Skip to main content

Full text of "Vascular-Surgery-Books_1aim.net"

See other formats


FM.qxd 4/21/04 3:37 PM Page iv 



ELSEVIER 
SAUNDERS 

The Curtis Center 
Independence Square West 
Philadelphia, Pennsylvania 19106 



VASCULAR TRAUMA, SECOND EDITION 

Copyright © 2004, Elsevier Science (USA). All rights reserved. 



ISBN: 0-7216-4071-0 



No part of this publication may be reproduced or transmitted in any form or by any means, electronic 
or mechanical, including photocopying, recording, or any information storage and retrieval system, 
without permission in writing from the publisher. Permissions may be sought directly from 
Elsevier's Health Sciences Rights Department in Philadelphia, PA, USA: phone: (+1) 215 238 7869, 
fax: (+1) 215 238 2239, e-mail: healthpermissions@elsevier.com. You may also complete your request 
on-line via the Elsevier Science homepage (http://www.elsevier.com), by selecting 'Customer Support' 
and then 'Obtaining Permissions'. 



NOTICE 



Surgery is an ever-changing field. Standard safety precautions must be followed, but as new research 
and clinical experience broaden our knowledge, changes in treatment and drug therapy may 
become necessary or appropriate. Readers are advised to check the most current product 
information provided by the manufacturer of each drug to be administered to verify the 
recommended dose, the method and duration of administration, and contraindications. It is the 
responsibility of the licensed prescriber, relying on experience and knowledge of the patient, to 
determine dosages and the best treatment for each individual patient. Neither the publisher nor 
the author assumes any liability for any injury and/or damage to persons or property arising from 
this publication. 

The Publisher 



Previous edition copyrighted 1978 

International Standard Book Number: 0-7216-4071-0 



Printed in the United States of America 

Last digit is the print number: 987654321 



FM.qxd 4/21/04 3:37PM Page ■ 



To those who serve and have served our 
country 

— in the military, both at home and in 

distant lands 

— in our nation's trauma centers 

— in the education of surgeons with an 

interest in vascular disease 

— in the development of new knowledge 

— in safety net hospitals 



FM.qxd 4/21/04 3:37 PM Page vii 




CONTRIBUTORS 



JOHN T. ANDERSON, MD 

Assistant Professor 

Department of Surgery 

Trauma Surgery and Surgical Critical Care 

University of California, Davis 

Sacramento, California 

JUAN A. ASENSIO, MD 

Associate Professor 

Department of Surgery 

University of Southern California Keck School of Medicine 

Los Angeles, California 

WALTER L. BIFFL, MD 

Associate Professor 

Department of Surgery 

Brown Medical School 

Chief, Division of Trauma and Surgical Critical Care 

Rhode Island Hospital 

Providence, Rhode Island 

F. WILLIAM BLAISDELL, MD 

Professor 

Department of Surgery 
University of California, Davis 
Sacramento, California 

KEVIN M. BRADLEY, MD 

Assistant Professor 

Department of Surgery 

Temple University School of Medicine 

Philadelphia, Pennsylvania 

ROBERT F. BUCKMAN, MD 

Professor 

Department of Surgery 

Drexel University College of Medicine 

Trauma Program Director 

Saint Mary Medical Center 

Langhorne, Pennsylvania 



VI 1 



FM.qxd 4/21/04 3:37 PM Pageviii 



Vlll CONTRIBUTORS 



JON M. BURCH, MD 

Professor 

Department of Surgery 

University of Colorado Health Sciences Center 

Denver, Colorado 

NEAL S. CAYNE, MD 

Assistant Professor 

Department of Surgery 

New York University School of Medicine 

Director of Endovascular Surgery 

New York University Medical Center 

New York, New York 

IRSHAD H. CHAUDRY, PhD 

Professor, Departments of Surgery, Microbiology, Physiology, and Biophysics 
Vice Chairmen, Department of Surgery 
Director, Center for Surgical Research 
The University of Alabama at Birmingham 
Birmingham, Alabama 

RAUL CIOMBRA, MD 

Associate Professor 

Department of Surgery 

Division of Trauma, Surgical Critical Care and Burns 

University of California, San Diego School of Medicine 

San Diego, California 

LORI D. CONKLIN, MD 

Surgical Resident 

Michael E. DeBakey Department of Surgery 

Baylor College of Medicine 

Houston, Texas 

MICHAEL E. DeBAKEY, MD 

Chancellor Emeritus 

Distinguished Service Professor 

Michael E. DeBakey Department of Surgery 

Baylor College of Medicine 

Houston, Texas 

DEMETRIOS DEMETRIADES, MD, PhD 

Professor 

Department of Surgery 

Division of Trauma and Critical Care 

Keck School of Medicine University of Southern California 

Los Angeles, California 

JAMES W. DENNIS, MD 

Professor 

Department of Surgery 

University of Florida Health Science Center 

Chief, Division of Vascular Surgery 

Shands Jacksonville Medical Center 

Jacksonville, Florida 



FM.qxd 4/21/04 3:37 PM Page ix 



CONTRIBUTORS IX 



DAVID V. FELICIANO, MD 

Professor 

Department of Surgery 

Emory University School of Medicine 

Chief of Surgery 

Grady Memorial Hospital 

Atlanta, Georgia 

ERIC R. FRYKBERG, MD 

Professor 

Department of Surgery 

University of Florida College of Medicine 

Chief, Division of General Surgery 

Shands Jacksonville Medical Center 

Jacksonville, Florida 

PRISCILLA J. GARCIA, MD 

Resident, Department of Anesthesia 
Baylor College of Medicine 
Houston, Texas 

NICHOLAS J. GARGIULO, III, MD 

Assistant Professor 

Department of Surgery 

The Albert Einstein College of Medicine 

Chief of Endovascular Surgery 

Jack D. Weiler Hospital 

Bronx, New York 

THOMAS S. GRANCHI, MD, MBA 

Associate Professor 

Michael E. DeBakey Department of Surgery 

Baylor College of Medicine 

Medical Director, Emergency Center 

Ben Taub General Hospital 

Houston, Texas 

ASHER HIRSHBERG, MD 

Associate Professor 

Michael E. DeBakey Department of Surgery 

Baylor College of Medicine 

Director of Vascular Surgery 

Ben Taub General Hospital 

Houston, Texas 

DAVID B. HOYT, MD 

Professor and Interim Chairman 

Department of Surgery 

University of California, San Diego 

Chief, Division of Trauma, Burns, and SICU 

UCSD Medical Center 

San Diego, California 



FM.qxd 4/21/04 3:37PM Page x 



CONTRIBUTORS 



RAO R. IVATURY, MD 

Professor 

Department of Surgery 

Virginia Commonwealth University 

Chief, Division of Trauma/Critical Care 

VCU Medical Center 

Richmond, Virginia 

DORAID JARRAR, MD 

Chief Resident 

Department of General Surgery 

The University of Alabama at Birmingham 

Birmingham, Alabama 

KAJ JOHANSEN, MD, PhD 

Clinical Professor 

Department of Surgery 

University of Washington School of Medicine 

Director, Peripheral Vascular Services 

Swedisky Medical Center 

Seattle, Washington 

M. MARGARET KNUDSON, MD 

Professor 

Department of Surgery 
University of California 
Director, Injury Research Center 
San Francisco General Hospital 
San Francisco, California 

ANNA M. LEDGERWOOD, MD 

Professor 

Department of Surgery 
Wayne State University 
Detroit, Michigan 

SCOTT A. LeMAIRE, MD 

Assistant Professor 

Division of Cardiothoracic Surgery 

Michael E. DeBakey Department of Surgery 

Baylor College of Medicine 

Houston, Texas 

MICHAEL R. LePORE, MD 

Medical Director of Peripheral Vascular Surgery 
Sarasota Memorial Hospital 
Sarasota, Florida 

CHARLES E. LUCAS, MD 

Professor 

Department of Surgery 
Wayne State University 
Detroit, Michigan 



FM.qxd 4/21/04 3:37PM Page xi 



CONTRIBUTORS XI 



KENNETH L. MATTOX, MD 

Professor and Vice Chairman 

Michael E. DeBakey Department of Surgery 

Baylor College of Medicine 

Chief of Staff and Chief of Surgery 

Ben Taub General Hospital 

Houston, Texas 

SAMUEL R. MONEY, MD 

Clinical Associate Professor 

Department of Surgery 

Tulane University 

Head, Section of Vascular Surgery 

Ochsner Clinic Foundation 

New Orleans, Louisiana 

ERNEST E. MOORE, MD 

Vice Chairman and Professor 

Department of Surgery 

University of Colorado Health Sciences Center 

Chief of Surgery and Trauma 

Denver Health 

Denver, Colorado 

JAMES A. MURRAY, MD 

Division of Trauma and Critical Care 

Department of Surgery 

Keck School of Medicine University of Southern California 

Assistant Professor of Surgery 

University of Southern California 

Los Angeles, CA 

TAKAO OHKI, MD, PhD 

Associate Professor 

Department of Surgery 

Albert Einstein College of Medicine 

Chief, Vascular and Endovascular Surgery 

Montefiore Medical Center 

Bronx, New York 

ABHIJIT S. PATHAK, MD 

Assistant Professor 

Department of Surgery 

Temple University School of Medicine 

Director, Surgical Intensive Care Unit 

Temple University Hospital 

Philadelphia, Pennsylvania 

DAVID C. RICE, MD, MB, BCH 

Assistant Professor 

Department of Thoracic and Cardiovascular Surgery 
University of Texas M.D. Anderson Cancer Center 
Houston, Texas 



FM.qxd 4/21/04 3:37 PM Page xii 



Xll CONTRIBUTORS 



NORMAN M. RICH, MD 

Leonard Heaton and David Packard Professor 

Chairman, Department of Surgery, USUHS 

Chief, Division of Vascular Surgery, Emeritus 

F. Edward Herbert School of Medicine 

Uniformed Services University of the Health Sciences 

Bethesda, Maryland 

AURELIO RODRIGUEZ, MD 

Professor 

Department of Surgery 

Drexel University College of Medicine 

Director, Division of Trauma Surgery 

Allegheny General Hospital Shock Trauma Center 

Pittsburgh, Pennsylvania 

SALVATORE J.A. SCLAFANI, MD 

Professor 

Department of Radiology 
State University of New York 
Director, Department of Radiology 
Kings County Hospital Center 
Brooklyn, New York 

BRADFORD G. SCOTT, MD 

Assistant Professor 

Michael E. DeBakey Department of Surgery 

Baylor College of Medicine 

Associate Trauma Medical Director 

Ben Taub General Hospital 

Houston, Texas 

STEVEN R. SHACKFORD, MD 

Stanley S. Fieber Professor and Chairman 

Department of Surgery 

University of Vermont College of Medicine 

Surgeon-in-Chief 

Fletcher Allen Health Care 

Burlington, Vermont 

MICHAEL J. SISE, MD 

Clinical Professor 
Department of Surgery 
UCSD School of Medicine 
Medical Director 
Scripps Mercy Hospital 
San Diego, California 

ERNESTO SOLTERO, MD 

Assistant Professor 

Michael E. DeBakey Department of Surgery 

Baylor College of Medicine 

Chief of Cardiovascular Surgery 

Michael E. DeBakey VA Medical Center 

Houston, Texas 



FM.qxd 4/21/04 3:37PM Pagexiii 



CONTRIBUTORS Xlll 



MICHAEL C. STONER, MD 

Fellow 

Division Vascular and Endovascular Surgery 

Massachusetts General Hospital 

Boston, Massachusetts 

FRANK J. VEITH, MD 

Professor and Vice Chairman 

Department of Surgery 

Albert Einstein College of Medicine 

The William J. von Liebig Chair in Vascular Surgery 

Montefiore Medical Center 

Albert Einstein College of Medicine 

Bronx, New York 

MATTHEW J. WALL, Jr, MD 

Professor 

Michael E. DeBakey Department of Surgery 

Baylor College of Medice 

Deputy Chief of Surgery 

Chief of Cardiothoracic Surgery 

Ben Taub General Hospital 

Houston, Texas 

PING WANG, MD 

Professor and Chief 

Division of Surgical Research 

Department of Surgery 

North Shore-Long Island Jewish Medical Center 

Manhasset, New York 



FM.qxd 4/21/04 3:37PM Page xv 



FOREWORD 



Nearly 25 years have passed since the first edition of Vascular Trauma 
was published, edited by Norman Rich and myself. The first edition 
included experiences from both the Korean and Vietnam Con- 
flicts. The Korean experiences demonstrated for the first time that arte- 
rial repair was imminently feasible in battle casualties without a serious 
hazard of infection. As cited by Hughes, a total of 304 arterial injuries 
underwent 269 repairs, with a 13% amputation rate as compared to the 
dismal 50% amputation rate following ligation in World War II. Our results 
in the U.S. Marine Corps were separately reported in the Annals of Surgery 
in 1955. 

The Korean experience quickly led to widespread adoption of 
arterial repair, following which over 7500 such injuries were repaired 
in the Vietnam Conflict. These were entered into the Walter Reed 
Vascular Registry organized by Norman Rich; 1500 late results were then 
evaluated. 

Arterial repair became possible primarily from the development of 
helicopter evacuation of wounded men. This coincided with the evolu- 
tion of techniques of vascular repair, improved resuscitation, and anti- 
biotics. Now, it is well established that the majority of arterial injuries can 
be effectively repaired if blood flow is restored within 6 to 7 hours after 
injury. After 7 to 8 hours, however, there is a rapid rise in the frequency of 
irreversible muscle necrosis, depending primarily upon extent of the 
associated soft tissue destruction with loss of collateral circulation. 

Initially, it was feared that arterial repair would result in a prohibi- 
tive degree of wound infection, especially in Korea where the widespread 
use of cow manure for fertilizer resulted in gross contamination of virtu- 
ally all injuries. Nonetheless, with adequate debridement, antibiotics and 
secondary closure, infection was rarely seen. 

Several valuable developments have occurred since 1978 that make 
vascular repair even more feasible than before. These include the use 
of soft tissue pedicle flaps to cover arterial repair after radical debride- 
ment, the early detection of the vascular compartment syndrome by 
tissue pressure monitoring, and the recent development of endovascular 
techniques. 

This book is of special importance because vascular injuries are uncom- 
mon in civilian trauma though increasing in frequency, primarily from 

XV 



FM.qxd 4/21/04 3:37PM Page xvi 



XVI FOREWORD 



automobile accidents and gunshot wounds. Civilian wounds fortunately 
often don't have the severe concomitant soft tissue destruction that often 
occurs with injury from high velocity missiles. Hence, extensive debride- 
ment and secondary closure are less often needed, but form the basis of 
the time-honored fact that the hazard of infection should virtually never 
prohibit arterial repair. 

The rarity of arterial injuries makes continuing efforts with educa- 
tion, training, and referral to specialized vascular centers most important. 
Extremities are still lost because the gravity of early crucial symptoms of 
limb-threatening ischemia was missed. It has been known for over four 
decades that limb-threatening ischemia produces loss of peripheral nerve 
function within a few minutes after onset, manifested by numbness and 
paralysis in the affected extremity; but this crucial basic physiological fact 
is simply unknown to a surprisingly large number of personnel treating 
injured patients. 

Another fact emphasizing the importance of experience is the fact 
that an acute vascular injury can be repaired with a success rate probably 
greater than 95% if treated with modern techniques within 6 to 7 hours 
after injury. If repair initially fails, however, prompt reoperation similarly 
has a success rate over 90%, for the cause of failure is usually thrombosis 
of an inadequate repair or thrombi incompletely removed at the first 
operation. Both of these preventable complications usually result from 
simple lack of experience. 

These facts clearly show the importance of this book for all physi- 
cians and staff treating injured patients. Strong adherence to the basic 
principles described makes arterial repair achievable in the vast majority 
of patients. The authors are to be congratulated on their significant 
contributions. 

Frank Spencer, MD 



FM.qxd 4/21/04 3:37 PM Page xvii 




FOREWORD 



Among traumatic injuries, those affecting the major vessels are of 
great importance, since they represent a serious threat to life and 
limb. These injuries assume special significance in light of the 
increasing number of trauma patients arriving at the hospital as a result 
of vehicular accidents, violent crimes, and other hazardous events. 

Until relatively recently, therapy for vascular injuries was limited to 
lifesaving control of hemorrhage. Even during World War II, surgical 
repair of arterial injuries was rarely attempted. The pioneering deve- 
lopments in vascular surgery that evolved in the early 1950s and the 
successful surgical repair of vascular injuries in the Korean War provided 
the basis for effective surgical treatment of this form of trauma. Accord- 
ingly, in the care of patients with vascular trauma only, salvage of life 
is no longer acceptable; the goal is also rapid restoration of normal 
circulatory dynamics. 

The authors, Norman Rich, Kenneth Mattox, and Asher Hirshberg, 
have had extensive and wide-ranging experience in the development of 
the most effective methods of treatment of vascular injuries, including 
civilian and military experience, especially during the Korean and Vietnam 
Wars. The authors have incorporated in this book a consideration of the 
entire subject of vascular injuries, including an interesting historical review 
of the topic; the relative frequency, and sites of their occurrence; and the 
clinical, anatomical, and surgical technical aspects of this subject. It 
will therefore be of immense value and usefulness to both civilian and 
military surgeons. 

Micheal E. DeBakey, M.D. 

Distinguished Service Professor 

and Olga Keith Wiess Professor of Surgery 

Michael E. DeBakey Department of Surgery 

Director, DeBakey Heart Center 

Chancellor Emeritus, Baylor College of Medicine 

Houston, Texas 



xvi 1 



FM.qxd 4/21/04 3:37PM Page xix 



PREFACE 



The first edition of this text was published in 1978 as a monograph 
written by Drs. Norman Rich and Frank Spencer. It was an exten- 
sive treatise on the pathophysiology, diagnosis, and management 
of traumatic injuries to blood vessels. It was and still is one of a kind. No 
other textbook on vascular trauma has been published before or since. 
The first edition was written in the aftermath of the Vietnam War, the 
first military conflict where modern principles of vascular surgery were 
applied to traumatic injuries of the blood vessels. Many of the lessons and 
concepts delineated in the first edition were based on the Vietnam Vas- 
cular Registry, an unprecedented effort, led by Dr. Rich, to systematically 
collect and analyze the vascular injuries in a large-scale military conflict. 
Yet with very few exceptions, most surgeons who performed vascular recon- 
structions in Vietnam did so only on a handful of patients. 

In the 1980s, as surgeons began to encounter increasing numbers of 
major injuries to blood vessels in the civilian population, there was a surge 
of interest in vascular trauma and an exponential rise in the number of 
publications on the subject. Many of the advances in the field originated 
at urban trauma centers, and particularly at the Ben Taub General Hos- 
pital in Houston, where the modern concepts of cardiovascular surgery, 
pioneered by Drs. Michael E. DeBakey, Stanley Crawford, Arthur Beall 
and others at Baylor College of Medicine, were developed into new man- 
agement strategies in vascular trauma by Dr. Kenneth L. Mattox and his 
team described in more than a hundred publications. 

Ten years ago, Dr. Rich enlisted the assistance of Dr. Mattox in writing 
the second edition of Vascular Trauma. The objective was to combine the 
military and civilian experience into one cohesive text. However, the task 
rapidly proved to be a "mission impossible" as the rapid developments in 
the fields of trauma systems, care of the injured patient, damage control 
surgery, vascular imaging and endovascular intervention constantly 
outpaced the revision of the original text. This led to the addition 
of Dr. Asher Hirshberg to the editorial team. He represents a new 
generation of surgeons formally trained in both trauma and vascular 
surgery. The plan for the book has subsequently changed from a mono- 
graph to a multi-authored text containing cutting-edge concepts and 
information in vascular trauma. At the same time, we wished to keep 
some of the original exciting "flavor" from the first edition of Vascular 
Trauma, so expertly and lovingly compiled by Drs. Rich and Spencer. 

xix 



FM.qxd 4/21/04 3:37 PM Page xx 



XX PREFACE 



Drs. Rich and Spencer assembled all their references at the end of 
the first edition, assuming that many of the references will be used in 
more than one chapter. Dr. Rich continued to collect key references during 
the next two decades, and his entire treasury of vascular trauma refer- 
ences, a unique and extremely valuable resource for any surgeon inter- 
ested in the care of patients with injuries to blood vessels, is given at the 
end of this book. We have attempted to assure that all citations in each 
chapter are in this reference list, but because of the enormity of this project, 
it is inevitable that some might have been omitted. It is also inevitable that 
some vascular trauma references might have been missed by all three 
editors. 

The editors are indebted to the numerous authors of this text, each 
one with a life-long commitment to the care of the injured. Ms. Mary Allen 
was the persistent force that brought the many aspects of this book together. 
In addition, we are grateful to the numerous individuals at Elsevier who 
contributed to the culmination of this endeavor. 

Norman M. Rich, MD 
Kenneth L. Mattox, MD 
Asher Hirshberg, MD 



chOl.qxd 4/16/04 3:20PM Page 3 




Historical and Military 
Aspects of Vascular Trauma 
(With Lifetime Reflections of 
Doctor Norman Rich) 



NORMAN M. RICH 



O 

o 



HISTORICAL OVERVIEW ON VASCULAR TRAUMA 

Control of Hemorrhage from the Time of Antiquity 

EARLY DIRECT VASCULAR RECONSTRUCTION 

MILITARY VASCULAR TRAUMA EXPERIENCE 

Balkin Wars 

World War I Experience 

World War II Experience 

Experiences during the Korean Conflict 

Experience in Vietnam 

Military Armed Conflicts following Vietnam 

CIVILIAN VASCULAR INJURIES 

HISTORICAL NOTES ON 20TH CENTURY PROGRESS WITH VENOUS 
INJURIES 

SPECIAL HISTORIC OBSERVATIONS 

Site of Injury 

Iatrogenic Injury 

Historic Observations on Mechanism of Injury 

Fractures 



O 

o 



chOl.qxd 4/16/04 3:20PM Page 4 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



o 



Posterior dislocation of the knee 

Blunt injury in presence of other vascular pathology 

Use of crutches 

Athletic injuries 

Vascular injury in children 

Radiation 

Vibratory tools 
Historic Classification of Vascular Injury 
HISTORY OF BALLISTICS AND VASCULAR INJURY 
HISTORICAL REFLECTIONS AND PROJECTIONS 



The advances in vascular surgery are typical 
of those in other fields of medicine and 
surgery. Each step is discovered and 
recorded only to be rediscovered by other 
individuals who failed to read and profit by 
the experience of others. 
Carl W. Hughes (1961) 



HISTORICAL OVERVIEW ON 
VASCULAR TRAUMA 

Although the first crude arteriorrhaphy was 
performed about 243 years ago, only in the 
past 40 years has vascular surgery become 
widely practiced with the anticipation of con- 
sistently obtaining good results. By the turn 
of this century, extensive experimental work 
and some early clinical applications had 
occurred, employing most of the techniques 
of vascular surgery in use today. In retrospect, 
it is almost astonishing that it took nearly 50 
years before the work of early pioneers such 
as Murphy, Goyanes, Carrel, Guthrie, and 
Lexer was widely accepted and applied in the 
treatment of vascular injuries. Since the days 
of Ambroise Pare in the mid-1 6th century, 
major advances in the surgery of trauma have 
occurred during the times of armed conflict, 
when it was necessary to treat large numbers 
of severely injured patients often under con- 
ditions far from ideal. This has been especially 
true with vascular injuries. 



Although German surgeons accomplished 
a limited number of arterial repairs in the early 
part of World War I, it was not until the Korean 
Conflict in the early 1950s that ligation of 
major arteries was abandoned as the standard 
treatment for arterial trauma. The results of 
ligation of major arteries following trauma 
were clearly recorded in the classic manuscript 
by DeBakey and Simeone (1946) , who found 
only 81 repairs in 2471 arterial injuries among 
U.S. troops in World War II. All but three of 
the arterial repairs were performed by lateral 
suture. Ligation was followed by gangrene and 
amputation in nearly one half of the cases. 
The pessimistic conclusion reached by many 
was expressed by Sir James Learmonth (1946) , 
who said that there was little place for defin- 
itive arterial repair in the combat wound. 

Between the end of World War II and the 
beginning of the Korean Conflict, advances 
in suture, noncrushing clamps, and arteriog- 
raphy were emerging. During the Korean 
Conflict continuing technology in polymer- 
ized material (plastic) added a new opportu- 
nity for vascular reconstruction. 

The possibility of successfully repairing arte- 
rial injuries was established conclusively, stem- 
ming particularly from the works of Hughes, 
Howard, Jahnke, and Spencer. In 1958, 
Hughes emphasized the significance of this 
contribution in a review of the Korean expe- 
rience, finding that the overall amputation 
rate was lowered to about 13%, compared to 
the approximately 49% amputation rate that 
followed ligation in World War II. 



chOl.qxd 4/16/04 3:20PM Page 5 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



During the Vietnam hostilities, more than 
600 young U.S. surgeons, representing most 
of the major surgical training programs in the 
United States, treated more than 7500 patients 
with vascular injuries. Rich and Hughes (1969) 
reported the preliminary statistics from the 
Vietnam Vascular Registry, established in 
1966 at Walter Reed General Hospital to 
document and follow all servicemen who sus- 
tained vascular trauma in Vietnam. The 
interim Registry report, encompassing 1000 
major acute arterial injuries, showed little 
change from the overall statistics presented 
in the preliminary report (Rich, 1970). Con- 
sidering all major extremity arteries, the 
amputation rate remained near 13%. 
Although high-velocity missiles created more 
soft tissue destruction in injuries seen in 
Vietnam, the combination of a stable hospi- 
tal environment and rapid evacuation of casu- 
alties, similar to that in Korea, made successful 
repair possible. Injuries of the popliteal artery 
remained an enigma, with an amputation rate 
remaining near 30%. 

In the past 40 years, civilian experience with 
vascular trauma has developed rapidly under 
conditions much more favorable than those 
of warfare. As might be predicted, several 
series have reported results that are signifi- 
cantly better than those achieved with mili- 
tary casualties in Korea and Vietnam. Mattox 
(1989) published the epidemiology of the 
largest civilian experience in managing vas- 
cular trauma in the history of the world. 

Control of Hemorrhage from 
the Time of Antiquity 

The control of hemorrhage following injury 
has been of prime concern to humans since 
the beginning. Methods have included various 
animal and vegetable tissues, hot irons, boiling 
pitch, cold instruments, styptics, bandaging, 
and compression. These methods were 
described in a historical review by Schwartz 
in 1958. Ancient methods of hemostasis used 
by Egyptians about 1600 bc are described in 
the Ebers' papyrus, discovered by Ebers at 
Luxor in 1873 (Schwartz, 1958) . Styptics pre- 
pared from mineral or vegetable matter were 
popular, including lead sulfate, antimony, and 



copper sulfate. Several hundred years later, 
copper sulfate again became popular during 
the Middle Ages in Europe and was known as 
the hemostatic "button." In ancient India, 
compression, cold elevation, and hot oil were 
used to control hemorrhage, while the 
Chinese about 1000 bc used tight bandaging 
and styptics. 

The writings of Celsus provide most of the 
knowledge of methods of hemostasis in the 
1st and 2nd centuries ad, Celsus was the first 
to record an accurate account of the use of 
ligature for hemostasis in 25 ad. During the 
first three centuries ad, Galen, Heliodorus, 
Rufus of Ephesus, and Archigenes advocated 
ligation or compression of a bleeding vessel 
to control hemorrhage. The prevailing sur- 
gical practice when amputation was done for 
gangrene was to amputate at the line of 
demarcation to prevent hemorrhage. Archi- 
genes, in the 1st century ad was apparently 
the first to advocate amputating above the line 
of demarcation for tumors and gangrene, 
using ligature of the artery to control 
hemorrhage. 

Rufus of Ephesus (1st century ad) noted 
that an artery would continue to bleed when 
partly severed, but when completely severed, 
it would contract and stop bleeding within a 
short time. Galen, the leading physician of 
Rome in the 2nd century ad, advised placing 
a finger on the orifice of a bleeding superfi- 
cial vessel for a period to initiate the forma- 
tion of a thrombus and the cessation of 
bleeding. He noted, however, that if the vessel 
were deeper, it was important to determine 
whether the bleeding was coming from an 
artery or a vein. If a vein, pressure or a styptic 
usually sufficed, but ligation with linen was 
recommended for arterial injury. Herophilus, 
the Greek physician and anatomist of the 3rd 
century bc described the difference between 
veins and arterial as "veins were weak and thin- 
walled, containing only blood, whereas arter- 
ies were thick-walled, containing air 'pneuma' 
and blood." 

Following the initial contributions of Celsus, 
Galen, and their contemporaries, the use of 
ligature was essentially forgotten for almost 
1200 years. Throughout the Middle Ages, 
cautery was used almost exclusively to 
control hemorrhage. Jerome of Brunswick 



chOl.qxd 4/16/04 3:20PM Page 6 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



(Hieronymus Brunschwig) , an Alsatian Army 
surgeon, actually preceded Pare in describ- 
ing the use of ligatures as the best way to stop 
hemorrhage (Schwartz, 1958). His recom- 
mendations were recorded in a textbook pub- 
lished in 1497 and provided a detailed account 
of the treatment of gunshot wounds. Ambroise 
Pare, with a wide experience in the surgery 
of trauma, especially on the battlefield, estab- 
lished firmly the use of ligature for control of 
hemorrhage from open blood vessels. In 1 552 , 
he startled the surgical world by amputating 
a leg above the line of demarcation, repeat- 
ing the demonstration of Archigenes 1400 
years earlier. The vessels were ligated with 
linen, leaving the ends long. Pare also devel- 
oped the "bee de corbin," ancestor of the 
modern hemostat, to grasp the vessel before 
ligating it (Fig. 1-1). Previously, vessels had 
been grasped with hooks, tenaculums, or the 
assistant's fingers. 

In the 17th century, Harvey's monumental 
contribution describing the circulation of the 
blood greatly aided the understanding of 
vascular injuries. Although Rufus of Ephesus 
apparently discussed arteriovenous commu- 
nications in the 1 st century ad, it was not until 
1757 that William Hunter first described the 
arteriovenous fistula as a pathologic entity. The 
historical development of the treatment of 
arteriovenous fistulas and false aneurysms are 
discussed in Chapter 24. Also, similar review 
of false aneurysms is included. As early as the 
2nd century ad, Antyllus described the phys- 
ical findings and management by proximal 




■ FIGURE 1-1 

Artist's concept of the bee de corbin, 
developed by Pare and Scultetus in the mid- 
16th century. It was used to grasp the vessel 
before ligating it. (From Schwartz AM: Surgery 
1958;44:604.) ■ 



and distal ligation. He was the first to docu- 
ment collateral circulation. 

The development of the tourniquet was 
another advance that played an important role 
in the control of hemorrhage. Tight bandages 
had been applied since antiquity, but subse- 
quent development of the tourniquet was slow. 
Finally, in 1674, a military surgeon named 
Morel introduced a stick into the bandage and 
twisted it until arterial flow stopped (Schwartz, 
1958). The screw tourniquet came into use 
shortly thereafter. This method of temporary 
control of hemorrhage encouraged more fre- 
quent use of the ligature, which required time 
for its application. In 1873, Freidrich van 
Esmarch, a student of Langenbeck, intro- 
duced his elastic tourniquet bandage for first 
aid use on the battlefield. Previously, it was 
thought that such compression would injure 
vessels irreversibly. His discovery permitted 
surgeons to operate electively on extremities 
in a dry, bloodless field. 

In addition to the control of hemorrhage 
at the time of injury, the second major area 
of concern for centuries was the prevention 
of secondary hemorrhage occurring days to 
weeks later. Because of its great frequency, styp- 
tics, compression, and pressure were used for 
several centuries after ligation of injured 
vessels became possible. Undoubtedly, the 
high rate of secondary hemorrhage after 
ligation was due to infection of the wound. 
Although John Hunter demonstrated the 
value of proximal ligation for control of a false 
aneurysm in 1 757, failure to control secondary 
hemorrhage resulted in the use of ligature only 
for secondary bleeding from the amputation 
stump. Subsequently, Bell (1801) and Guthrie 
(1815) performed ligation both proximal and 
distal to the arterial wound with better results 
than those previously obtained. 

Some of the first clear records of ligation 
of major arterial were written in the 19th 
century and are of particular interest. The first 
successful ligation of the common carotid 
artery for hemorrhage was performed in 1 803 
by Fleming but was not reported until 14 years 
later by Coley (1817), because Fleming died 
a short time after the operation was per- 
formed. A servant aboard the HMS Tonnant 
attempted suicide by slashing his throat. 



chOl.qxd 4/16/04 3:20PM Page 7 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



When Fleming saw the patient, it appeared 
that he had exsanguinated. There was no pulse 
at the wrist and the pupils were dilated. It was 
possible to ligate two superior thyroid arter- 
ies and one internaljugular vein. A laceration 
of the outer and muscular layers of the carotid 
artery was noted, as well as a laceration of the 
trachea between the thyroid and cricoid car- 
tilages. This allowed drainage from the wound 
to enter the trachea, provoking violent seizures 
of coughing. Although the patient seemed to 
be improving, approximately 1 week follow- 
ing the injury, Flemming recorded that "on 
the evening of the 1 7th, during a violent parox- 
ysm of coughing, the artery burst, and my 
poor patient was, in an instant, deluged with 
blood!" 

The dilemma of the surgeon is appreciated 
by the statement, "In this dreadful situation I 
concluded that there was but one step to take, 
with any prospect of success; mainly, to cut- 
down upon, and tie the carotid artery below 
the wound. I had never heard of such an 
operation being performed; but conceived 
that its effects might be less formidable, in 
this case, than in a person not reduced by 
hemorrhage." 

The wound rapidly healed following liga- 
tion of the carotid artery and the patient 
recovered. 

Ellis (1845) reported the astonishing expe- 
rience of successful ligation of both carotid 
arteries in a 21-year-old patientwho sustained 
a gunshot wound of the neck while he was 
setting a trap in the woods on October 21, 
1844, near Grand Rapids, Michigan, when he 
was unfortunately mistaken for a bear by a 
companion. Approximately 1 week later, Ellis 
had to ligate the patient's left carotid artery 
because of a hemorrhage. An appreciation 
of the surgeon's problem can be gained by 
Ellis' description of the operation, "We placed 
him on a table, and with the assistance of 
Doctor Piatt and a student, I ligatured the 
left carotid artery, below the omohyoideus 
muscle; an operation attended with a good 
deal of difficulty, owing to the swollen state 
of the parts, the necessity of keeping up pres- 
sure, the bad position of the parts owing to 
the necessity of keeping the mouth in a certain 
position to prevent his being strangulated by 



the blood, and the necessity of operating by 
candlelight." 

There was recurrent hemorrhage on the 
11th day after the accident and right carotid 
artery pressure helped control the blood loss. 
It was, therefore, necessary to ligate also the 
right carotid artery 4/ 2 days after the left 
carotid artery had been ligated. Ellis (1845) 
remarked, "For convenience, we had him in 
the sitting posture during the operation; 
when we tightened the ligature, no disagree- 
able effects followed; no fainting; no bad 
feeling about the head; and all the percepti- 
ble change was a slight paleness, a cessation 
of pulsation in both temporal arteries, and of 
the hemorrhage." 

The patient recovered rapidly with good 
wound healing and returned to normal daily 
activity. There was no perceptible pulsation 
in either superficial temporal artery. 

The importance of collateral circulation in 
preserving viability of the limb after ligation 
was well understood for centuries. The fact 
that time was necessary for establishment of 
this collateral circulation was recognized. 
Halsted (1912) reported cure of an iliofemoral 
aneurysm by application of an aluminum band 
to the proximal artery without seriously affect- 
ing the circulation or function of the lower 
extremity. The importance of asepsis had now 
been recognized, and the frequency of sec- 
ondary hemorrhage and gangrene following 
ligation promptly decreased. Subsequently, 
Halsted (1914) demonstrated the roll of col- 
lateral circulation by gradually completely 
occluding the aorta and other large arteries 
in dogs by means of silver or aluminum bands, 
which were gradually tightened over a period 
of time. 



EARLY DIRECT VASCULAR 
RECONSTRUCTION 



About two centuries after Pare established the 
use of the ligature, the first direct repair of 
an injured artery was accomplished. This 
event, about 243 years ago, is credited as the 
first documented vascular repair. Hallowell 
(1762) , acting on a suggestion by Lambert in 



chOl.qxd 4/16/04 3:20PM Page 8 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 




Figure-of-eight suture 
Pin 



Laceration 



Brachial Artery 



■ FIGURE 1-2 

The first arterial repair performed by Hallowell, acting on a suggestion by Lambert in 1759. The 
technique, known as the farrier's (veterinarian's) stitch, was followed in repairing the brachial artery 
by placing a pin through the arterial walls and holding the edges in apposition with a suture in a 
figure-of-eight fashion about the pin. (From Lambert. Med Obser Inq 1762;30:360.) ■ 



1759, repaired a wound of the brachial artery 
by placing a pin through the arterial walls and 
holding the edges in apposition by applying 
a suture in a figure-of-eight fashion about the 
pin (Fig. 1-2). This technique (known as the 
Farrier stitch) had been used by veterinari- 
ans but had fallen into disrepute following 
unsuccessful experiments. Table 1-1 outlines 
early vascular techniques. 

Unfortunately, others could not duplicate 
Hallowell's successful experience, almost 
surely because of the multiple problems of 
infection and lack of anesthesia. There was 
one report by Broca (1762) of a successful 
suture of a longitudinal incision in an artery. 
However, according to Shumacker (1969) , an 
additional 127 years passed following the 
Hallowell-Lambert arterial repair before a 
second instance of arterial repair by lateral 



TABLE 1-1 






VASCULAR REPAIR PRIOR TO 1900 


Technique Year 


Surgeon 


Pin and thread 


1759 


Hallowell 


Small ivory clamps 


881 


Gluck 


Fine needles and silk 


889 


Jassinowski 


Continuous suture 


890 


Burci 


Invagination 


896 


Murphy 


Suture all layers 


899 


Dorfler 



Adapted from Guthrie GC: Blood Vessel Surgery and Its 
Application. New York: Longmans, Green, 1912. 



suture of an artery in a man was reported by 
Postempski in 1886. 

With the combined developments of anes- 
thesia and asepsis, several reports of attempts 
to repair arteries appeared in the latter part 
of the 19th century. The work ofjassinowsky, 
who is credited in 1889 for experimentally 
proving that arterial wounds could be sutured 
with preservation of the lumen, was later 
judged by Murphy in 1897 as the best exper- 
imental work published at that time. In 1865, 
Henry Lee of London attempted repair of arte- 
rial lacerations with suture (Shumacker, 1969). 
Gluck in 1883 reported 19 experiments with 
arterial suture, but all experiments failed 
because of bleeding from the holes made by 
suture needles. He also devised aluminum and 
ivory clamps to unite longitudinal incisions 
in a vessel, and it was recorded that the ivory 
clamps succeeded in one experiment on the 
femoral artery of a large dog. Von Horoch of 
Vienna reported six experiments, including 
one end-to-end union, in 1887, all of which 
thrombosed. In 1889, Bruci sutured six lon- 
gitudinal arteriotomies in dogs; the procedure 
was successful in four. In 1890 Muscatello suc- 
cessfully sutured a partial transection of the 
abdominal aorta in a dog. In 1894 Heiden- 
hain closed by catgut suture, a 1-cm opening 
in the axillary artery made accidentally while 
removing the adherent carcinomatous glands. 
The patient recovered without any circulatory 
disturbance. In 1883, Israel, in a discussion of 
a paper by Gluck, described closing a lacera- 
tion in the common iliac artery created 



chOl.qxd 4/16/04 3:20PM Page 9 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



during an operation for peri typhli tic abscess. 
The closure was accomplished by five silk 
sutures. However, Murphy (1897) did not 
believe it could be possible from his personal 
observations to have success in this type of 
arterial repair. In 1896, Sabanyeff successfully 
closed small openings in the femoral artery 
with sutures. 

The classic studies ofjohn B. Murphy (1897) 
of Chicago contributed greatly to the devel- 
opment of arterial repair and culminated in 
the first successful end-to-end anastomosis of 
an artery in 1 896. Previously, Murphy had care- 
fully reviewed earlier clinical and experi- 
mental studies of arterial repair and had 
evaluated different techniques extensively in 
laboratory studies. Murphy attempted to 
determine experimentally how much artery 
could be removed and still allow an anasto- 
mosis. He found 1 inch of calf's carotid artery 
could be removed and the ends still approx- 
imated by invagination suture technique 
because of the elasticity of the artery. He con- 
cluded that arterial repair could be done with 
safety when no more than three fourths of an 
inch of an a artery had been removed, except 
in certain locations such as the popliteal fossa 
or the axillary space where the limb could be 
moved to relieve tension on the repair. He 
also concluded thatwhen more than one half 
of the artery was destroyed, it was better to 
perform and end-to-end anastomosis by 
invagination rather than to attempt repair of 
the laceration. This repair was done by intro- 
ducing sutures into the proximal artery, 
including only the two outer coats, and 
using three sutures to invaginate the proxi- 
mal artery into the distal one, reinforcing 
the closure with an interrupted suture 
(Fig. 1-3). 

In 1 896 Murphy was unable to find a similar 
recorded case involving the suture of an artery 
after complete division, and he consequently 
reported his experience (1897) in one patient 
and then and carried out a number of exper- 
iments to determine the feasibility of his pro- 
cedure. Murphy's patient was a 19-year-old 
male shot twice, with one bullet entering the 
femoral triangle. The patientwas admitted to 
Cook County Hospital in Chicago on Sep- 
tember 19, 1896, approximately 2 hours after 
wounding. There was no hemorrhage or 




**& 




■ FIGURE 1-3 

The first successful clinical end-to-end 
anastomosis of an artery was performed in 
1896. Sutures were placed in the proximal 
artery, including only the few outer costs, and 
three sutures were used to invaginate the 
proximal artery into the distal one; the closure 
was reinforced with an interrupted suture. 
(From Murphy JB: Exp Clin Res Med Rec 
1897;51:73-104.) ■ 



increased pulsation noted at the time. Murphy 
first saw the patient 15 days later, October 4, 
1896, and found a large bruit surrounding 
the site of the injury. Distal pulses were barely 
perceptible. Two days later, when demon- 
strating this patient to students, a thrill was 
also detected. An operative repair was decided. 
Because of the historical significance, the 
operation report is quoted: 



chOl.qxd 4/16/04 3:20PM Page 10 



10 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



Operation, October 7, 1896. An incision five 
inches long was made from Poupart's 
ligament along the course of the femoral 
artery. The artery was readily exposed about 
one inch above Poupart's ligament; it was 
separated from its sheath and a provisional 
ligature thrown about it but not tied. A careful 
dissection was then made down along the 
wall of the vessel to the pulsating clot. The 
artery was exposed to one inch below the 
point and a ligature thrown around it but not 
tied; a careful dissection was made upward to 
the point of the clot. The artery was then 
closed above and below with gentle 
compression clamps and was elevated, at 
which time there was a profuse hemorrhage 
from an opening in the vein. A cavity, about 
the size of a filbert, was found posterior to the 
artery communicating with its caliber, the 
aneurysmal pocket. A small aneurysmal sac 
about the same size was found on the 
anterior surface of the artery over the point of 
perforation. The hemorrhage from the vein 
was very profuse and was controlled by 
digital compression. It was found that one- 
eighth of an inch of the arterial wall on the 
outer side of the opening remained, and on 
the inner side of the perforation only a band 
of one-sixteenth of an inch of adventitia was 
intact. The bullet had passed through the 
center of the artery, carried away all of its wall 
except the strands described above, and 
passed downward and backward making a 
large hole in the vein in its posterior and 
external side just above the junction of the 
vena profunda. Great difficulty was 
experienced in controlling the hemorrhage 
from the vein. After dissecting the vein above 
and below the point of laceration and placing 
a temporary ligature on the vena profunda, 
the hemorrhage was controlled so that the 



vein was greatly diminished in size, but when 
the clamps were removed it dilated about 
one-third the normal diameter or one-third the 
diameter of the vein above and below. There 
was no bleeding from the vein when the 
clamps were removed. Our attention was then 
turned to the artery. Two inches of it had been 
exposed and freed from all surroundings. The 
opening in the artery was three-eighths of an 
inch in length; one-half inch was resected 
and the proximal was invaginated into the 
distal for one-third of an inch with four double 
needle threads which penetrated all of the 
walls of the artery. The adventitia was peeled 
off the invaginated portion for a distance of 
one-third of an inch: a row of sutures was 
placed around the edge of the overlapping 
distal end, the sutures penetrating only the 
medial of the proximal portion; the adventitia 
was then brought over the end of the union 
and sutured. The clamps were removed. Not 
a drop of blood escaped at the line of suture. 
Pulsation was immediately restored in the 
artery below the line of approximation and it 
could be felt feebly in the posterior tibial and 
dorsalis pedis pulses. The sheath and 
connective tissue around the artery were then 
approximated at the position of the suture 
with catgut, so as to support the wall of the 
artery. The whole cavity was washed out with 
a five percent solution of carbolic acid and 
the edges of the wound were accurately 
approximated with silk worm-gut sutures. No 
drainage. The time of the operation was 
approximately two and one-half hours, most 
of the time being consumed in suturing the 
vein. The artery was easily secured and 
sutured, and the hemorrhage from it readily 
controlled. The patient was placed in bed 
with the leg elevated and wrapped in 
cotton. 



The anatomic location of the injuries, the 
gross pathology involved and the repair for 
Murphy's historically successful arterial anas- 
tomosis are shown in Figure 1-4. Murphy men- 
tioned that a pulsation could be felt in the 
dorsalis pedis artery 4 days following the oper- 
ation. The patient had no edema and no 



disturbance of his circulation during the 
reported 3 months of observation. 

Subsequently, Murphy (1897) reviewed the 
results of ligature of large arteries before the 
turn of the century. He found that the abdom- 
inal aorta had been ligated 10 times with only 
one patient surviving for 10 days. Lidell 



chOl.qxd 4/16/04 3:20PM Page 11 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



11 



Femoral artery 



Femoral vein 




Posterior 



Anterior 




Aneurysmal pockets 
on the anterior and 
posterior surface of 
the femoral a. 



C 






■ FIGURE 1-4 

The first successful end-to-end arterial anastomosis in man by Murphy in 1896. A, The anatomic 
location of the injury. B, The close pathology involved. C, Degree of destruction, portion resected 
and appearance after invagination of femoral artery. See text for details including venous repair. 
(From Murphy JB: Med Rec 1897;51:73-104.) ■ 



reported only 16 recoveries after ligation of 
the common iliac artery 68 times, a mortality 
of 77% . Balance and Edmunds reported a 40% 
mortality following ligation of afemoral artery 
aneurysm in 31 patients. Billroth reported 



secondary hemorrhage from 50% of large 
arteries ligated in continuity. Wyeth collected 
106 cases of carotid artery aneurysms treated 
by proximal ligation, with a mortality rate of 
35%. 



chOl.qxd 5/3/04 4:35PM Page 12 



12 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



In 189 7 Murphy summarized techniques he 
considered necessary for arterial suture. They 
bear a close resemblance to principles gen- 
erally followed today: 

1. Complete asepsis 

2. Exposure of the vessel with as little injury 
as possible 

3. Temporary suppression of the blood 
current 

4. Control of the vessel while applying the 
suture 

5. Accurate approximation of the walls 

6. Perfect hemostasis by pressure after the 
clamps are taken off 

7. Toilet of the wound 

Murphy also reported that Billroth, Schede, 
Braun, Schmidt, and others had successfully 
sutured wounds in veins. He personally had 
used five silk sutures to close an opening 
three eighths of an inch in the common 
jugular vein. Several significant accomplish- 
ments occurred in vascular surgery within 
the next few years. Matas (1903) described 
his technique with endoaneurysmorrhaphy 
for aneurysm, a technique that remained 
the standard technique for aneurysms for 
more than 40 years. In 1906 Carrel and 
Guthrie performed classic experimental 
studies over a period with many significant 
results. These included direct suture repair 
of arteries, vein transplantation, and trans- 
plantation of blood vessels, organs, and limbs 
(Fig. 1-5). 

In 1912 Guthrie independently published 
his continuing work on vascular surgery. Fol- 
lowing Murphy's successful case in 1896, the 
next successful repair of an arterial defect 
came 10 years later when Goyanesused a vein 
graft to bridge an arterial defect in 1906. 
Working in Madrid, Goyanes excised a 
popliteal artery aneurysm and used the accom- 
panying popliteal vein to restore continuity 
(Fig. 1-6). 

He used the suture technique, developed 
by Carrel and Guthrie, of triangulation of the 
arterial orifice with three sutures, followed by 
continuous suture between each of the three 




■ FIGURE 1-5 

The triangulation method of suturing vessels. 
Initially conceived in 1902 by Carrel, this 
method was used by Carrel and Guthrie in their 
monumental contributions in the direct suture 
repair of arteries, vein transplants, and 
transplantation of blood vessels and organs. 
(Courtesy the New York Academy of Medicine 
Library.) ■ 

A year later in Germany, Lexer (1907) first 
used the saphenous vein as an arterial sub- 
stitute to restore continuity after excision of 
an aneurysm of the axillary artery. In his 1969 
review, Shumacker commented thatwithin the 
first few years of this century, the triangula- 
tion stitch of Carrel (1902), the quadrangu- 
lation method of Frouin (1908), and the 
Mourin modification (1914) (Fig. 1-7) had 
developed. 

By 1910 Stich reported more than 100 cases 
of arterial reconstruction by lateral suture. His 
review also included 46 repairs by end-to-end 
anastomosis or by insertion of a vein graft 
(Nolan, 1968). It is curious with this promis- 
ing start that more than 30 years had elapsed 
before vascular surgery was widely employed. 
A high failure rate, usually by thrombosis, 
attended early attempts at repair, and few 
surgeons were convinced that repair of an 
artery wasworthwhile. Matas (1913) stated that 
vascular injuries, particularly arteriovenous 
aneurysms, had become a conspicuous feature 
of modern military surgery, and he felt that 
this class of injury must command the closest 
attention of the modern military surgeon: 



chOl.qxd 4/16/04 3:20PM Page 13 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



13 



Artery 




■ FIGURE 1-6 

The first successful repair of an arterial defect 
utilizing a vein graft. Using the triangulation 
technique of Carrel with endothelial coaptation, 
a segment of the adjacent popliteal vein was 
used to repair the popliteal artery. (From 
Goyanes DJ: El Siglo Med 1906;53:546,561 .) ■ 



A most timely and valuable contribution to the 
surgery of blood vessels resulted from 
wounds in war. . . . Unusual opportunities for 
the observation of vascular wounds inflicted 
with modern military weapons . . . based on 
material fresh from the field of action, and 



fully confirmed the belief that this last war, 
waged in closed proximity to well equipped 
surgical centers, would also offer an 
unusual opportunity for the study of the 
most advanced methods of treating injuries 
of blood vessels. 



MILITARY VASCULAR 
TRAUMA EXPERIENCE 



Balkin Wars 

In 1913 Soubbotitch (Fig. 1-8) described the 
experience of Serbian military surgeons 
during the Serbo-Turkish and Serbo-Bulgar- 
ian Wars. Seventy-seven false aneurysms and 
arteriovenous fistulas were treated. There were 
45 ligations, but 32 vessels were repaired, 
including 19 arteriorrhaphies, 13 venorrha- 
phies, and 15 end-to-end anastomoses (11 
arteries and 4 veins) . It is impressive that infec- 
tion and secondary hemorrhage were avoided. 
Matas (1913), in discussing Soubbotitch's 
report, emphasized that a notable feature was 
the suture (circular and lateral repair) of 
blood vessels and the fact that it had been used 
more frequently in the Balkan Conflict than 
in previous wars. He also noted that judging 
by Soubbotitch's statistics, the success obtained 
by surgeons in the Serbian Army Hospital in 
Belgrade far surpassed that obtained by other 
military surgeons in previous wars, with the 
exception perhaps of the remarkably favor- 
able results in the Japanese Reserve Hospital 
reported by Kikuzi during the Russo- 
Japanese War (1904/1905). It is ironic that 





■ FIGURE 1-7 

The original triangulation stitch 
of Carrel in 1902 was modified 
to a quadrangulation method 
by Frouin in 1908. Another 
modification, as shown here, 
was that of Mourin in 1914. 
(From Moure P. Les Greffes 
Arterielles, 1914.) ■ 



chOl.qxd 4/16/04 3:20PM Page 14 



14 I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 




■ FIGURE 1-8 

Dr. V. Soubbotitch (front-row center), Lt Col., Serbian Army Reserves, is flanked by members of his 
staff at the Belgrade State Hospital (circa 1912-1913). The reference provides additional details 
regarding the Matas-Soubbotitch connection. (From Rich NM, Clagett GP, Salander JM, Piscevic S: 
Surgery 1983;93:17-19.) ■ 



the vascular experience continued in the 
Balkans, in the early 1990s. Additional infor- 
mation regarding Soubbotitch came directly 
from Geza De Takats (Fig. 1-9) who was in 
Belgrade early in World War II. Table 1-2 iden- 
tifies the Soubbotitch experience. 



World War I Experience 

During the early part of World War I, with the 
new techniques of vascular surgery well estab- 
lished, the German surgeons attempted repair 
of acutely injured arteries and were success- 
ful in more than 100 cases (Nolan, 1968). 
During the first 9 months of World War I, low- 
velocity missiles caused arterial trauma of a 



TABLE 1- 


-2 




TREATMENT OF TRAUMATIC 


ANEURYSMS FROM THE SERBO- 


TURKISH AND SERBO-BULGARIAN 


WARS 










Partial Circular 




Ligation 


Suture Suture 


Arteries 


41 


8 11 


Veins 


4 


9 4 



Modified from Soubbotitch V: Lancet 1913:2:720-721. 



chOl.qxd 4/16/04 3:20PM Page 15 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



15 



MILITARY SURGICAL HERITAGE 

DEPARTMENT OF SURGERY, USUHS 

Geza de Takats 1892 - 1985 




1st. LT, MC Austro-Hungarian Army 
At Funeral of Emperor Franz Joseph 1916 
A Dedicated Friend of Military Surgeons 
Consultant, Great Lakes Naval Hospital, 1946 - 



■ FIGURE 1-9 

An internationally acclaimed pioneer 
in vascular surgery Geza De Takats is 
among the early contributors to our 
military surgical heritage. (From Rich 
NM: Am J Surg 1993;166:91-96.) ■ 



limited extent. In 1915, however, the wide- 
spread use of high explosives (the high explo- 
sive artillery shells replaced the Shrapnel shell 
in use since British action in Surinam in 1904) 
and high-velocity bullets, combined with mass 
casualties and slow evacuation of the wounded, 
made arterial repair impractical. 

Bernheim (1920) , who had performed vas- 
cular research in a Hunterian Laboratory, per- 
formed the first vascular repair using 
saphenous vein in the United States at Johns 
Hopkins University and went to France with 
the specific intent of repairing arterial injuries. 
Despite extensive prior experience and 
equipment, however, he concluded that 
attempts atvascular repair were unwise (1920) . 
He wrote 



Opportunities for carrying out the more 
modern procedures for repair or 
reconstruction of damaged blood vessels 
were conspicuous by their absence during 
the recent military activities. . . . Not that 
blood vessels were immune from injury; not 
that gaping arteries and veins and 
vicariously united vessels did not cry out for 
relief by fine suture or anastomosis. They 
did, most eloquently, and in great numbers, 
but he would have been a foolhardy man 
who would have essayed sutures of arterial 
or venous trunks in the presence of such 
infections as were the rule in practically all 
of the battle wounded. 



chOl.qxd 4/16/04 3:20PM Page 16 



16 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



The great frequency of infection with sec- 
ondary hemorrhage virtually precluded arte- 
rial repair. In addition, there were inadequate 
statistics about the frequency of gangrene fol- 
lowing ligation, and initial reports subse- 
quently proved to be unduly optimistic. Poole 
(1927), in the Medical Department History 
of World War I, remarked that if gangrene 
was a danger following arterial ligation, 
primary suture should be performed and the 
patient watched very carefully. 

Despite the discouragement of managing 
acute arterial injuries in World War I, fairly 
frequent repair of false aneurysms and arte- 
riovenous fistulas was carried out by many 
surgeons. These cases were treated after the 
acute period of injury, when collateral circu- 
lation had developed with the passage of time 
and ensured viability of extremities. Matas 
(1921) recorded that most of these repairs 
consisted of arteriorrhaphy by lateral or 
circular suture, with excision of the sac or 
endoaneurysmorrhaphy. 

Makins (1919), who served in World 
War I as a British surgeon, recommended 
ligating the concomitant vein when it was 
necessary to ligate a major artery. He 
thought that this reduced the frequency of 
gangrene. This hypothesis was debated 
for more than 20 years before it was finally 
abandoned. 

Goodman (1918), in describing his expe- 
rience at the number 1 (Presbyterian U.S.A.) 
General Hospital in France during World War 
I, reported a successful closure with continu- 
ous silk suture of 5-mm longitudinal openings 
in both the popliteal artery and the popliteal 
vein in one patient with a shell fragment. 
However, the patient was followed for only 9 
days before being transferred to the Base 
Hospital. Goodman reported, "An attempt to 
obtain further information covering the case 
is now underway and will be embodied in a 
subsequent report." 

If there was any further follow-up infor- 
mation obtained or reported, it became 
obscured in the available literature. At least 
this military surgeon recognized the impor- 
tance of obtaining long-term follow-up infor- 
mation to thoroughly evaluate his method of 
managing vascular trauma. 



In 1987, Shumacker identified that 
Weglowski was a neglected pioneer in vascu- 
lar surgery. Weglowski served first in a Russian 
Military Hospital and later as Surgeon General 
of the Polish Army. Based on his experience 
with more than 600 patients, he summarized 
his recommendations in 1919 that all arterial 
injuries and post-traumatic aneurysms, includ- 
ing those of the aorta, carotid, iliac, and sub- 
clavian arteries, and the arteries of the 
extremities, should be repaired by vascular 
suture either immediately after the injury or 
after 1 month for pulsating hematomas. In 
1924 Weglowski presented the results of 193 
personal vascular repairs including 46 by 
lateral sutures, 12 by end-to-end anastomosis 
and 56 using venous grafts. Ligation was 
required in the remaining 79 patients because 
of infection and the risk of postoperative 
bleeding. His results were surprisingly good. 
In 1994, Nunn wrote in detail about Ernst 
Jeger, who he called a "forgotten pioneer in 
cardiovascular surgery." Jeger's work was 
recorded only in the German language. Jeger 
described his research in a book, Die Chirgiirie 
der Blutgefasse und des Herzens in 1913. He was 
drafted as a physician for the German Army 
in 1914 and died tragically in a Russian prison 
camp in 1915. Jeger did report his operative 
experience with 10 soldiers with vascular 
injuries. He had success in seven patients using 
lateral suture or end-to-end anastomosis. 
Based on his successful experience, he rec- 
ommended increased use of vascular repair 
in war injuries. 



World War II Experience 

Experiences with vascular surgery in World 
War II are well recorded in the classic review 
of DeBakey and Simeone (1946), analyzing 
2471 arterial injuries. Almost all were treated 
by ligation, with a subsequent amputation rate 
near 49%. Only 81 repairs were attempted, 
78 by lateral suture and 3 by end-to-end anas- 
tomosis, with an amputation rate of approxi- 
mately 35%. The use of vein grafts was even 
more disappointing: They were attempted in 
40 patients, with an amputation rate of nearly 
58%. Early patency of a venous graft in the 



chOl.qxd 4/16/04 3:20PM Page 17 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



17 



arterial system was demonstrated angio- 
graphically. DeBakey (Fig. 1-10) has had the 
opportunity to contribute to the development 
and progress in vascular surgery over more 
than 50 years, from 1946 to 1996. 

The controversial question of ligation of the 
concomitant vein remained, although few 
observers were convinced that the procedure 
enhanced circulation. The varying opinions 
were summarized by Linton in 1949. 

A refreshing exception to the dismal World 
War II experience in regard to ligation and 
gangrene was the case operated on by Doctor 
Allen M. Boyden: an acute arteriovenous 
fistula of the femoral vessels repaired shortly 
after D-Day in Normandy. The following com- 
ments are taken from his field notes about 26 



years later Boyden (personal communication, 
1970) and emphasize the value of adequate 
records, even in military combat: 



High explosive wound left groin, 14 June 

1944, at 2200 hours. Acute arteriovenous 

aneurysm femoral artery 
Preoperative blood pressure 140/70; pulse 

104 
Operation: 16 June 1944, nitrous oxide and 

oxygen 
Operation: 1910 to 22 hours 
One unit blood transfused during the 

operation 

Continued 



MILITARY SURGICAL HERITAGE 

Department of Surgery, USUHS 

Michael E. DeBakey, MD 




Colonel, MC, AUS Third AMEDD Surgical Consultant 1946 

Consultant Department of Surgery USUHS, 1977- 

Visiting Board Department of Surgery USUHS, 1978- 

Advisor, USU Surgical Associates, 1980- President 1990 

Michael E. Debakey International Military Surgeon's Award 



■ FIGURE 1-10 

Doctor DeBakey is recognized for 
his numerous and valuable 
contributions to surgery, in general, 
and specifically to military vascular 
surgery. The Michael DeBakey 
International Military Surgeons 
Award is presented at the Uniformed 
Services University Surgical 
Associates Day each spring at the 
Uniformed Services University of the 
Health Sciences, Bethesda, 
Maryland. (From Rich NM: Am J 
Surg 1993;166:91-96.) ■ 



chOl.qxd 4/16/04 3:20PM Page 18 



18 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



Arteriovenous aneurysms isolated near 

junction with profunda femoris artery 
Considerable hemorrhage 
Openings in both artery and vein were sutured 

with fine silk 
Postoperative blood pressure 120/68; pulse 

118 
Circulation of the extremity remained intact 

until evacuation 



As this case demonstrated Boyden's inter- 
est in vascular surgery, the Consulting Surgeon 
for the First Army presented him with one half 
of the latter's supply of vascular instruments 
and material. This supply consisted of two sets 
of Blakemore tubes, two bulldog forceps, and 
a 2-mL ampule of heparin! 

The conclusion that ligation was the treat- 
ment of choice for injured arteries was 
summarized by DeBakey and Simeone in 1946, 
"It is clear that no procedure other than lig- 
ation is applicable to the majority of vascular 
injuries which come under the military sur- 
geons' observation. It is not a procedure of 
choice. It is a procedure of stern necessity, for 
the basic purpose of controlling hemorrhage, 
as well as because of the location, type, size 
and character of most battle injuries of the 
arteries." 

In retrospect it should be remembered that 
the average time lag between wounding and 
surgical treatment was more than 10 hours in 
World War II, virtually precluding successful 
arterial repair in most patients. Of historical 
interest is the nonsuture method of arterial 
repair used during World War II (Figs. 1-11 
and 1-12). 

Although considerable time and effort 
were expended following World War II in an 
attempt to provide additional long-term 
follow-up information, the results of this 
effort are not generally available. Individual 
follow-up has been possible in a random way 
for some patients, such is the case of an acute 
femoral arteriovenous fistula that was repaired 
shortly after D-Day in Normandy on June 16, 
1944 (Boyden, 1970) . This was an unusual case 
because it involved the successful repair of 
both the common femoral artery and the 




■ FIGURE 1-11 

Completed unsutured vein graft of the popliteal 
artery which was complicated by a severe 
compound fracture of the tibia. This was 
representative of 40 cases utilizing the double- 
tube graft technique in World War II as 
advocated by Blakemore, Lord and Stefko in 
1942. (From DeBakey ME, Simeone FA: Ann 
Surg 1946;123:534-579.) ■ 



common femoral vein in a combat zone at a 
time when ligation of vascular injuries was the 
accepted principle. Unfortunately, when it was 
possible more than 26 years later to obtain 
follow-up data on this patient, it was learned 
that hemorrhage occurred in the left inguinal 
wound after the patient was evacuated to a 
General Hospital in England. Ligation of both 
the common femoral artery and the common 
femoral vein was required. Additional follow- 
up was not possible because the patient died 
of tuberculosis approximately 16 months 
after receiving the wound. Shumacker (1946, 
1947a, 1947b, 1948a, 1948b) (Fig. 1-13) 
(Table 1-3) made many valuable contribu- 
tions to vascular surgery with the U.S. Army, 
as did Rob (Fig. 1-14) with the British Army. 



Experiences During the 
Korean Conflict 

The successful repair of arterial injuries in the 
Korean Conflict, in pleasant contrast to the 
experiences of World War I and World War 



chOl.qxd 4/16/04 3:20PM Page 19 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



19 





Proximal 

S^rRubber shod 
(Artery c| a m P 





Kelly clamp 

Distal end of 
vein placed into 
Distal proximal end of artery 
5 



■ FIGURE 1-12 

The various steps of a nonsuture method 
of bridging arterial defects designed 
during World War II (1)The Vitallium tube 
with its two ridges (sometimes grooves). 
(2) The exposed femoral artery and vein 
retracted and clamps placed on a 
branch. (3) The removed segment of 
vein is irrigated with saline solution. (4) 
The vein has been pushed through the 
inside of the Vitallium tube, and the two 
ends everted over the ends of the tube 
held in place with one or two ligatures of 
fine silk. (5) Distal end of the segment of 
vein is placed into the proximal end of 
the artery and held there by two ligatures 
of fine silk. (6) The snug ligature near the 
end of the Vitallium tube is tied to 
provide apposition of the artery and vein. 
(7) The completed operation, showing 
the bridging of a 2-cm gap in the femoral 
artery. (From Blakemore AH, Lord JW, 
Stefko PL: Surgery 1942;12:488-508.) ■ 



MILITARY SURGICAL HERITAGE 

DEPARTMENT OF SURGERY, USUH8 



Harrli B Shumlcktl, Jr. 




Captain, 118th General, (Johns Hopkins) 

Camp Edwarda, Cap* Cod 1942 

Prolasaor of Surgery, USUHS 

1 July 1081 - 



■ FIGURE 1-13 

Doctor Harris Shumacker made 
many valuable contributions in World 
War II to the early development of 
military vascular surgery. 
Subsequently, he received 
appropriate recognition by being 
named the first, and only, 
distinguished professor in the 
Department of Surgery at the 
Uniformed Services University of the 
Health Sciences, Bethesda, 
Maryland. (From Rich NM: Am J 
Surg 1993;166:91-96.) ■ 



chOl.qxd 4/16/04 3:20PM Page 2 



20 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



Lil 

<r 

Z> 
H 
Z> 
CO 

Q 

z 

LU 

I 

o 



LU 

>- 
CD 

Q 
LU 

< 
Q. 
LU 

cr 

CO 

I 

> 

cr 

LU 

H 

cr 
< 

x 
o 

X 



CO 

I 



CO 
LU 



03 W 
< < 

H O 



3 
CO 
0) 



(0 

Q. 
3 ^ 

* P 
o =. 



CD 

> g 

S o 

if 

a> >> 
A- W 



o 

0) 

Q. 



o 

o .2 o 

B (0 C 
(B 0) £. 

3- 1 
D 



< Q. 



a) 

(A 

<B o 

o z 



« J J 

"0 ~cd "0 
o o o 



"a -a 

-„ 

m c m S< 

5 -BE | 

E « E => 

o z o c 

£ « Jz § 

^ u tr o 

o ra -a 



C £Z C 

b „ 

o o o oi(3 n i 

X X X CD C 

LU LU LU CO Q_ 



C\J C\J C\J -r- 



■o 
a> 

o -2 

_ ° — 



LO LO LO 



LO CM LO 
-<- CXI t- 



c 










"a 




"D 




o 


TJ 


T3 






.i= 




.^= 


"a 


<D 










JZ 


"D 


JZ 




JZ 


_E 


CO 




*-' 


.?= 


^ 


JZ 






E 




0) 


JZ 


CD 




















■i 


CO 


CO 


z* 




"a 


— 


"a 


CO 


O 


"co 


"co 


o 




"a 


£ 


"a 


"co 


£ 
O 


XI 


TJ 


Q_ 




E 


to 


E 


TJ 


"co 


"CO 


"CO 




"co 




"co 


"CO 


o 


JZ 


JZ 


JZ 


TJ 


JZ 


JZ 


JZ 


CJ 


CJ 


o 


JZ 


CJ 


o 


CJ 





CO 


CO 


CO 


CO 




CO 


CO 


_l 












x; 








m 


CD 


CO 




CD 


< 


CD 


CD 



r- lo 

LD N CO 



LO LO 

CO LO CD CO 



o lo h- 


CO CO O) 'sT 


CM CO CM 


CM CM 1- CM 



< 



E 


E 


E 


E 


E 


E 


E 


E 


o 
o 


co 


<n 


CO 


CO 


CO 


CO 


<n 


CO 


^ 


>^ 


>. 


>^ 


>> 


^ 


>^ 


^ 


CO 


















CO 


z 


z 


z 


z 


z 


z 


z 


z 


CO 





CD 


CD 







c 











23 


c 


c 


c 


c 


z 


c 


c 


o 
c 


CO 


CO 


CO 


CO 


CO 


CO 


CO 


CO 





















_C0 


_C0 


_C0 


_C0 


_C0 


CO 


CO 


CO 


> 


z 


z 


z 


z 


z 


z 


z 


z 


,o 


o 


CJ 


o 


CJ 


CJ 


CJ 


CJ 


CJ 




o 


CJ 


CJ 


CJ 


CJ 


CJ 


CJ 


CJ 





CO 


CO 


CO 


CO 


CO 


CO 


CO 


CO 




CO 


co 


CO 


CO 


CO 


CO 


co 


CO 


< 



CO 

o ^ 
£ 2 

CO C\J 



CO 

"a 



CO 


m 


> 








m 




m 


n 





o 


b 


ffl 


fc 



< 



^ in (D n 



E ra 



E S 

co 
0j >• 



n3 o 
n3 D) 



0) co 

« E 



D_ OT 



> X ■ 



CD 







c/3 o r^ 

5; co cm 

o E '- 

E ^ o> 



<5 E 
< 2 



C/3 



chOl.qxd 4/16/04 3:20PM Page 21 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



21 



MILITARY SURGICAL HERITAGE 

DEPARTMENT OF SURGERY, USUHS 



Charla* G. Rob 




ilt Royal Amy Mit Tc al Corps 

Ti w a ar l , North Africa January 1943 

Prof»»«or of Surgery, USUHS 

1 July 19B3 - 



■ FIGURE 1-14 

Doctor Charles Rob has had a 
distinguished career, both in the 
United Kingdom and in the United 
States; he has been recognized for 
his international contributions in 
vascular surgery. Dr. Rob continues 
to serve as professor and senior 
advisor at the Uniformed Services 
University of the Health Sciences, 
Bethesda, Maryland. (From Rich 
NM: Am J Surg 1993;166:91-96.) ■ 



II, was due to several factors. There had been 
substantial progress in the techniques of vas- 
cular surgery, accompanied by improvements 
in anesthesia, angiography, blood transfusion, 
and antibiotics. Perhaps of the greatest impor- 
tance was rapid evacuation of wounded men 
often by helicopter, permitting their transport 
from time of wounding to surgical care often 
within 1 to 2 hours (Fig. 1-15). In addition, 
a thorough understanding of the importance 
of debridement, delayed primary closure, and 
antibiotics greatly decreased the hazards of 
infection. 

Initially in the Korean Conflict, attempts at 
arterial repair were disappointing. During one 
report of experiences at a surgical hospital 
for 8 months between September 1951 and 



April 1952, only 11 of 40 attempted arterial 
repairs were thought to be successful (Hughes, 
1959). Only 6 of 29 end-to-end anastomoses 
were considered initially successful, and all 6 
venous grafts failed. In another report from 
a similar period, only 4 of 18 attempted repairs 
were considered successful. Warren (1952) 
emphasized that an aggressive approach was 
needed, with the establishment of a research 
team headed by a surgeon experienced in vas- 
cular grafting. Surgical research teams were 
established in the Army, and there was 
improvement in results of vascular repairs in 
1952. Significant reports were published by 
Jahnke and Seeley (1953), Hughes (1955, 
1958), and Inui, Shannon, and Howard 
(1955). Hughes (Fig. 1-16) continued his 



chOl.qxd 4/16/04 3:20PM Page 22 



22 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



■ FIGURE 1-15 

Helicopter evacuation of the wounded during the 
Korean Conflict helped reduce the lag time 
between injury and definitive surgical care. 
Continued improvement in Vietnam in helicopter 
evacuation of the wounded allowed some 
patients with vascular trauma to reach a 
definitive surgical center within 15 to 30 minutes. 
(US Army photograph.) ■ 




MILITARY SURGICAL HERITAGE 

DEPARTMENT OF SURGERY, USUHS 



■ FIGURE 1-16 

Doctor Carl Hughes had a 
distinguished military career 
providing valuable contribution 
during the Korean Conflict and, 
subsequently, during the Vietnam 
War. He continues to serve as 
professor at the Uniformed Services 
University of the Health Sciences, 
Bethesda, Maryland. (From Rich 
NM. Am J Surg 1993;166:91-96.) ■ 




WS5 HASH, CNH Won ¥■»•», Kor*» 1MS 

MG, MC, US* (RET) 
Pr o Umr ot lan)MV, USUHS 1M4-1MC 
1 twawy, USUHS ISM- 



chOl.qxd 4/16/04 3:20PM Page 23 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



23 



efforts in vascular surgery rising to the rank 
of Major General. He continues at the Uni- 
formed Services University of the Health Sci- 
ences, a Distinguished Professor of Surgery. 
Howard reflected in 1998 on his clinical and 
research experiences during the Korean Con- 
flict. Similar work in the Navy was done with 
the U.S. Marines during 1952 and 1953 by 
Spencer and Grewe (1955). These surgeons 
worked in specialized research groups under 
fairly stabilized conditions, considering that 
they were in a combat zone (Fig. 1-17). 
Spencer reflected on his experiences during 
the Korean Conflict during his Presidential 
Address to the American Surgical Association 
in 1998. 

Brigadier General Sam Seeley, who was 
Chief of the Department of Surgery at Walter 
Reed Army Hospital in 1950, had the fore- 
sight to establish Walter Reed Army Hospital 
as a vascular surgery center, and this made it 
possible for patients with vascular injuries to 
be returned there for later study (Fig. 1-18) . 
In a total experience with 304 arterial injuries, 
269 were repaired and 35 ligated (Hughes, 
1958). The overall amputation rate was 13%, 
a marked contrast to that of about 49% in 
World War II. Because the amputation rate is 
only one method of determining ultimate 



Rights were not granted to include this figure in electronic media. 
Please refer to the printed publication. 



■ FIGURE 1-18 

An autogenous greater saphenous vein graft 
was utilized in 1952 at Walter Reed General 
Hospital to repair a traumatized proximal 
popliteal artery. Each anastomosis is an 
everting type with intima-to-intima held by 
everting mattress sutures. (Rich NM, Hughes 
CW. Bull Am Coll Surg 1972;57:35.) ■ 



success or failure in arterial repair, it is impor- 
tant to emphasize thatjahnke (1958) revealed 
that in addition to the lowered rate of limb 
loss, limbs functioned normally when arter- 
ial repair was successful. The arteriovenous 
fistula experience is expanded on in 
Chapter 24. 




■ FIGURE 1-17 

Postoperative ward in a mobile army surgical 
hospital (MASH) shows some of the conditions 
at the time of the Korean Conflict when it was 
demonstrated that arterial repair could be 
successful, even under battlefield conditions. 
(Hughes CW. Milit Med 1959;124:30-46.) ■ 



Experience in Vietnam 

In Vietnam the time lag between injury and 
treatment was reduced even further by the 
almost routine evacuation by helicopter, com- 
bined with the widespread availability of sur- 
geons experienced in vascular surgery. In one 
study of 750 patients with missile wounds in 
Vietnam, 95% of the patients reached the hos- 
pital by helicopter (Rich, 1968) (Fig. 1-18). 
This prompt evacuation, however, similarly 
created an adverse effect on the overall 
results, for patients with severe injuries from 
high-velocity missiles survived only long 
enough to reach the hospital. During initial 
care, they expired. These patients would 
never have reached the hospital alive in pre- 
vious military conflicts. 



chOl.qxd 4/16/04 3:20PM Page 24 



24 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



In the initial Vietnam studies, between 
October 1, 1965,andjune30, 1966, there were 
177 known vascular injuries in U.S. casualties, 
excluding those with traumatic amputation 
(Heaton and colleagues, 1966) . One hundred 
sixteen operations were performed on 106 
patients with 108 injuries (Table 1-4). These 
results included the personal experience of 
one of us (N.M.R.) at the Second Surgical Hos- 
pital. The results reported included a short- 
term follow-up of approximately 7 to 10 days 
in Vietnam. In Vietnam, amputations were 
required for only 9 of the 108 vascular injuries, 
a rate of about 8%. Subsequently, more 
detailed analysis from the Vietnam Vascular 
Registry (Rich and Hughes, 1969; Rich, 1970) 
found the amputation rate of approximately 
13%, identical to that of the Korean Conflict. 
Almost all amputationswere performed within 
the first month after wounding. 

The Vietnam Vascular Registry (Figs. 1-19 
and 1-20) was established at Walter Reed 
General Hospital in 1966 to document and 
analyze all vascular injuries treated in Army 
Hospitals in Vietnam. A preliminary report 




■ FIGURE 1-19 

During the war in Vietnam, most patients were 
rapidly treated in fixed installations. An early 
example is the 2 nd Surgical Hospital at An Khe 
in January, 1966. Ninety-five per cent of the 
wounded reached a hospital by helicopter. 
(Rich NM, Georgiade NG. Plastic and 
Maxillo-facial Trauma Symposium. CV Mosby, 
St Louis, 1969.) ■ 



(Rich and Hughes, 1969) involved the com- 
plete follow-up of 500 patients who sustained 
718 vascular injuries (Table 1-5). Although 
vascular repairs on Vietnamese and allied mil- 
itary personnel were not included, the Reg- 
istry effort was soon expanded to include all 
U.S. service personnel, rather than limiting 
the effort to soldiers. 

Fisher (1967) collected 154 acute arterial 
injuries in Vietnam covering the 1965 to 1966 
period. There were 108 arterial injuries with 
significant information for the initial review 
from Army hospitals. In 1967, Chandler and 
Knapp reported results in managing acute vas- 
cular injuries in the U.S. Navy hospitals in 
Vietnam. These patients were not included in 
the initial Vietnam Vascular Registry report, 
but after 1967, an attempt was made to 
include all military personnel sustaining 
vascular trauma in Vietnam. This included 
active-duty members of the U.S. Armed 
Forces treated at approximately 25 Army 
hospitals, six Navy hospitals, and one Air Force 
hospital. 

As with any registry, success of the Vietnam 
Vascular Registry has depended on the coop- 
eration of hundreds of individuals within the 
military and civilian communities. In the initial 
report from the Registry, the names of 20 sur- 
geons who had done more than five vascular 
repairs were included (Rich and Hughes, 
1969) . In the first edition of Vascular Trauma, 
the names of many more who contributed to 
the Vietnam Vascular Registry are included 
(Rich and Spencer, 1978). 

We would be remiss if we did not gain some- 
thing positive from an experience with as many 
negative aspects as the U.S. involvement in 
Southeast Asia between 1965 and 1972. The 
Vietnam Vascular Registry provides a unique 
opportunity for long-term follow-up of thou- 
sands of young men with vascular repairs. The 
challenge remains and the potential is great. 
Additional historical details regarding the Reg- 
istry activities are included in the first edition 
of Vascular Trauma. Unfortunately, the major- 
ity in the United States did not want to hear 
the word Vietnam for nearly 25 years. Only 
recently has the value of the Vietnam Vascu- 
lar Registry been appreciated appropriately 
and it is hoped that the efforts can be 
completed. 



chOl.qxd 4/16/04 3:20PM Page 25 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



25 



CO 

DL 
CO 

o 

I 

>- 

DC 
< 

CO 
LU 

dc 

3 



< 

DC 
LU 

H 
DC 
< 
00 

o 



UJ 

> 



>~ 

DC 
LU 
(5 
DC 

CO 
DC 
< 

_l 
Z> 
O 
CO 

£ 

X 



LU 

o 

z 

LU 

DC 
LU 
Dl 
X 
LU 

> 



oa cc 







E 






< 




C 






o 












4-1 




(A 


ra 


, . 


ii 


3 


a) c 


3 


Q. 


to a) 


(0 

cc 


E 
< 

6 

z 


3 <n 

li 

jS 'to 

O CL 

F a> 
CC 

CO cfci 

o to 

E 
o 
o 

tD 

XI 

E 
o 

_c 

H 

i- to 

3q? 


i- 












IB 






a. 
a> 






cc 




to 


o 




<5 


a> 




c 


a 




5 


H 




to 
'to 
o 
E 
o 
"to 
to 
c 

< 

M_ tO 

o a> 
6 5 

£• 
o> 

c 
< 



oo-^oooocoldioi 



o^cooooo^-'-ieo 



N030)^^^N(DO|CO 



h~ CT> CD t- t- ^t- 00 



OOOOO-'-OOOI-i- 



OO^OOOOO^ICM 



fOCMOlO^OT-CDLOIN 



rat^ooi-^nNin 



T-COlfli-OWCO^N 



8 E 

o E « 

CD c to £ — 

ti CO = ,- .tO 



o >..E5 

c -3 o 

o ^ 2 

O < CD 



CO 



c 'o CO 

t CD 

c CD = 

D X O IS O 

C/3 LU O CO D_ 



CO o 

E E 

CD 



E 
< 



"D £5 



I 

E 



chOl.qxd 4/16/04 3:20PM Page 2 6 




ACUTE ARTE8IAL INJURIES IN VIETNAM 




■ a 



IS 



■ FIGURE 1-20 

4, This exhibit representing the management of 
acute arterial trauma in Vietnam was presented 
from material in the Vietnam Registry to the 
Clinical Congress of the American College of 
Surgeons in Chicago in 1970. At least 1 10 
surgeons who had previously performed arterial 
repairs in Vietnam visited the exhibit. B, 
Identification card sent to armed forces 
personnel who were wounded in Vietnam. This 
card was issued in an attempt to identify 
participation in the Registry and with the hope 
that additional long-term follow-up information 
will be generated. (A, From A. F.I. P. photograph; 
B, From Walter Reed General Hospital.) ■ 



VIETNAM VASCULAR REGISTRY 



CERTIFY THAT 




HAS A HtWMAWtNT FILE AT 
B WALTER REED GENERAL HDSPITAL. 



TABLE 1-5 

MANAGEMENT OF ARTERIAL TRAUMAIN VIETNAM CASUALTIES PRELIMINARY 
REPORT FROM THE VIETNAM VASCULAR REGISTRY* 





End-to-End 


Vein 


Lateral 


Prosthetic 






Artery 


Anastomosis 


Graft 


Suture 


Graft 


Thrombectomy 


Ligation 














^H 


Common carotid 


2 


6(2) 


3 




(2) 


1 


Internal carotid 






2 






1 


Subclavian 


1 












Axillary 


6(3) 


12(3) 


2(3) 


(D 


(3) 


(1) 


Brachial 


57(8) 


32(10) 


2(1) 




1(9) 


1(2) 


Aorta 






3(1) 








Renal 












1 


Iliac 


1 


1 




1(1) 


(1) 


(1) 


Common femoral 


4(2) 


11(1) 


4(1) 


1(2) 


(2) 


(4) 


Superficial femoral 


63(5) 


37 (14) 


7(7) 


(4) 


2(6) 


(4) 


Popliteal 


31(5) 


28(13) 


6(4) 




(10) 


2(4) 


TOTAL 


165 (23) 


127 (43) 


29(17) 


2(8) 


3(33) 


6(16) 



'Numbers in parenthesis represent additional procedures performed after the initial repair in Vietnam and repair of major arterial 

injuries not initially treated in Vietnam. 

Modified from Rich NM, Hughes CE: Vietnam vascular registry: a preliminary report. Surgery 65:218-226, 1969. 



chOl.qxd 4/16/04 3:20PM Page 27 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



27 



TABLE 1-6 

LOCATION OF EXTREMITY VENOUS INJURIES IN VIETNAM (1965-1969), ISRAEL 
(1 973), LEBANON (1 969-1 982), AND CROATIA (1 991 -1 992) 





Vietnam (%) 


Israel (%) 


Lebanon (%) 


Croatia (%) 


Vein 


(n 


= 361) 


(n = 26) 


(n 


= 348) 


(n = 41) 


Subclavian 




1 


8 




3 


— 


Axillary 




6 




3 


10 


Brachial 




15 


31 




— 


20 


Iliac 




3 


— 




10 


12 


Femoral 




43 


35 




51 


39 


Popliteal 




32 


27 




32 


20 



From Leppaniemi A, Rich NM, Browner BD (eds): Techniques in Orthopaedics, vol 10, pp 265-271, 1995, Philadelphia, JB. 
Lippincott. 



Military Armed Conflicts 
Following Vietnam 

From Beruit (1982) to Grenada (1983), 
Panama (1989), the GulfWar (1991) , Somalia 
(1992), as well as recent experiences in 
Croatia, Rwanda, and Haiti (and even the 
2001 /2002 War against Terrorism) , no United 
States military surgeons have had more than 
an antidotal vascular case or two to manage. 
The data from Vietnam remain pertinent and 
valuable today! Leppaniemi (1995) makes 
comparisons between Vietnam and recent 
wars in Israel (1973), Lebanon (1969tol982) 
and Croatia (1992) (Table 1-6). Roostar 
(1995) adds the Soviet experience in 
Afghanistan in the early 1980s (Table 1-7). 
There are military sources such as Croatia and 
Serbia; however, these are generally limited 
in coverage considering the large numbers of 
casualties. The following report is outlined 
because of the information contained. Luetic 
and colleagues, at the "Doctor Ozren Novosel" 
Clinical Hospital at the University of Zagreb, 
has documented experience in the manage- 
ment of military vascular injuries in Croatia. 
He presented a single center experience in 
the recent conflict, documenting results from 
April through December 1991. Luetic and col- 
leagues (1993) managed 1020 casualties with 
76 patients sustaining 120 vascular injuries. 
This is a relatively high 7.5% of the casualties 
with vascular trauma. Also, patients averaging 



TABLE 1-7 

LOCATION OF INJURIES IN THE 
AFGHANISTAN WAR 









Artery 




Location 


Artery 


Vein 


and Vein 


Total 


Carotid 


3 


— 


1 


4 


Subclavian 


10 


2 


3 


15 


Axillary 


6 


— 


1 


7 


Brachial 


39 


— 


6 


45 


Radial 


7 


— 


1 


8 


Iliac 


5 


— 


2 


7 


Femoral 


45 


9 


22 


76 


Popliteal 


19 


2 


1 


22 


Tibial 


22 


— 


— 


22 


TOTAL 


156 


13 


37 


206 



Modified from Roostar L: Treatment plan used for vascular 
injuries in the Afghanistan war. Cardiovasc Surg 3:42-45, 
1995. 



1.58 vascular injuries is a very high percent of 
multiple injuries. It is particularly pertinent 
to note that the casualties were transported 
after initial treatment in forward surgical facil- 
ities, reaching the university hospital within 
3 to 18 hours, with a mean time of 7 hours. 
The most common injuries were to the 
popliteal artery (12.5%) and the brachial veins 
(10%). There was a relatively high incidence 
of concomitant fractures, occurring in 90.4% 
of the cases. They routinely employed exter- 
nal fixation of the concomitant fractures. They 



chOl.qxd 4/16/04 3:20PM Page 28 



28 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



used venous interposition grafts in 45 arter- 
ial injuries and 20 venous injuries. Prostheses 
were used in only three arterial injuries. They 
did have a relatively high incidence of arteri- 
ovenous fistulas and pseudoaneurysms, with 
the former occurring in seven patients and 
the latter in six patients (9.8%) with one 
patient having injury to the popliteal artery 
and five having injuries to the superficial 
femoral artery. Sepsis, deep venous throm- 
bosis, and extensive myonecrosis contributed 
to the required amputations, with three 
patients not receiving definitive surgical repair 
until 12 hours after injury. All patients requir- 
ing amputations had concomitant injury to 
bone, nerve, soft tissue, veins, and arteries. 
There was a mortality rate of 3.9%, with three 
patients dying. 



CIVILIAN VASCULAR INJURIES 



Several differences exist between civilian and 
military vascular injuries. First, military injuries 
are characteristically in young persons without 
arterial disease. They frequently result from 
high-velocity missiles with extensive soft tissue 
destruction, often with injuries of multiple 
organ systems and in circumstances in which 
surgical treatment is less than ideal. Civilian 
injuries, however, are usually from wounds 
associated with minimal soft tissue destruction. 
Prompt treatment and excellent hospital facil- 
ities are usually available. Although young civil- 
ians are commonly injured, there is also a 
significant percentage of older patients who 
often have preexisting arterial disease. In addi- 
tion, there are frequent injuries from blunt 
trauma, such as automobile or industrial acci- 
dents, and fractures of long bones. Finally, an 
increasing number of vascular injuries are 
being seen as a complication of diagnostic pro- 
cedures involving cannulation of peripheral 
arteries, as in angiography or cardiac 
catheterization. 



The frequency of arterial injuries in civil- 
ian life has increased greatly in the past decade. 
This is due to more automobile accidents, the 
appalling increase in gunshot and stab 
wounds, and the increasing use of therapeu- 
tic and diagnostic techniques involving the 
cannulation of major arteries. As recently as 
1950, most general surgeons had little expe- 
rience or confidence in techniques of arter- 
ial repair. The experiences in the Korean 
Conflict, combined with the widespread teach- 
ing of techniques of vascular surgery in sur- 
gical residencies, resulted in a great increase 
in frequency of arterial repair between 1950 
and 1960. 

One of the first large series of civilian arte- 
rial trauma was reported by Morris, Creech, 
and DeBakey in 1957. They described a series 
of 136 patients with acute arterial injuries 
treated over 7 years at Baylor University-affil- 
iated hospitals in Houston (Fig. 1-21). Sixty- 




Currently accepted principles regarding 
management of acute vascular injuries is 
largely based on military experience. 
Theodore Drapanas (1970) 



■ FIGURE 1-21 

Anatomic locations of arterial injuries in the 
civilian experience in Houston. In addition to 
the fact that nearly one half of the injuries 
involved the brachial and femoral arteries, 
exactly 50% of the injuries also involved 
arteries supplying the upper extremity. (From 
Morris GC Jr, Creech O Jr, DeBakey ME: Am J 
Surg 1957;93:565-567.) ■ 



chOl.qxd 5/3/04 4:33PM Page 29 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



29 



TABLE 1-8 

THE CIVILIAN EXPERIENCE IN ATLANTA COMPARING TWO CONSECUTIVE 5-YEAR 
PERIODS SHOWS THE INCREASE IN INCIDENCE OF INJURY AND THE MARKED 
IMPROVEMENT OF SUCCESSFUL REPAIR FROM 36 PERCENT TO 90 PERCENT. 
THERE WAS ALSO AN ASSOCIATED REDUCTION IN THE MORTALITY RATE BY ONE- 
THIRD AND AMPUTATION RATE BY ONE HALF 



100 



80 



60 



40 



20 



1950 1951 1952 1953 1954 1955 1956 1957 1958 1959 

■ Percentage of cases treated by repair 

D Percentage of repairs successful (restoration of distal pulses) 

From Ferguson IA, Byrd WM, McAfee DK: Experiences in the management of arterial injuries. Ann Surg 153:980-986, 1961. 




eight injuries, one half of the group, involved 
the upper extremities, and forty-seven injuries 
(about 35%) involved the lower extremities. 
There were injuries of either the abdominal 
or the thoracic aorta and 11 of the carotid 
artery. One hundred twenty of the patients, 
88%, were male, and most of the injuries were 
caused by acts of violence. Primary arterial 
repair was possible in a high percentage of 
these patients. 

In 1961, Ferguson, Byrd, and McAfee, 
reported from Grady Memorial Hospital in 
Atlanta, 200 arterial injuries treated over 10 
years. The superficial femoral artery was 
injured most often, 39 patients, or nearly 20% 
of the total group. However, 54 of the 200 
patients had injuries of minor arteries such 
as the radial or ulnar arteries. The propor- 
tion of patients treated by arterial repair 
increased from less than 10% in 1950 to more 
than 80% in 1959. In the latter part of the 
study, ligation was done only for injuries of 
minor arteries, such as the radial or ulnar, or 
certain visceral arteries. The mortality rate was 
reduced by one third and the amputation rate 



by one half when two consecutive 5-year 
periods were compared. The rate of success 
of arterial repair improved from 36% to 90% 
(Table 1-8). As in Houston, most resulted 
from acts of violence (Table 1-9) . Automo- 
bile, industrial, and domestic accidents 
accounted for most of the remaining injuries. 



TABLE 1-9 

TYPE OF ACUTE ARTERIAL INJURIES IN 
CIVILIAN PRACTICE, HOUSTON, PRIOR 
TO 31 JULY 1956 



No. 



No. 



Transection 
Laceration 
Contusion 
Spasm 

TOTAL 



71 

56 

6 

3 

136 



52.2 

41.2 

4.4 

2.2 

100.0 



Modified from Morris GC Jr, Creech O Jr, DeBakey ME: Acute 
arterial injuries in civilian practice. Am J Surg 93:565-572, 
1957. 



chOl.qxd 4/16/04 3:20PM Page 3 



30 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



In 1963, Smith, Foran, and Caspar 
described experiences with 59 patients with 
61 vascular injuries in Detroit, including both 
acute and chronic arterial injuries (Table 
1-10). They properly emphasized that a 
careful distinction must be made between 
results with acute and chronic lesions. Col- 
lateral circulation has usually developed when 
a chronic lesion is treated and the problem 
of soft tissue is not present. Acts of violence, 
gunshot and stab wounds, caused 18 injuries, 
44% of the penetrating lesions (Table 1-11). 
Their patients included ten industrial injuries 
and eight iatrogenic injuries resulting either 
from surgical operations or from diagnostic 
procedures. These eight included three 
injuries of the external iliac artery during 
inguinal herniorrhaphy, three arterial injuries 
following diagnostic arterial catheterization, 
one injury of the internal iliac artery during 
removal of a herniated intravertebral disc, and 
one arteriovenous fistula developing after a 
mass suture ligature of the renal pedicle 
during nephrectomy. 



In 1964, Patman, Poulos, and Shires 
described experiences with 256 patients, with 
a total of 271 arterial injuries, treated at the 
Parkland Memorial Hospital in Dallas over 1 2 
years starting in July 1949. As in other U.S. 
series, most resulted from acts of violence, 
and only a few resulted from industrial or 
automobile accidents. Multiple arterial 
injuries occurred in 6% of the group. 
Although chronic lesions from trauma were 
included, it was noteworthy that these were 
few: only 6 arteriovenous fistulas and 12 false 
aneurysms among the entire group of 256 
patients. 

A somewhat different group of cases was 
reported from Europe by Vollmar in 1968. In 
an analysis of 85,000 injured patients treated 
in the Heidelberg University Surgery Clinic 
between 1953 and 1966, there were only 172 
arterial lesions, an incidence of 0.3%. In 
marked contrast to the U.S. experience, only 
1% of the injuries were due to gunshot 
wounds. Most patients were injured in indus- 
trial accidents (Table 1-12). Approximately 



TABLE 1-10 

CIVILIAN ARTERIAL TRAUMA IN DETROIT, 


61 ARTERIAL INJURIES 




Type of Trauma 


Laceration 


28 


Early 


Lesions 


Spasm 






Transection 




Thrombosis 




Penetrating injuries 
Nonpenetrating injuries 

TOTAL 


10 
_2 
12 


6 
2 
8 




2 
4 
6 


2 
2 



Modified from Smith RF, Szilagyi DE, Pfeifer JR: Arterial trauma. Arch Surg 86:825-835, 1963. 



TABLE 1-11 

ETIOLOGIC FACTORS CAUSING 61 ARTERIAL INJURIES, 59 PATIENTS IN CIVILIAN 
SERIES IN DETROIT 



42 Penetrating Injuries 

Gunshot 13 

Industrial 10 

Iatrogenic 8 

Household 6 

Stab 5 



% 

21.3 

16.4 

13.1 

9.8 

8.2 



19 Nonpenetrating Injuries 

Industrial 1 1 

Auto 4 

Athletic 2 

Household 2 



18.0 
6.6 
3.3 

3.3 



Modified from Smith RF, Szilagyi DE, Pfeifer JR: Arterial trauma. Arch Surg 86:825-835, 1963. 



chOl.qxd 4/16/04 3:20PM Page 31 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



31 



TABLE 1-12 

TYPE OF ACCIDENT RESPONSIBLE 
FOR 169 PATIENTS WITH ARTERIAL 
INJURIES, HEIDELBERG UNIVERSITY 
SURGICAL CLINIC 1953-1966 



Etiology 




No. 


% 


Industrial ace 


dent 


59 


35 


Domestic accident 


44 


26 


Suicide 




32 


19 


Traffic 




24 


14 


Iatrogenic 




10 


6 


TOTAL 




169 


100 



Modified from Vollmar J: In Hiertonn T, Rybeck B (eds): 
Traumatic arterial lesions. Stockholm: Forsvarets 
Forskningsanstalt, 1968. 



TABLE 1-13 

NATURE OF 197 ARTERIAL LESIONS IN 
168 PATIENTS, HEIDELBERG 
UNIVERSITY, SURGICAL CLINIC 



Type 



No. 



% 



Sharp penetrating 



Blunt 



Cut 


95 


48 


Stab 


19 


10 


Shot 


3 


1 


Closed 


22 


11 


Open 


58 


30 



Modified from Vollmar J: In Hiertonn T, Rybeck B (eds): 
Traumatic arterial lesions. Stockholm: Forsvarets 
Forskningsanstalt. 1968. 



59 



41 



one fourth resulted from simple domestic acci- 
dents. Twelve resulted from automobile acci- 
dents. It was significant in this group that 41 % 
of the total resulted from blunt trauma (Table 
1-13). 



In 1968, Saletta and Freeark described expe- 
riences with 57 patients with partially severed 
major peripheral arteries treated in Chicago 
(Table 1-14). Most of the injuries resulted 
from physical violence from gunshot wounds 



TABLE 1-14 

LOCATION AND CAUSE OF INJURY IN 57 PATIENTS WITH PARTIALLY 
SEVERED ARTERIES 



Location and Artery 



No. 



Gunshot 



Knife 



Etiology 

Glass Blunt 



Other 



Head and neck 








Temporal 


3 






Internal carotid 


2 


2 




External carotid 


2 


1 


1 


Vertebral 


1 




1 


Lingual 


1 




1 


Upper extremity 








Axillary 


6 


2 


3 


Brachial 


5 


1 


4 


Innominate 


1 


1 




Subclavian 


1 




1 


Lower extremity 








Femoral 


15 


11 


3 


Common femoral 


6 


3 


1 


Popliteal 


6 


6 




External iliac 


4 


3 


1 


Deep femoral 


2 


1 


1 


Anterior tibial 


1 






Posterior tibial 


1 






Tibioperoneal 


1 


1 





1 



1 (needle) 
1 (needle) 



1 (tin can) 



Modified from Saletta JD, Freeark RJ: The partially severed artery. Arch Surg 97:198-205, 1968. 



chOl.qxd 4/16/04 3:20PM Page 32 



32 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



or knives. Also, in 1968 Dillard, Nelson, and 
Norman described the treatment of 85 arte- 
rial injuries in St. Louis over 8 years begin- 
ning in 1958 (Table 1-15). Eighty-one percent 
of the injuries involved an extremity. Pene- 
trating injuries from knives or glass caused 35 
of the injuries, 31 resulted from gunshot 
wounds, and 19 were caused by blunt trauma 
or crushing injuries (Table 1-16). 

Two large series are those of Drapanas and 
colleagues (1970) from New Orleans, which 
included 226 arterial injuries, and the cumu- 
lative report by Perry, Thai, and Shires from 
Dallas (1971), which included 508 arterial 
injuries. At Charity Hospital in New Orleans, 
226 patients with arterial injuries were treated 
between 1942 and 1969 (Drapanas and col- 
leagues, 1970) (Fig. 1-22). Of 226 patients, 
173 had major arterial injuries and 53 had 
minor injuries. The most frequently injured 
arteries were the brachial (39 injuries) and 
the superficial femoral (31 injuries). There 
was an unusually large number (23) of aortic 
injuries involving the thoracic or abdominal 
aorta. 

In 1971, Perry Thai, and Shires reported 
additional series of 259 arterial injuries from 
Dallas. About 55% of these were associated 
with gunshot wounds (Table 1-17). Combined 
with the 1964 report (Patman, Poulos, and 
Shires) , there were a total of 508 injuries 
(Table 1-18). These included 442 injuries of 



TABLE 1-15 

DISTRIBUTION OF ARTERIAL INJURIES, 
ST. LOUIS, MISSOURI, 1958-1966 



Artery 



% 



Axillary artery 


6 


7.1 


Brachial artery 


26 


30.6 


Subclavian artery 


4 


4.7 


Thoracic aorta 


8 


9.4 


Abdominal aorta and branches 


8 


9.4 


Iliac artery 


2 


2.3 


Common femoral artery 


7 


8.2 


Superficial femoral artery 


14 


16.5 


Popliteal artery 


10 


11.8 


TOTAL 


85 


100.0 



TABLE 1-16 

ETIOLOGY AND ANATOMIC 
DISTRIBUTION OF 85 ARTERIAL 
INJURIES, ST. LOUIS, MISSOURI, 1958- 
1966 

A. Knife or glass penetrating injuries 





Upper extremities 


15 




Thoracic aorta 


3 




Abdominal aorta and branches 


5 




Lower extremities 


12 




TOTAL 


35(41.2%) 


B. 


Penetrating gunshot injuries 






Upper extremities 


13 




Thoracic aorta 


2 




Abdominal aorta and branches 


2 




Lower extremities 


14 




TOTAL 


31 (36.5%) 


C. 


Blunt trauma or crush injuries 






Upper extremities 


8 




Thoracic aorta 


3 




Abdominal aorta and branches 


3 




Lower extremities 


5 




TOTAL 


19(22.3%) 



Modified from Dillard BM, Nelson DL, Norman HG Jr: Review 
of 85 major traumatic arterial injuries. Surgery 63:391-395, 
1968. 



Modified from Dillard BM, Nelson DL, Norman HG Jr: Review 
of 85 major traumatic arterial injuries. Surgery 63:391-395, 
1968. 



arteries in the extremities, representing 87% 
of the total. There were also 42 cervical arte- 
rial injuries and 24 visceral arterial injuries. 
Moore and colleagues in 1971 reported 250 
vascular injuries treated in Galveston, 45% of 
which occurred in the extremities (Table 
1-19) . In this series, 40% of the cases involved 
either the chest, the abdomen, or the head 
and neck, a percentage higher than that in 
other reports. The injuries resulted from 
either gunshot or stab wounds in 60% of the 
group (Table 1-20) . Thirteen percent resulted 
from blunt trauma, and iatrogenic injuries 
were responsible for ten percent. Among the 
25 iatrogenic injuries, there were 16 acute arte- 
rial thromboses resulting from a total of more 
than 3000 cardiovascular radiographic 
procedures, a frequency of less than 1% 
(Table 1-21). 

Smith, Elliot, and Hageman (1974) 
reported a survey of 268 patients in Detroit 
with 285 penetrating wounds of the limbs and 
neck. There were 127 peripheral arterial 
injuries identified. Kelly and Eiseman (1975) 



chOl.qxd 4/16/04 3:20PM Page 33 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



33 



Sup. mes.~3 




S. femoral-31 



■ FIGURE 1-22 

Distribution of 226 acute civilian arterial injuries 
covering a 30-year period in New Orleans 
starting in 1942. Eighty percent of the injuries 
involve arteries to the extremities. (From 
Drapanas T, Hewitt RL, Weichert RF 3rd, Smith 
AD: Surg 1970;172:351-360.) ■ 



reported 43% of 143 patients in Denver with 
vascular injuries sustained gunshot wounds. 
The brachial artery was injured most often, 
37 times. Hardy and colleagues (1975) in 
Jackson recorded 192 arterial injuries from 
firearms (155 gunshot and 37 shotgun), 91 
stab wounds and lacerations, 48 injuries from 
blunt trauma, and 20 iatrogenic injuries. The 
series included 36 aortic injuries. However, 
approximately two thirds involved extremity 
vessels. Cheek and colleagues (1975) reviewed 
200 operative cases of major vascular injuries 
in Memphis, which included 155 arterial 
injuries. Bole and colleagues (1976) reported 
126 arterial injuries in 122 patients in New 
York City from 1968 to 1973. 

Reynolds, McDowell, and Diethelm (1979) 
documented results in managing 191 con- 
secutive patients treated for arterial injuries 
during an 8-year period at the University of 
Alabama Medical Center starting in 1970. Most 
of their patients sustained penetrating 
wounds, either gunshot or shotgun; however, 
there were also 46 patients, or 24%, who had 
blunt trauma associated with their arterial 
injuries. Barros D'Sa and colleagues (1980) 
from the Queen's University Hospital at 
Belfast reviewed their experience with missile- 
induced vascular trauma over 7/ 2 years of 
serious hostilities involving the civilian pop- 
ulation of northern Ireland. They docu- 
mented the results in managing 113 patients 
with 191 vascular injuries treated at the Royal 
Victoria Hospital. It is particularly important 
to note that treatment commenced within 1 
hour in 87% of the patients. Etiology of the 



TABLE 1-17 

CAUSE AND TYPE OF INJURY, CIVILIAN ARTERIAL TRAUMA, DALLAS, TEXAS 



Cause 


No. 




Arterial Injury 






Gunshot wound 


143 


55.2 


Laceration 


133 


51.4 


Edged instruments 


93 


35.5 


Transection 


99 


38.2 


Blunt trauma 


24 


9.3 


Puncture 


18 


6.9 


TOTAL 


259 


100.0 


Contusion 


7 


2.7 








Spasm 


2 


0.8 








TOTAL 


259 


100.0 



Modified from Perry MO, Thai ER, Shires GT: Management of arterial injuries. Ann Surg 173:403-408, 1971. 



chOl.qxd 4/16/04 3:20PM Page 34 



34 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



TABLE 1-18 

DISTRIBUTION OF CIVILIAN ARTERIAL INJURIES, DALLAS, TEXAS, 508 ARTERIAL 
INJURIES 



Extremity (89.0%) 




Cervical (8.3%) 




Visceral (4.7%) 




Aorta 


26 


Common carotid 


24 


Celiac 


2 


Innominate 


1 


Internal carotid 


8 


Splenic 


2 


Subclavian 


23 


External carotid 


6 


Superior mesenteric 


7 


Axillary 


38 


Vertebral 


4 


Renal 


9 


Brachial 


78 


TOTAL 


42 


Hepatic 


4 


Radial 


58 






TOTAL 


24 


Ulnar 


39 










Common iliac 


20 










External iliac 


11 










Hypogastric 


7 










Common femoral 


11 










Superficial femoral 


93 










Profunda femoral 


8 










Popliteal 


17 










Tibial 


12 










TOTAL 


442 











Modified from Perry MO, Thai ER, Shires GT: Management of arterial injuries. Ann Surg 173:403-408, 1971. 



TABLE 1-19 

LOCATION OF VASCULAR TRAUMA IN 
250 CIVILIAN INJURIES, GALVESTON, 
TEXAS, 1960-1970 



TABLE 1-20 

LOCATION OF VASCULAR TRAUMA IN 
250 CIVILIAN INJURIES, GALVESTON, 
TEXAS, 1960-1970 



Location 

Head and neck 
Thoracic outlet 
Chest 
Abdomen 
Extremities 

TOTAL 



Type 



10 


Gunshot 


16 


Stab 


15 


Blunt 


14 


latragenic 


45 


Other 


100 


TOTAL 



Modified from Moore CH, Wolma FJ, Brown RW, Derrick JR: 
Vascular trauma. A review of 250 cases. Am J Surg 122:576- 
578, 1971. 



% 

39.0 
25.0 

13.0 

10.0 

13.0 

100.0 



Modified from Moore CH, Wolma FJ, Brown RW, Derrick JR: 
Vascular trauma. A review of 250 cases. Am J Surg 122:576- 
578, 1971. 



chOl.qxd 4/16/04 3:20PM Page 35 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



35 



TABLE 1-21 

25 CASES OF IATROGENIC VASCULAR INJURIES, GALVESTON, TEXAS, 1960-1970 



Procedure 



Injury 



Treatment 



No. 



Central venous catheterization 


Thrombosis 


Thrombectomy 


16 


Lumbar laminectomy 


Arteriovenous fistula 


Repair of fistula 


3 


Osteotomy of hip 


Arteriovenous fistula 


Repair of fistula 


1 


Renal hemodialysis 


False aneurysm 


Resection of aneurysm 


2 


arteriovenous shunts 








Pelvic irradiation 


Femoral artery rupture 


Aortofemoral bypass 


1 


Fracture of humerus 


Volkmann's ischemia 


Open reduction; free 


1 


closed reduction 




artery and fasciotomy 




Subclavian catheterization 


Arteriovenous fistula 


Repair of fistula 


1 


TOTAL 






25 



Modified from Moore CH, Wolma FJ, Brown RW, Derrick JR: Vascular trauma. A review of 250 cases. Am J Surg 122:576-578, 
1971. 



injuries is outlined in Table 1-22. A special 
group of 38 patients had "knee capping" con- 
tributed to most of the popliteal vascular 
injuries. 

Koivunen (1982) documented the experi- 
ence in managing vascular trauma in a rural 
population in Missouri. During a 10-year 
period, they identified 89 cases of vascular 
trauma. Recognizing that the considerable 
delay in abdominal vasculature accounted for 
33.7% of the injuries. Multiple injuries were 
common, with 1057 patients having two or 
more concurrent vascular injuries. There 
were three patients who had four or more sep- 
arate vascular injuries. The increasing inci- 



TABLE 1-22 

INCIDENCE OF VASCULAR INJURIES 
RELATED TO TYPE 



Wounding Missile 




Patien 






No. 




% 


Bullet 








Low velocity 


48 




42.5 


High velocity 


28 




24.8 


Uncertain velocity 


22 




19.5 


= ragments from explosions 


15 




13.2 


TOTAL 


113 







From Barros D, Sa AA, Hassard TH, Livingston RH, et al: 
Missile-induced vascular trauma. Injury 12:13-30, 1980. 



dence of vascular trauma in urban centers is 
emphasized by the marked increase from an 
average of 27 patients per year in the early 
1960s to nearly a tenfold increase to the 
current average of 213 patients peryear. These 
data can be compared and contrasted to other 
civilian and military experience, as noted in 
Table 1-23. 

Feliciano and colleagues (1984) reported 
a 1-year experience with 456 vascular and 
cardiac injuries among 312 patients during 
1982 at the Ben Taub Hospital in Houston. 
More than 87% of the injuries were pene- 
trating, as identified in Table 1-24. Specifi- 
cally, there were 408 vascular injuries and 48 
cardiac injuries. Thirty-four percent of the 
patients had two or more vascular or cardiac 
injuries. The majority were penetrating, with 
more than 87% secondary to gunshotwounds, 
stab wounds, or shotgun wounds. The largest 
number of injuries occurred in the extremi- 
ties, 39.9%, with the brachial artery being the 
most common arterial injury. There was a rel- 
atively large number of abdominal vascular 
injuries, accountingfor 31.9% of the total. The 
most common venous injury occurred in the 
internal jugular vein in 26 patients. 

Mattox and colleagues (1989) detailed a 
unique epidemiologic evolutionary profile 
from the civilian trauma registry at Baylor 
College of Medicine in Houston. During a 30- 
year period from 1958 to 1987, where con- 
sistent evaluation and treatment philosophy 



chOl.qxd 4/16/04 3:20PM Page 36 



36 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



TABLE 1-23 

LOCATION OF REPORTED VASCULAR INJURIES IN MAJOR WARS 



Neck 



Chest Abdomen Upper Extremity Lower Extremity Total 



Makins (WWI 4 ) 


176 


— 


11 


367 


648 


1202 


DeBakey(WWII 2 ) 


34 


— 


49 


871 


1517 


2471 


Hughes (Korea 3 ) 


14 


— 


7 


109 


304 


304 


Rich (Vietnam 7 ) 


76 


4 


354 


416 


840 


1377 


TOTAL 


300 


4 


421 


1763 


3179 


5667 



LOCATION OF CARDIOVASCULAR INJURIES IN CIVILIAN EPIDEMIOLOGIC 
VASCULAR TRAUMA REPORTS 



Author 


City 


Year 


Neck 


Chest 


Abdomen 


Upper 
Extremities 


Lower 
Extremities 


Total 


Morris 19 


Houston 


1957 


16 


5 


13 


62 


39 


136 


Ferguson 13 


Atlanta 


1961 


15 


1 


32 


93 


56 


200 


Smith 23 


Detroit 


1962 


3 


2 


8 


25 


21 


57 


Treiman 24 


Los Angeles 


1966 


14 


10 


56 


67 


86 


233 


Dilard 10 


St. Louis 


1968 


4 


8 


10 


32 


31 


85 


Drapanas 11 


New Orleans 


1970 


28 


11 


31 


97 


59 


226 


Perry 21 


Dallas 


1971 


65 


14 


75 


213 


141 


508 


Moore 16 


Galveston 


1971 


45 


57 


35 


56 


37 


250 


Cheek 9 


Memphis 


1975 


46 


10 


88 


30 


60 


200 


Kelly 15 


Denver 


1975 


14 


— 


62 


52 


47 


175 


Hardy 16 


Jackson 


1975 


39 


41 


66 


98 


116 


360 


Bole 8 


New York 


1976 


8 


12 


31 


25 


50 


126 


Sirinek 22 


San Antonio 


1983 


17 


35 


218 


— 


— 


270 


TOTAL 






315 


206 


725 


850 


763 


2859 



From Mattox KL, Feliciano DV, Burch J, et al: Five thousand seven hundred sixty cardiovascular injuries in 4459 patients. 
Epidemiologic evolution, 1958 to 1987. Ann Surg 209:698-707, 1989. 



TABLE 1-24 

MECHANISMS OF INJURY; ALL 
VASCULAR AND CARDIAC INJURIES 



Mechanism 

Gunshot wound 
Stab wound 
Shotgun wound 
Laceration 
Iatrogenic 
Blunt 

TOTAL 



No. 

166(53.2%) 
88 (28.2%) 
18(5.8%) 
17 (5.4%) 
14(4.5%) 
9 (2.9%) 

312 (100%) 



87.2% 



From Feliciano DV, Bitando CG, Mattox KL, et al: Civilian 
trauma in the 1980s. A 1-year experience with 456 vascular 
and cardiac injuries. Ann Surg 199:717-724, 1984. 



existed, they treated 5760 cardiovascular 
injuries in 4459 patients. Eighty-six percent 
of the patients were male, with an average age 
of 30years. Penetrating trauma accounted for 
more than 90.0% of the injuries, with 51.1% 
resulting from gunshot wounds, 31.1% from 
stab wounds, and 6.8% from shotgun wounds 
(Table 1-25). The remaining injuries were 
iatrogenic or secondary to blunt trauma. Table 
1-26 outlines the specific cardiovascular 
injuries by etiology and grouped by body 
region. Of particular interest and note, and 
in marked contrast with military experience 
and many previous civilian series, truncal 
injuries, including the neck, accounted for 
66% of all injuries treated. The lower extrem- 
ities, including the groin, accounted for only 
19% of the injuries, specifically, injuries to the 
six patients. In the decade of the 1980s, there 
was marked increase in the number of 



chOl.qxd 4/16/04 3:20PM Page 37 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



37 



TABLE 1-25 

ETIOLOGY OF PATIENT CARDIOVASCULAR INJURIES PER 5-YEAR TIME INTERVAL 



Etiology 



1958-1963 1964-1968 1969-1973 1974-1978 1979-1983 1984-1988 Total 



Gunshot wound 


42 


236 


436 


501 


625 


456 


2296 


Stab/laoeration 


64 


110 


161 


229 


362 


463 


1389 


Blunt trauma 


1 


17 


58 


90 


62 


76 


304 


Shotgun wound 


1 


15 


45 


55 


61 


37 


214 


Iatrogenic 


1 


1 








4 


25 


31 


Other/unknowns 


54 


20 


111 


25 


3 


12 


225 


TOTAL 


163 


399 


811 


900 


1117 


1069 


4459 



From Mattox KL, Feliciano DV, Burch J, et al: Five thousand seven hundred sixty cardiovascular injuries in 4459 patients: 
Epidemiologic evolution 1958 to 1987. Ann Surg 209:698-707, 1989. 



manuscripts devoted to the management of 
civilian vascular trauma culminated by the 
extensive review of Mattox and colleagues. 

During the 1990s, there continued to be a 
significant number of reports of the man- 
agement of civilian arterial injuries. Oiler and 
colleagues (1992) established a State Trauma 
Registry that had an early report of 1148 vas- 
cular injuries suffered by 9 78 patients in North 
Carolina over 39 months (October 1987 to 
January 1991) (Table 1-27). Whether human 
made or natural, there will continue to be 
trauma patients with associated vascular 
injuries. Internationally Kurtoglu and col- 
leagues (1991) described treating 115 periph- 
eral arterial injuries in Istanbul (Table 1-28) . 
There will be additional details throughout 
the text emphasizing the recent experience 
of the past decade. The following, however, 
identifies areas of historical significance 
related to specific considerations involving 
vascular injuries. 



HISTORICAL NOTES ON 20th 
CENTURY PROGRESS WITH 
VENOUS INJURIES 



It could be recorded in history that 
outstanding contributions based on 
experience of managing Vietnam casualties 
by American military surgeons did as much 
to stimulate and direct interest and success 



in repair of venous injuries as was 
established during the Korean Conflict with 
repair of arterial injuries. Vietnam Vascular 
Registry, Rich (1977) 

Several excellent historic reviews of the 
development of venous trauma have been pub- 
lished (Haimovici, 1963; Shumacker, 1969; 
Rob, 1972). Two earlier outstanding refer- 
ences are Guthrie (1912) andMurphy (1897). 
As early as 1816, Travers supposedly closed a 
small wound in a femoral vein. In 1830, 
Guthrie reported more precisely that he 
closed a laceration of the internaljugular vein 
by placing a tenaculum through the cut 
edges, after which he tied a suture around the 
tenaculum to constitute a lateral ligature. In 
1878, Agnew used lateral sutures to close 
venous wounds. Only a year earlier, Eck had 
performed the first vascular anastomosis by 
suturing the portal vein to the inferior 
vena cava. Schede in 1882 in Germany is 
generally given credit for performing the 
first successful lateral suture repair of a lac- 
eration in a vein in clinical practice, and he 
advocated repair ofwounds of the femoral vein 
in man. 

In the late 19th century, other surgeons who 
with apparent success sutured wounds of veins 
include Billroth, Braun of Koenigsberg, and 
Schmidt. In his experimental laboratory, 
Hirsch in 1881 successfully repaired divided 
veins in dogs. When Dorfler in 1889 outlined 
his method of arterial repair, he recom- 
mended the same technique for repairing 



chOl.qxd 4/16/04 3:20PM Page 38 



38 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



TABLE 1-26 

SPECIFIC CARDIOVASCULAR INJURIES BY ETIOLOGY AND GROUPED BY 
BODY REGION 



Gunshot Stab wound/ 
wound laceration 



Blunt Shotgun Unknown/ 

trauma wound lateogenic Other 



Total 



Carotid artery 


115 


45 


6 


14 


— 


10 


190 


Jugular vein 


116 


154 


4 


9 


— 


13 


296 


Vertebral artery 


18 


13 


3 


3 


— 


2 


40 


Subclavian vessel 


91 


50 


8 


6 


— 


13 


168 


Heart 


220 


261 


32 


3 


5 


8 


539 


Coronary artery 


3 


10 


— 


— 


3 


1 


14 


Ascending aorta 


15 


12 


3 


3 


— 


— 


33 


Innominate artery 


20 


8 


7 


2 


2 


— 


39 


Pulmonary artery 


43 


25 


7 


3 


— 


1 


79 


Desc thorac aorta 


25 


5 


59 


— 


— 


— 


89 


Aortic arch 


13 


7 


1 


1 


— 


— 


22 


Thorac vena cava 


34 


15 


4 


1 


— 


1 


55 


Innominate vein 


25 


15 


2 


— 


— 


— 


42 


Pulmonary vein 


29 


5 


4 


1 


— 


1 


40 


Azygous vein 


13 


2 


— 


1 


— 


— 


16 


Thoracic duct 


3 


8 


— 


1 


— 


— 


12 


Int mammary artery 


18 


71 


3 


— 


— 


6 


98 


Intercostal artery 


25 


54 


— 


— 


— 


2 


81 


Abdominal aorta 


180 


40 


5 


17 


2 


5 


249 


Inf vena cava 


353 


100 


44 


21 


— 


17 


535 


Mesentric artrey 


136 


45 


14 


7 


— 


14 


216 


Portal venous 


116 


44 


22 


3 


— 


4 


189 


Iliac artery 


172 


30 


11 


11 


2 


6 


232 


Iliac vein 


224 


32 


9 


11 


1 


12 


289 


Renal vessel 


86 


33 


32 


4 


— 


8 


163 


Epigastric artery 


3 


14 


— 


3 


— 


1 


52 


Hepatic veins 


36 


6 


8 


1 


— 


1 


21 


Axillary vessel 


85 


40 


3 


6 


1 


8 


143 


Brachial artery 


184 


163 


14 


38 


10 


37 


446 


Radial/ulnar art 


38 


169 


1 


10 


2 


41 


261 


Cephalic/basilic vein 


4 


3 


— 


1 


1 


— 





Femoral artery 


316 


58 


14 


70 


5 


37 


500 


Femoral vein 


184 


34 


7 


36 


— 


19 


280 


Popliteal artery 


88 


3 


36 


18 


— 


11 


156 


Popliteal vein 


45 


5 


9 


14 


— 


9 


68 


Tibial artery 


31 


8 


11 


9 


— 


9 


68 


Tibial vein 


4 


1 


— 


1 


— 


1 


7 


Saphenous vein 


12 


— 


1 


1 


— 


1 


15 


TOTAL 


3134 


1543 


385 


341 


56 


293 


5760 



From Mattox KL, Feliciano DV, Burch J, et al: Five thousand seven hundred sixty cardiovascular injuries in 4459 patients: 
Epidemiologic evolution 1958 to 1987. Ann Surg 209(6):698-707, 1989. 



veins. Haimovici (1963) described Dorfler's 
method, "The essential features of this method 
consisted of the use of fine, round needles 
and fine silk and his suture was continuous, 
embracing all of the coats of the vessel. From 



his experience, although limited to 16 cases, 
he concluded that aseptic silk thread in the 
lumen of the vessel does not necessarily lead 
to thrombosis and, therefore, the penetration 
of the intima was not contraindicated." 



chOl.qxd 4/16/04 3:20PM Page 3 9 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



39 



TABLE 1-27 

VASCULAR INJURIES IN A RURAL 
STATE: A REVIEW OF 978 PATIENTS 
FROM A STATE TRAUMA REGISTRY 



Vessels Injured by Region 

Head carotid and neck 

Common 

Internal 

External 

Unspecified 

Jugular internal 

Other vessels of neck 
Thorax 

Aorta 

Innominate/subclavian A & V 

Pulmonary vessels 

Intercostals, mammary, superior 
vena cava, other, unspecified 
Abdomen/pelvis 

Aorta 

Interior vena cava 

Celiac and mesenteric artery 

Porta and splenic vessel 

Renal vessel 

Iliac vessels 

Ovarian, other, unspecified 
Upper extremity 

Axillary vessels 

Brachial vessels 

Radial artery 

Ulnar artery 

Digital 

Other, unspecified 
Lower extremity 

Common femoral artery 

Superficial femoral artery 

Femoral vessel 

Popliteal artery 

Popliteal vein 

Popliteal vessels 

Tibital vessels 

Plantar, other, unspecified 



No. (%) 

133(9.9) 

12 

17 
9 

10 

22 

63 
166(12.4) 

58 

37 

14 

57 

211 (15.7) 

16 

38 

40 

19 

23 

45 

30 
361 (26.9) 

17 

93 

79 

81 

69 

22 
271 (20.2) 

16 

54 

25 

38 

12 

14 

63 

49 



From Oiler DW, Rutledge R, Clancy T, et al: Vascular injuries 
in a rural state: a review of 978 patients from a state trauma 
registry. J Trauma 32:740-746, 1992. 



In 1889, Kummel performed the first clin- 
ical end-to-end anastomosis of a femoral vein. 
In 1901, Clermont successfully reunited the 
ends of a divided vena cava with a continuous 
fine silk suture. A month later, the lumen of 
the vena cava was found to be smooth and 
unobstructed at the site of the anastomosis. 
Jensen in 1903 was successful in four of seven 



TABLE 1-28 

ETIOLOGIC FACTORS MANAGEMENT 
OF VASCULAR INJURIES OF THE 
EXTREMITIES (115 CASES) 



Etiologic Factors Number of Cases 



Penetrating trauma 
Stab wounds 
Traffic accidents 
Gunshot injury 
Industrial accidents 
Failing from heights 
latragenic 



50 
25 
22 
9 
5 
2 
2 



% 

43 
21 
19 
8 
5 
2 
2 



From Kurtoglu M, Ertekin C, Bulut T, et al: Management of 
vascular injuries of the extremities: 1 15 cases, Int Angiol 
10:95-99, 1991. 



operations in anastomosing transected veins, 
using a continuous suture technique. 

In World War I the clinical use of lateral 
suture repair of venous lacerations was 
reported by Goodman (1918). He reported 
experiences with five patients with vascular 
injuries in whom a lateral suture repair of 
venous lacerations was done in four, involv- 
ing two popliteal and two superficial femoral 
veins. The defects ranged from 5 to 20 mm in 
length. The results are unknown because there 
was no follow-up evaluation. 

The importance of venous repair was min- 
imized by the proposal of Makins in 1917, 
which was that the concomitant vein should 
be ligated when an arterial injury was treated 
by ligation. The results reported by Makins to 
support this hypothesis were later found to 
have no statistical significance (Table 1-29) . 
The influence persisted even until World 
War II. Data from World War II showed no 
benefit from ligation of the concomitant 
vein, however (DeBakey and Simeone, 
1946). During the Korean Conflict, repair 
of injuries of major veins was again under- 
taken in selected patients (Hughes, 1959). 
This was expanded in Vietnam (Rich, 1970 to 
1995). 

One of the most bizarre recommendations 
in the history of vascular surgery is the mid- 
19th century recommendation that ligation 
of the concomitant uninjured artery should 
be done when a venous injury was treated by 



chOl.qxd 4/16/04 3:20PM Page 40 



40 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



TABLE 1-29 

A COMPARISON OF THE RESULTS OF LIGATIONS OF THE ARTERY ALONE WITH 
THOSE OF SIMULTANEOUS LIGATIONS OF ARTERY AND VEIN 



Artery 




Artery 


Alone 






Artery 


and Vein 






No. of 


Good 






Percent 


No. of 


Good 




Percent 




Cases 


Result 


G 


angrene 


Gangrene* 


Cases 


Result 


Gangrene 


Gangrene* 


Subclavian 


4 


3 




1 


25.0 


1 


1 


— 


0.0 


Axillary 


6 


5 




1 


16.6 


4 


4 


— 


0.0 


Brachial 


13 


10 




3 


23.0 


1 


1 


— 


0.0 


= emoral 


32 


24 




8 


25.0 


32 


25 


7 


21.0 


Popliteal 


24 


14 




10 


41.6 


28 


22 


6 


21.4 


Tibial 


4 


4 




— 


0.0 


1 


1 


— 


0.0 


Carotid 


18 


12 




6 


3.3 


4 


3 


1 


25.0 


TOTAL 


101 


72 




29 


28.0 


71 


57 


14 


19.7 



*AII the percentages were added to the table by the author, except the total percentages, which appear in Makins' original table 
Modified from Montgomery ML: Arch Surg 1932;24:1016-1027. 



ligation (Rich and Rob, 1996) . Apparently this 
astonishing recommendation was first made 
by Gensoul in 1883, who feared the hazards 
of venous engorgement if the vein alone was 
ligated. Other surgeons (Dupuytren, 1839; 
Chassaignac, 1855; Langenbeck, 1861; Pilch er, 
1886) made similar recommendations, 
although these were intended primarily to 
minimize hemorrhage with venous injuries 
(Simeone, Grillo, and Rundle, 1951). 

Moreover, during the Korean Conflict, 
there was a renewal of interest in repair of the 
involved vein during the elective repair of 
arteriovenous fistulas that usually was per- 
formed several months after the initial injury. 
Traditionally such fistulas were treated by lig- 
ation of both the artery and the vein. The tech- 
nique of repair gradually evolved to include 
repair of the artery and often repair of the 
concomitant vein. Successful results in such 
patients generated some enthusiasm for repair 
of acute venous injuries (Hughes, 1958): 



. . noted 63 percent major vein injuries 
accompanying major artery injuries. A 
number of other vein injuries were treated in 
which there was no arterial involvement. 
Most of these veins were treated by ligation, 
but in some, ligations resulted in various 



degrees of venous stasis. On rare occasion, 
massive venous stasis resulted in 
amputation of the extremity. To eliminate this 
complication two investigators . . . [sic] 
Hughes and Spencer independently . . . 
began the repair of major veins. They 
reported 20 major veins repaired, all by 
lateral suture except one which was 
repaired by direct anastomosis. Some of 
these are known to have thrombosed later 
without complications. No embolic 
complications resulted. 



A review concerned primarily with the 
management of venous injuries in civilian 
practice was published by Caspar and Treiman 
in 1960; it described injuries of 52 major veins 
in 51 patients. Venous reconstruction when 
performed was usually by lateral suture. 

During the Vietnam Conflict, with the 
interest and experience resulting from the 
necessity of treating thousands of vascular 
injuries, there was a significant effort to 
perform venous repairs in the last 5 years of 
the conflict (from 1968 to 1972). In a sym- 
posium on venous surgery in the lower extrem- 
ities atWalter Reed Army Institute of Research, 
the combined experiences of both civilian and 
military surgeons were summarized (Swan and 



chOl.qxd 4/16/04 3:20PM Page 41 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



41 



colleagues, 1975) . Venous Trauma, by Hobson, 
Rich, and Wright (1983) , reviews civilian and 
military experience with venous injuries. 

Venous injuries are unimportant to many 
surgeons, so the true frequency is not docu- 
mented accurately. This is particularly true in 
the case of combined arterial and venous 
injuries. Analysis of the Vietnam experience 
found numerous cases in which venous trauma 
was not documented in the records. The first 
major interest in the frequency of venous 
injuries in military trauma was during the 
Korean Conflict. In analysis of 180 acute vas- 
cular injuries (Table 1-30), Hughes (1954) 
found nearly as many injuries in major veins 
(71) as there were in major arteries (79). Sim- 
ilarly, in civilian practice, the frequency of 
venous trauma was documented only occa- 



TABLE 1-30 

INCIDENCE OF ACUTE VASCULAR 
TRAUMA IN KOREAN CASUALTIES 



Vessel 


No. 


% 


Major 


arteries 


79 


43.9 


Major 


veins 


71 


39.4 


Minor 


arteries 


30 


16.7 


TOTAL 




180 


100.0 



Modified from Hughes CW: Acute vascular trauma in Korean 
casualties: analysis of 180 cases. Surg Gynecol Obstet 
99:91-100, 1954. 



sionally, most reports describing only arterial 
trauma until 1980. There are numerous large 
series of arterial injuries reported thatgive no 
details regarding venous trauma and this con- 
tinues through 1996. The report by Caspar 
and Treiman (1960) is one of the first to have 
a detailed analysis of venous injuries alone. 
In a group of 228 patients with vascular injuries 
at the Los Angeles County General Hospital 
over a period of 10 years, about 22% (51 
patients) had venous injuries. The superficial 
femoral vein was most commonly injured 
(nine times) . The inferior vena cava and the 
internal jugular vein were each injured eight 
times, and the brachial veins seven. In 1966 
40 patients were added to the original series 
in a supplementary report by Treiman. The 
frequency of venous injury in the different 
locations is shown in Table 1-31. Mullins, 
Lucas, and Ledgerwood (1980) published a 
large series of civilian venous injuries from 
Detroit. 

In the preliminary Vietnam Vascular Reg- 
istry report, approximately one fourth of the 
patients had venous trauma (Table 1-32) 
(Rich and Hughes, 1969). There were only 
28 injuries of isolated veins, and most of the 
venous injuries were combined with arterial 
trauma. The increased incidence of venous 
trauma when associated with arterial trauma 
was emphasized in an interim Registry report, 
which documented concomitant venous 
injuries in 37.7% of cases with acute major 
arterial trauma (Table 1-33) (Rich, 1970). 



TABLE 1-31 










INCIDENCE OF VENOUS INJURIES 










No. 


No. 


Total 




Vein 


1948-1958 


1958-1963 


1948-1963 




Axillary brachial 


8 


5 


13 


14.1 


Innominate subclavian 


3 


5 


8 


8.7 


Superior vena cava 


1 





1 


1.1 


Inferior vena cava 


8 


4 


12 


13.0 


Iliac 


7 


4 


11 


12.0 


Femoral 


11 


6 


17 


18.5 


Other 


14 


16 


30 


32.6 


TOTAL 


52 


40 


92 


100.0 



Modified from Treiman RL, Doty D, Gaspar MR: Acute vascular trauma a fifteen year study. Am Surg 1 11:469-473, 1966. 



chOl.qxd 4/16/04 3:20PM Page 42 



42 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



TABLE 1-32 

INCIDENCE OF VENOUS TRAUMA; 
PRELIMINARY VIETNAM VASCULAR 
REGISTRY REPORT (500 PATIENTS) 



Total vascular injuries 


718 




Venous injuries 


194 


(27.0%) 


Isolation 


28 


(14.4%) 


Combined 


166 


(85.6%) 



Modified from Rich NM, Hughes CW: Vietnam vascular 
registry: a preliminary report. Surgery 65:218-226, 1969. 



TABLE 1-33 

CONCOMITANT VENOUS TRAUMA 
ASSOCIATED WITH ACUTE ARTERIAL 
TRAUMA 



Cases 
Venous injuries 



1000 
377 



(37.7%) 



Modified from Rich NM, Baugh JH, Hughes CW: Acute 
arterial injuries in Vietnam: 1000 cases. J Trauma 
1970;10:359-369. 




■ FIGURE 1-23 

Clinical success is demonstrated 
angiographically by the patent compilation vein 
graft used to repair an injured common femoral 
vein. (Courtesy Dr. William G. Sullivan.) ■ 



Combat situations provide fertile opportuni- 
ties for young surgeons to learn by managing 
many similar injuries in a short period under 
similar circumstances and this has been 
emphasized repeatedly. Over an 8-year period 
in Vietnam (from 1965 to 1972), this oppor- 
tunity was provided for approximately 600 
young U.S. surgeons. The consensus devel- 
oped toward an increased emphasis for repair 
of major lower extremity veins that were 
injured with a particular emphasis for repair 
of the popliteal vein. Valid statistical data are 
still badly needed to determine the best 
method of venous repair, especially when end- 
to-end venous anastomosis or vein grafts are 
required. Only by such long-term evaluation 
can the reliability of different types of venous 
reconstruction be determined. Figure 1-23 
demonstrates patency of a compilation graft 
of autogenous greater saphenous vein used 
successfully to repair a defect in the left 
common femoral vein. Although this repair 
was successful in the immediate postoperative 



period, it is also important to know whether 
long-term patency can be anticipated. The 
second important area in which long-term 
data are needed is the frequency of signifi- 
cant venous insufficiency following ligation. 
Because venous insufficiency may not develop 
for several years, often after repeated episodes 
of phlebitis induced by stasis from the origi- 
nal vein ligation, long periods of observation 
are necessary. Another consideration is the 
possibility of delayed venous reconstruction 
in some patients with chronic venous insuffi- 
ciency following ligation. In such patients, 
serial venography may be useful. This is 
shown in a report by Rich and Sullivan (1972) 
of a patient with recanalization of an autoge- 
nous vein graft in the popliteal vein (Figs. 
1-24 and 1-25). Early recanalization can be 
seen in Figure 1-26. Additional phlebograms 
area needed in the extended follow-up, and 
some findings have been encouraging, such 
as the long-term patency %/ % years after lateral 
suture repair (Fig. 1-27). 



chOl.qxd 4/16/04 3:20PM Page 43 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



43 




■ FIGURE 1-24 

This venogram was performed 72 hours 
postoperatively at the 12th Evacuation Hospital 
in the Republic of Vietnam. It revealed 
thrombosis of the autogenous cephalic vein 
graft placed in the right popliteal vein. (From 
Rich NM, Sullivan WG: J Trauma 1972;12:919- 
920.) ■ 



The importance of repair of the popliteal 
vein when associated with injuries of the 
popliteal artery is discussed in further detail 
in Chapter 18. Recently, additional experience 
has been accumulated regarding the use of 
adjunctive measures. Schramek and Hash- 
monai (1974) and Schramek and colleagues 
(1975) in Israel have used a branch of the pro- 
funda femoris artery to reconstruct a distal 
arteriovenous fistula with an autogenous vein 
graft for repair of the femoral in three patients 
(Fig. 1-28). 

A study from the Vietnam Vascular Registry 
(Rich and colleagues, 1976) evaluates the man- 
agement and long-term follow-up of 110 
patients with isolated popliteal venous trauma. 
Nearly an equal number were repaired and 
ligated. Thrombophlebitis and pulmonary 
embolism were not significant complications 
in this series. The only pulmonary embolus 
occurred after ligation of an injured popliteal 
vein. However, therewas a significant increase 




■ FIGURE 1-25 

An additional venogram was performed at 
Walter Reed General Hospital approximately 
4 1 / 2 months following a venogram performed in 
Vietnam (see Fig. 1-24). Note recanalization of 
the 3-cm cephalic vein graft in the right 
popliteal vein. (From Rich NM, Sullivan WG: J 
Trauma 1972:12:919-920.) ■ 



in edema in the involved extremity following 
ligation of the popliteal vein (Table 1-34) . 
Rich (1977) also provided a 10-year follow-up 
of 51 Vietnam casualties who had lower 
extremity venous injuries repaired using au- 
togenous interposition venous grafts. Only 
onepatient (2%) developed thrombophlebitis 
in the postoperative period and this was tran- 
sitory in nature (Table 1-35) . 

Although repair rather than ligation of arte- 
rial injuries has been widely and enthusiasti- 
cally accepted for the past 40 years, the same 
approach has not developed for venous 



chOl.qxd 4/16/04 3:20PM Page 44 



44 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 




■ FIGURE 1-26 

The venogram shows minimal recanalization of 
a segment of the left greater saphenous vein, 
which was used to repair the right popliteal vein 
2 1 / 2 months earlier in Vietnam. Also note some 
of the remaining collateral venous 
development. Interestingly, the patient had no 
distal edema. (From Rich NM, Hobson RW: In 
Swan KE, et al. [eds]: Venous Surgery in the 
Lower Extremity. St. Louis: Warren H. Green 
Publishers, Inc., 1975.) ■ 



■ FIGURE 1-27 

Venogram demonstrating patency of the 
popliteal vein at its junction with the superficial 
femoral vein. Note the metallic fragments that 
caused the injury. The vein was repaired by 
lateral suture 3 1 / 2 years earlier in Vietnam. 
Repair of concomitant venous injuries is 
advocated as one of the methods that will help 
lower the relatively high amputation rate 
associated with popliteal artery trauma. (From 
Rich NM, Jarstfer BS, Geer TM: J Cardiovasc 
Surg 1974;15:340-351.) ■ 



injuries (Fig. 1-29) . In many instances, these 
have been simply treated by ligation. There 
are several reasons for this paucity of interest 
in repair. First, many veins can be ligated and 
little or no disability follows. Even when very 
large veins are ligated, the extremity may not 
be threatened, although months or years later 
venous insufficiency may appear. Second, the 
effectiveness of repair of many venous injuries 
is uncertain. With the low pressure in the 
venous system, thrombosis is much more 
common than it is after repair of arterial 



injuries. Acquisition of data to show the effec- 
tiveness of repair is particularly difficult 
because there is no simple method for patency 
of a venous reconstruction; with arterial 
repair, simple palpation of a peripheral pulse 
is usually adequate. 

The degree of disability from chronic 
venous insufficiency is not recognized by 
many, because it may become evident only 
months or years after injury. A clinical example 
of disability including venous stasis, edema, 
skin pigmentation, and ulceration following 



chOl.qxd 4/16/04 3:20PM Page 45 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



45 




■ FIGURE 1-28 

Operative photograph showing the H-type 
arteriovenous fistula, which measures 
approximately 1 cm in length and 8 mm in 
diameter (arrow), constructed approximately 2 
to 3 cm distal to the suture line of the vein graft 
(between two vascular forceps) in the femoral 
vein of the canine model. Patency of the 
autogenous vein graft in the venous system 
was enhanced by the adjuvant distal 
arteriovenous fistula. (From Rich NM, Levin PM, 
Hutton JE Jr: In Swan KE, et al. Venous Surgery 
in the Lower Extremity. St. Louis: Warren H. 
Green Publishers, Inc., 1975.) ■ 



TABLE 1-34 

INCIDENCE OF EDEMA FOLLOWING 
LIGATION AND REPAIR OF INJURED 
POPLITEAL VEINS 



Management 



No. 



Ligation 
Repair 



57 
53 



With 
Edema 

29 
7 



50.9 
13.2 



Modified from Rich NM, Hobson RW, Collins GJ Jr, Anderson 
CA: The effect of acute popliteal venous interruption. Ann 
Surg 183:365-368, 1976. 



TABLE 1-35 

COMPLICATIONS OF VENOUS REPAIR 
USING AUTOGENOUS VENOUS 
GRAFTS 



Complication 



Thrombophlebitis 


1 


2.0 


Pulmonary embolism 





0.0 


Amputation 





0.0 


Death 





0.0 



Edema 

None 
Early 
Residual 

TOTAL 



34 


66.6 


11 


21.6 


6 


11.8 


51 


100.0 



From Rich NM, Collins GJ, Andersen CA, McDonald PT: 
Autogenous venous interposition grafts in repair of major 
venous injuries. J Trauma 17:512-520, 1977. 



ligation of the superficial femoral and greater 
saphenous veins is shown in Figure 1-30. 
Because of the uncertainty of the importance 
and the effectiveness of repair of venous 
injuries, an analysis and a preliminary report 
from the Vietnam Vascular Registry were pre- 
pared in 1970; this report encouraged the 
repair of major veins in the lower extremities 
(Rich, 1970). Although data thus far are 
meager and the effectiveness of some types 
of venous reconstruction is yet unproved, 
certain clinical guidelines are now well 
established. 

Venous trauma remains a continuing chal- 
lenge with controversy regarding appropriate 
management. Selective references (1980- 
1996) emphasize experience from an expand- 
ing literature. Ironically, John B. Murphy 
emphasized in 1987 in Chicago that injured 
veins, like injured arteries, should be repaired. 
Including all medium and large-caliber veins 
in a general description might add to the exist- 
ing confusion. There is a considerable dif- 
ference in injury to the inferior or superior 
vena cava compared and contrasted to the axil- 
lary or superficial femoral veins, with the latter 
two being duplicated more frequently. Addi- 
tional controversial exchanges have centered 
on the management of injured veins by liga- 
tion or repair particularly in larger caliber 



chOl.qxd 4/16/04 3:20PM Page 46 



46 I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 




■ FIGURE 1-29 

This exhibit, entitled "Management of Venous Injuries: Clinical and Experimental Evaluation," has 
been used to stimulate an increased interest in the repair of venous injuries. Although repair of 
arterial injuries has been accepted during the past 20 years, all too often the repair of venous 
injuries has been treated with minimal interest and even disdain. (A. F.I. P. photograph.) ■ 



lower extremity veins. It has been widely rec- 
ognized that most patients can tolerate liga- 
tion of injured veins, although the contest has 
been in identifying the challenge to determine 
which patients will not tolerate the ligation of 
medium and larger veins, again, particularly 
in the lower extremities. General agreement 
emphasizes that the patient's overall condi- 
tion must be considered primarily, and the 
requirement to save a patient's life when mul- 
tiple injuries are present may necessitate lig- 
ation of injured veins. Prevention of long- 
term disability, particularly from lower extrem- 
ity swelling, on the other hand, should be 
considered and this is what has emphasized 
the importance to place a priority on the 
repair of major lower extremity veins when 
possible. 

Venous repair may be important in at least 
three circumstances. First, when popliteal 
injuries, repair of the vein may be necessary 
to prevent loss of the leg despite successful 
arterial reconstruction. This observation was 
first made during the Korean Conflict and has 
been confirmed repeatedly since that time 
(Hobson and Rich, 1995; Rich, 1995). Amajor 
factor in this decision is the anatomy of the 
popliteal space, where an injury often criti- 
cally impedes venous return from the lower 
extremity. Second, venous repair may be nec- 



essary in the presence of massive soft tissue 
injury in the extremities, where the wide- 
spread loss of soft tissue interrupts venous 
return to a crucial degree. Third, repair should 
be routinely considered with large veins, 
especially when the damage is proximal to the 
profunda femoris, to prevent chronic venous 
insufficiency. This includes the common 
femoral, and the external and common iliac 
veins. 

For a long time, a natural concern with 
repair of venous injuries was the fear of pro- 
ducing venous thrombosis and pulmonary 
embolization. Though an apparently likely 
hazard, this dangerous sequence has been sur- 
prisingly absent. Conceivably, small emboli 
may not be recognized clinically, but the 
absence of clinically detectable pulmonary 
emboli has been uniformly documented in 
both the Vietnam Vascular Registry and in civil- 
ian reports (1980 to 1995). 

There have been an increasing number of 
reports from the civilian community in the 
United States and from a variety of locations 
around the world since the documented 
experience from U.S. surgeons in Vietnam. 
Confirming and conflicting military and civil- 
ian experiences have been reported. Civilian 
reports have identified that ligation of injured 
veins, including those in the lower extremi- 



chOl.qxd 4/16/04 3:20PM Page 47 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



47 




■ FIGURE 1-30 

Chronic venous insufficiency has been seen in 
the Registry with increasing frequency in 
patients who had lower extremity venous 
ligation in Vietnam. In addition to edema, other 
changes similar to the postphlebitic syndrome 
have been evident, including venous stasis 
changes in the skin and even some superficial 
ulcerations. Some of these changes are 
present in the right lower extremity of this 
patient, who had ligation of his superficial 
femoral vein. (Walter Reed General Hospital 
1969. Vietnam Vascular Registry #225, 
NMR.) ■ 



ties, did notresultin significantmorbidity. Rec- 
ognizing that civilian and military wounds are 
considerably different, this should not be a 
surprise. Civilian wounds had in general less 
soft tissue destruction, less interruption of lym- 
phatics, and less interruption of venous col- 
laterals with fewer associated fractures with 
wounds resulting more often from knives or 
low-velocity handguns in contrast to the more 
massive military wounds, which lead to increas- 



ing morbidity with ligation of major caliber 
veins, particularly in the lower extremities. 
Ideally, determination of patients injeopardy 
for complications would be desired. Nonin- 
vasive examinations, ambulatory venous pres- 
sures, and phlebography can all be of 
assistance. These studies are, however, often 
impractical. Also, it is important to empha- 
size the inconsistency in venous anatomy. It 
would be helpful to know the anatomy before 
making claims regarding success or failure of 
management whether by ligation or repair. 
In summation, it is important to emphasize 
the difference in wounds in civilian and mil- 
itary experiences around the world recog- 
nizing that many civilian wounds have become 
more military in nature in recent years. This 
latter fact emphasizes the validity of continu- 
ing to analyze the military experiences. Long- 
term follow-up remains a major requirement. 
Nevertheless, it is becoming increasingly 
obvious that there are patients who suffer life- 
long disability from ligation of medium and 
larger caliber lower extremity veins. Many 
studies have evaluated the pathophysiology of 
acute interruption of major caliberveins. Cor- 
relation with clinical experiences and long- 
term follow-up are part of the remaining 
challenge. 



SPECIAL HISTORIC 
OBSERVATIONS 

Site of Injury 

The specific site of arterial injury is impor- 
tant. All of the cited series, except that of 
Mattox in 1989, emphasize the predomi- 
nance of extremity arterial injuries. On the 
other hand, arterial injuries in the thorax or 
abdomen may be more difficult to diagnose 
or may present additional problems in man- 
agement. There has been a relatively high mor- 
tality rate associated with arterial injuries at 
the base of the neck as a result of uncontrol- 
lable hemorrhage and cerebral ischemia. In 
1964, Pate and Wilson described experiences 
with 21 patients with arterial injuries at the 
base of the neck treated at the City of Memphis 
Hospital over a 12 year period. As would be 



chOl.qxd 4/16/04 3:20PM Page 48 



48 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



expected, there was a significant percentage 
of permanent crippling neurologic injuries. 
The interesting observation was made that 
93% of the patients who were stabbed were 
injured on the left side, suggesting that most 
of the assailants were right handed. 

Until the past 20 years, little had been pub- 
lished about intra-abdominal vascular injuries, 
perhaps because most victims died of exsan- 
guinating hemorrhage. In 1968, Perdue and 
Smith reported a group of 90 patients with 
126 separate injuries treated in Atlanta over 
a period of 10 years beginning in 1956. Most 
injuries resulted from low-velocity bullet 
wounds. Five were injured with a shotgun and 
fourteen were stab wounds. Mattox's epi- 
demiologic study reported in 1989 empha- 
sized the increasing number of truncal 
vascular injuries from civilian vascular expe- 
rience in contrast to the military vascular expe- 
rience where extremity arteries are involved 
predominantly. 



Iatrogenic Injury 

Of historical interest is the fact that one of 
the first hospital-incurred vascular injuries 
(iatrogenic injuries) was described approxi- 
mately lOOyears ago. Murphy (1897) reported 
that Heidenhain used a catgut suture to 
close a 1-cm laceration of the axillary artery, 
accidentally injured during removal of 
adherent carcinomatous glands on May 28, 
1894. The patient made a good recovery with 
no disturbance of the circulation in the 
extremity. 

Several reports have described arterial 
injuries complicating the removal of a herni- 
ated nucleus pulposus; usually the injury 
involves the common iliac artery. One of the 
first detailed reports of this complication was 
made by Seeley (1954). The injury resulted 
from the anatomic location of the iliac vessels 
on the anterior surface of the lumbar verte- 
brae, especially at the intervertebral spaces 
between the fourth and fifth lumbar vertebrae 
and between the fifth lumbar and first sacral 
ribs. In addition to the anatomic susceptibil- 
ity to injury, the use of a pituitary rongeur for 
removal of the intervertebral discs was found 
to predispose to this type of injury (Fig. 1-31) . 




Common Iliac Vs. Rt. Common Iliac A. 

■ FIGURE 1-31 

Manner in which the common iliac artery can 
be injured while using an angled pituitary 
rongeur at the intervertebral space between L4 
and L5 during removal of a herniated nucleus 
pulposus. (From Seeley SF, Hughes CW, 
Jahnke EJ Jr: Surgery 1954;35:421-429.) ■ 



At least eight such cases have been seen at 
Walter Reed General Hospital over a period 
of 25 years, including the report by Jarstfer 
and Rich (1976). Salander and colleagues 
(1984) expanded the Walter Reed Army 
Medical Center report to six patients oper- 
ated on from 1949 to 1982 for vascular injury 
following lumbar disc surgery. All six patients 
had common iliac artery injuries. 

Arteriovenous fistulas have occurred at 
numerous sites after ligation in continuity of 
an artery and a vein, such as a renal artery 
and vein following nephrectomy, the splenic 
artery and vein following splenectomy, or the 
superior thyroid artery and vein following 
thyroid lobectomy. Pritchard and colleagues 
(1977) reported an interesting case of trau- 
matic popliteal arteriovenous fistula follow- 
ing meniscectomy treated at the Mayo Clinic. 
Jimenez and colleagues (1988) presented an 
interesting case of a popliteal artery and 
venous aneurysm as a complication of arthro- 
scopic meniscectomy. There was an associated 
arteriovenous fistula. It was emphasized that 



chOl.qxd 4/16/04 3:20PM Page 4 9 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



49 



this is a rare finding following open surgical 
techniques. 

In 1968, Dillard, Nelson, and Norman 
described an arterial injury developing from 
a Kirschner wire placed through the popliteal 
artery while applying skeletal traction for a 
femoral fracture. Saletta and Freeark (1972) 
described injury of the profunda femoris 
artery caused by a drill point during an ortho- 
pedic procedure. Injuries of the femoral 
artery and vein have occurred during inguinal 
herniorrhaphy, especially during attempts to 
control hemorrhage with deep, blindly 
inserted sutures. A series of 11 iatrogenic 
injuries were reported by Lord and colleagues 
in 1958 (Table 1-36). Although retrograde 
dissection of an iliac artery and the aorta 
have been uncommon, Kay, Dykstra, and Tsuji 
(1966) have emphasized this catastrophic 
complication of cannulation and perfusion 
of the common femoral artery in open 
heart surgery. Aust, Bredenberg, and Murray 
(1981) reported five cases of arterial compli- 
cations associated with total hip replacement. 
All five injuries resulted from intraoperative 
injury. 

Kozloff and colleagues (1980) presented a 
report of eight patients seen over 18 months 
who had significant iliofemoral arterial com- 
prise secondary to cannulation for car- 
diopulmonary bypass or intra-aortic balloon 
pumping. Perler and colleagues (1983) doc- 
umented an incidence of vascular complica- 
tions of 8.8 percent utilizing the intra-aortic 
balloon pump in 794 patients at the Massa- 
chusetts General Hospital in Boston. Eighty- 
seven major vascular complications occurred 
in 70 patients. Specifically, 36 patients had a 
limb ischemia and arterial trauma occurred 
in 20. No limbswere lost. Todd and colleagues 
(1983) identified vascular complications 
related to percutaneous intra-aortic balloon 
pump inserted in 112 patients (Table 1-37). 
While six patients had reversal of ischemic 
signs following removal of the device, nine 
patients required exploration of the femoral 
artery for thrombectomy, femoral laceration 
repair, or false aneurysm repair. 

Vascular injuries following angiographic 
procedures have increased in number with the 
rapid development of precise techniques of 
angiography. The actual incidence of these 



injuries has decreased somewhat with the avail- 
ability of skilled vascular radiologists, specifi- 
cally trained for angiography, but the 
increasing utilization of such diagnostic tech- 
niques has resulted in an overall increase in 
the number of cases seen. Complications 
include hemorrhage, hematoma formation, 
false aneurysm, arteriovenous fistula, subin- 
timal dissection (with and without thrombus), 
distal embolization of thrombi material, and 
breakage of a guidewire or catheter. 

In 1971, Bolasny and Killen reviewed the 
frequency and management of arterial injuries 
following angiography at Vanderbilt Univer- 
sity over a period of 2/2 years, starting in 
January 1968. Almost 4000 angiographic pro- 
cedures were performed, following which 
there were 33 vascular injuries requiring 
surgical intervention (0.8%) (Table 1-38). 
Twenty-six of thirty-three complications were 
thrombosis at the site of catheterization. 
Sixteen involved the femoral artery, three the 
axillary, and five the brachial. In three 
instances, there was extensive dissection of the 
intima in association with thrombosis (Fig. 
1-32). 

Two patients developed arteriovenous fis- 
tulas and one had a distal embolus from the 
puncture site in the femoral artery. Almost 
none of the arterial injuries resulted simply 
from the needle puncture, in only one case 
did the injury occur from uncomplicated 
passage of a single arterial catheter. Most 
injuries occurred when manipulation of the 
catheter was "difficult" or multiple catheters 
were inserted. Complications were more 
common with arteries with atherosclerotic 
plaques. Spasm alone did not cause serious 
problems in any patient. Among numerous 
other recent papers describing complications 
associated with angiographic procedures is the 
1973 report by Brener and Couch from 
Boston. They reported a thrombosis rate of 
13% in using the brachial route for angio- 
cardiographic catheterization (Table 1-39) . 
Their overall complication rate was 6% when 
the femoral route was used, and 28% when 
the brachial route was used. 

Rich, Hobson, and Fedde (1974) described 
the Walter Reed Hospital experience with hos- 
pital-incurred vascular trauma. This was 
updated by Youkey and colleagues in 1983 



chOl.qxd 4/16/04 3:20PM Page 50 



50 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



CO 

LU 
CL 
D 
Q 
LU 

o 
o 

CC 
D. 

LU 

> 

fee 

LX 
LU 
D. 
O 

LU 

> 

h- 

o 

LU 

_l 
LU 

O 

Z 

LX 
D 
Q 

> 
CC 

D 



LX 
< 

_l 

D 

O 

CO CO 

CM < 

i> 

j O 

II 



c 
a) 

E 
u 

oi 

<u 
c 
as 






■a 
o 

3 

£■ 
a> 



Q. 

o 



a> 

< 





E o 
2 E 






o 

CD 




**- 3 






0' 




m W 






> 




co Q- 






to 














<n d 
>. o 








CL 


>. >. 


co -43 >. 


>> 


>. 


O >, >, 




■n [D i- 






pos 
over 
over 


a) a) 


" ir cd 


CD 





> > 


ied 2 
aspi 
ecov 


> 


> 


o o 


O 


o 


o o 


CJ 


o 


t 
-p 





CD 





rr cc 


Q cc 


CC 


cc 


CM CC CC 



m 


r/i 


1 ) 


1 ) 


f- 


h 


1 ) 


1 1 






in 


c/i 


CD 


CD 




6 6 



< < 



"O 
C JZ 
CO o 










CO CO 




u 


££ 


z 


c 



CO 

CO 



> .5 
™ 



CO h=- CO 



n 









CD 


CD 


CJ 


U 


r 


C 


1 > 


i 1 


f- 


t- 


f- 


f- 





O 





en 








F 




w 






CO 
CO 

E 


c 


CJ 
5^ 


_C0 



c 







r 


-XL 
O 


c 


CO 

c 

CO 




CO 


CD 
CL 


CO 

"o 



F 


CJ 

CD 




T5 
CO 


LL 


N 




LU 




LL 


CO 





























-' 


m 


en 




in 




CO 


CO 


tj- 


Lfl 




LCI 




LO 




0) 


O) 




O) 




0) 



m 


>. 


>> 




CM 


r*- 


_C 


"Nf 


FJ 


O 






<n 






(i) 












) 








T— 


CJ 



CO 


CO CO 




>^ 


~z. >■ >- 



n 


cl) 




CD 





■— 


^ 




1 






1 1 


r 


CJ 


F 





■n 


F 




( 


1 ) 


O) 





CJ 



o 
E 
E 
o 
O 



CL CO 



C "D 

O Jo 

CD c 

_l Q LL1 



= 3 £ 

C C cz 
OOO 

E E E 
E E E 

OOO 

OOO 



CD 





■" 

D_ 


C 
< 


co .y 

> cc 


CO 








B 


10 

LO 


LO 

en 


lo en 

LO ^t 




T — 


T — 


1 — -1 — 



- 




CO 


>s 


rn 


JZ 


co 


-* 


CM 


CM 













m 










CD 




















] 












CM 


CD 


CJ 


*- 



chOl.qxd 4/16/04 3:20PM Page 51 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



51 



TABLE 1-37 






INCIDENCE OF VASCULAR 




COMPLICATIONS 








No. 


(%) of 




Complications 






Survivors 






Until 




Total 


Balloon 




Group 


Removal 




(N =102) 


(N =67) 


Total no. of clinically 


15(14.7) 


15(22.4) 


evident vascular 






complications 






Limb ischemia 


6(5.9) 


6(8.9) 


responding to 






balloon removal 






Limb ischemia requiring 


6(5.9) 


6 (8.9) 


thrombectomy 






Hemorrhage requiring 


3(2.9) 


3(4.5) 


operation 







From Todd GJ, Bregman D, Voorhees AB, Reemtsma K: 
Vascular complications associated with percutaneous intra- 
acute balloon pumping. Arch Surg 118:963-964, 1983. 



TABLE 1-38 

ARTERIAL INJURY RESULTING FROM 
3934 ANGIOGRAPHIC PROCEDURES: 
0.8% INCIDENCE, VANDERBILT 
UNIVERSITY MEDICAL CENTER, 1 
JANUARY, 1968-1 JULY 1977 



Cases 



26 



% 



78.8 



Complications 

Thrombosis at site of entry 

Femoral 18 

Axillary 3 

Brachial 5 

Intimal dissection with occlusion 
Arteriovenous fistula 
Embolus from puncture site 
Perforation with hemorrhage 

TOTAL 



Modified from Bolasny BL, Killen DA: Surgical management 
of arterial injuries secondary to angiography. Ann Surg 
174:962-964, 1971. 




■ FIGURE 1-32 

Arch aortogram in a patient with angiographic 
dissection of the subclavian artery shows an 
intimal dissection caused by transfemoral 
selective arteriography. View is of the origin of 
the arch vessels. Arrow indicates intimal 
"septum" in the first portion of the left 
subclavian artery. (From Bolasny BL, Killen DA: 
Ann Surg 1971;174:962-964.) ■ 



TABLE 1-39 

INCIDENCE OF ANGIOGRAPHIC 
CATHETER COMPLICATIONS 





9.1 
6.1 
3.0 
3.0 
100.0 


Complications 


Femoral 
(223 Patients) 


Brachial 
(96 Patients) 


3 
2 

1 

1 


Thrombosis 
Stenosis 
Embolus 
False aneurysm 

TOTAL 


2 (1%) 

7 (3%) 

4 (2%) 

13(6%) 


12(13%) 
14(15%) 


33 


26 (28%) 



Modified from Brener BJ, Couch MP: Surgical arterial 
complications of left heart catheterization and their 
management. Am J Surg 125:521-526, 1973. 



chOl.qxd 4/16/04 3:20PM Page 52 



52 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



noting many similarities. Natali and Ben- 
hemori (1979) reviewed an interesting group 
of 125 cases of iatrogenic vascular injuries 
excluding angiographic injuries from Paris. 
Included were three cases in which there was 
inadvertent arterial stripping during a vein 
stripping operation. 

Alpert and colleagues (1980) presented 
five patients who sustained limb ischemia 
in neonates after umbilical artery catheteri- 
zation. Gangrene developed in three patients 
and two patients died from primary illness, 
with the third patient surviving after leg 
amputation. In the remaining two infants 
who had advanced ischemia, there was a favor- 
able response to catheter removal and 
heparinization. 

Cronenwett, Walsh, and Garrett (1988) 
identified an unusual case of multiple tibial 
artery pseudoaneurysms that appeared 4 
years after balloon catheter embolectomy. 
They reviewed the literature and found 46 
cases of balloon catheter injuries reported, 
including arterial disruption (29), intimal 
rupture (12),orcathetermalfunction (5). The 
injuries resulted in hemorrhage (13), arteri- 
ovenous fistula (12), pseudoaneurysm (4), 
thrombosis (3) , dissection (5) , accelerated ath- 
erosclerosis (4), and catheter fragment 
embolism (5). Only 41% of these complica- 
tions were recognized during the initial 
operation. 

Gurri and Johnson (1980) reviewed oper- 
ative management of 42 patients who sus- 
tained brachial arterial injury following 
cardiac catheterization at the University of 
North Carolina at Chapel Hill. 

Adar, Bass, and Walden (1982) reviewed a 
University Hospital experience with iatrogenic 
vascular complications. They emphasized that 
a concerted effort to study these injuries can 
lead to a decrease in incidence. 

Orcutt and colleagues (1985) reviewed 46 
patients who were treated for iatrogenic vas- 
cular injuries at the University of Texas Health 
Science Center in San Antonio during a 6- 
year period ending in December 1982. Diag- 
nostic procedures led to 24 injuries and 
therapeutic procedures were responsible for 
22 vascular injuries. 

Flanigan and colleagues (1983) docu- 
mented a 32-month period involving iatro- 



genic pediatric vascular injuries in 79 extrem- 
ities in 76 children in Chicago. They empha- 
sized that iatrogenic pediatric vascular injuries 
are common and can result in significant limb 
growth impairment. 

Historic Observations on 
Mechanism of Injury 

FRACTURES 

Although an arterial injury can occur with 
almost any type of fracture or dislocation, it 
is surprising that such an injury does not occur 
more often. The usual injury is confusion with 
spasm and subsequent thrombosis, rather than 
laceration or transection (Collins andjacobs, 
1961; Makins, Howard, and Green, 1966). 
Such injuries commonly have been over- 
looked in the past, confusing the signs of acute 
arterial insufficiency with soft tissue trauma, 
hemorrhage, and "arterial spasm." The avail- 
ability of angiography has greatly facilitated 
the management of such problems; the ques- 
tion of arterial injury in a patient with a frac- 
ture can be resolved simply by performing an 
angiogram. Fractures of the midshaft of the 
femur may lacerate the superficial femoral 
artery (Kirkup, 1963), whereas fractures of the 
distal tibia and fibula may lacerate the poste- 
rior and anterior tibial arteries (Miller, 1957) . 
Pelvic fractures have traumatized the iliac 
arteries, whereas medial angulation of the 
radial fragments of a fracture of the neck of 
the humerus may lacerate the axillary or 
brachial artery (Hughes, 1958) . 



POSTERIOR DISLOCATION OF 
THE KNEE 

Dislocation of the knee has frequently been 
associated with injury of the popliteal artery, 
often leading to amputation. In one series of 
22 dislocated knees, the popliteal artery was 
injured in 13 patients, an incidence of nearly 
60% (Kennedy, 1959). Similarly, Hoover, 
reporting from the Mayo Clinic in 1961, found 
9 popliteal artery occlusions associated with 
14 knee dislocations. Less commonly, dislo- 
cation of the elbow has injured the brachial 



chOl.qxd 4/16/04 3:20PM Page 53 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



53 



or radial arteries. Anterior dislocation of the 
shoulder has injured the axillary artery 
(McKenzie and Sinclair, 1958) . Trauma to the 
axillary artery may be compounded by trauma 
to the subscapular and humeral circumflex 
branches; this may make the injury more 
serious by destroying important collateral 
pathways for arterial flow to the upper extrem- 
ity. Fractures or the clavicle may injury the 
subclavian artery, the subclavian vein and the 
brachial plexus. 



BLUNT INJURY IN PRESENCE OF 
OTHER VASCULAR PATHOLOGY 

The types of blunt trauma that may in unusual 
instances injure an artery are almost endless. 
Gibson (1962) described injury from direct 
blunt force. Instances of intimal dissection, 
prolapse, and eventual thrombosis were 
reported by Elliott in 1956 and Moore in 1958. 
Ngu and Konstam (1965) reported the case 
of a woman who developed traumatic dissec- 
tion of the abdominal aorta following blunt 
trauma from a surfboard. 



USE OF CRUTCHES 

Lesions of the axillary artery have resulted 
from long-term use of crutches (Rob and 
Standeven, 1956). In 1973, Abbott and Darling 
added eight cases of axillary artery aneurysm 
secondary to crutch trauma from the Massa- 
chusetts General Hospital between 1965 and 
1971 to a review of the English literature that 
contained only 11 cases of arterial thrombo- 
sis and 2 cases of arterial aneurysms. Ettien 
(1980) reported the case of a crutch-induced 
aneurysm of the axillary artery, which resulted 
in distal embolism. 



marked, amputations have not been necessary. 
In baseball pitchers, thrombosis of the axil- 
lary artery has developed apparently as a result 
of the motion of the throwing arm from a 
position of exaggerated hyperabduction 
through a wide downward arc with great 
force. The two possibilities of injury are a 
tear of the intima from repeated stretching 
or twisting and compression from hypertro- 
phy of the pectoralis minor tendon, causing 
repetitive trauma to the artery (Whelan and 
Baugh, 1967). The importance of complete 
angiography in these unusual instances of 
arterial trauma has recently been emphasized. 
Aneurysms of the ulnar artery in the wrist 
or palm, which without angiographic inves- 
tigation would have gone undetected, have 
been found to be responsible for distal 
emboli. 



VASCULAR INJURY IN CHILDREN 

Meagher and colleagues (1979) performed a 
retrospective evaluation of vascular trauma in 
infants and children in Houston. They iden- 
tified 53 cases of blunt and penetrating vas- 
cular injuries in pediatric patients. The 
brachial artery, superficial femoral artery, and 
inferior vena cava were the vessels most often 
involved. There were 41 major arterial and 32 
major venous injuries. 

Richardson and colleagues (1981) reviewed 
the management of arterial injuries in 29 
children treated at the University of Louisville. 
Blunt trauma was responsible for 1 1 injuries, 
gunshot wounds for 9, penetrating injuries 
by sharp objects for 5 injuries, and angio- 
graphic-related injury occurred in the remain- 
ing 4. The femoral artery was most often 
injured. 



ATHLETIC INJURIES 

In baseball players, effort thrombosis of the 
subclavian artery and vein has been described. 
In addition, the syndrome has developed in 
the index finger of the catching hand, where 
the major force of the baseball is received. 
Although signs of ischemia may become 



RADIATION 

The extent of arterial trauma secondary to 
radiation therapy is not completely under- 
stood. However, it is generally believed that 
only smaller vessels are usually affected. Fre- 
quent observations have been made that there 
seems to be an increase in the friable nature 
of the vena cava during a retroperitoneal node 



chOl.qxd 4/16/04 3:20PM Page 54 



54 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



dissection following radiation for pelvic 
carcinoma. 



VIBRATORY TOOLS 

Chronic use of vibratory tools such as an air 
hammer has caused thrombosis of the distal 
arteries (Barker and Hines, 1944; De Takats, 
1959). 

Historic Classification of 
Vascular Injury 

The types of arterial injury that can occur can 
be conveniently divided into five groups, as 
follows: lacerations, transections, contusions, 
spasm, and arteriovenous fistulas (Fig. 1-33) . 
In almost every series reported, laceration or 
transection accounts for 85% to 90% of the 
total injuries seen. 

A laceration varies from a simple puncture 
wound to almost complete transection of 



the arterial wall. Transection varies from 
simple division of the artery to actual loss of 
substance from a high velocity bullet, often 
with injury of the ends of the divided artery. 
Contusion ranges from a trivial hematoma in 
the adventitia to diffuse fragmentation and 
hematomas throughout the arterial wall. In 
the most severe form, there is fracture of the 
intima, subsequent prolapse into the lumen 
and eventual thrombosis. Spasm is a definite 
entity that can occur in the absence of any 
organic injury, but it is extremely rare. It can 
be demonstrated simply in the laboratory 
by repetitively stretching an artery. This ini- 
tiates a sustained contraction of "spasm" of 
the concentric bands of smooth muscle in the 
media of the arterial wall. When it occurs, it 
is important to appreciate that spasm is a 
mechanical myogenic response and not a 
neurogenic response that is typically see in 
smaller arterial tributaries under the influence 
of the sympathetic nervous system. Arteri- 
ovenous fistulas classically occur with a for- 
tuitous injury of concomitant artery and 



i 



Laceration 





Transection 




1 



Incomplete 
Transection 




Contusion and 
Segmental Spasm 



'■";S'.<*?" 



Contusion and 
Thrombosis 




Contusion and 
True Aneurysm 




Pulsating Hematoma 
or False Aneurysm 





External 
Compression 



Arteriovenous Fistula 

■ FIGURE 1-33 

Common types of arterial trauma. Lacerations and transections account for the vast majority of 
arterial injuries. Transections may be associated with avulsions with missing segments of artery. 
External compression can be caused by displaced bone from comminuted fractures. ■ 



chOl.qxd 4/16/04 3:20PM Page 55 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



55 



vein, but the overall frequency of their occur- 
rence is small. False aneurysms evolve from 
lacerations of an artery temporarily sealed by 
blood clot. Eventually the thrombosis lique- 
fies and the lesion begins to expand. Often 
only after the appearance of an expanding 
lesion is the presence of an arterial injury first 
recognized. 



HISTORY OF BALLISTICS AND 
VASCULAR INJURY 



Primary damage and wounding results from 
direct crushing of tissue in front of the 
moving missile and from stretching and 
tearing in a wide range around the missile 
path. The stretching results from the 
formation of a large temporary cavity behind 
the missile which leaves a region of 
extravasated blood on collapse. The cavity 
formation is explosive in character and a 
comparison is drawn between a shot into 
tissue and an underwater explosion. 
Harvey (1947) 



arteries. This can be demonstrated best exper- 
imentally. The effort is warranted in view of 
an alarming increase in the number of 
gunshot wounds, including those involving 
arterial trauma, even in civilian experience. 
The mechanical disruption of arteries by high- 
velocity missiles has presented additional 
problems in arterial repair. Controversy 
regarding the extent of arterial trauma and 
the significance of this trauma to the even- 
tual success of the arterial repair stimulated 
additional experimental work based on clin- 
ical impressions from both the Korean and 
Vietnam experiences. Many misconceptions 
regarding wound ballistics have been cor- 
rected through experimental research. Even 
a lower velocity missile creates a temporary 
cavity, as shown in Figure 1-34. Nevertheless, 
the wounding power of high-velocity missiles, 
in comparison to that of lower velocity mis- 
siles, is greatly accentuated by the additional 
energy in the larger temporary cavity. Within 
microseconds after impact, the missile trans- 
fers energy to the tissues struck. Herget 
(1956) emphasized that high internal pres- 
sures and Shockwaves as high as 100 atmos- 
pheres (1500 pounds per square inch) exist 
in the temporary cavity as the tissue along the 



Primary damage and wounding results 
from direct crushing of tissue in front of the 
moving missile and from stretching and 
tearing in a wide range around the missile 
path. The stretching results from the forma- 
tion of a large temporary cavity behind the 
missile, which leaves a region of extravasated 
blood on collapse. The cavity formation is 
explosive in character and a comparison is 
drawn between a shot into tissue and an under- 
water explosion (Harvey, 1947) . 

Experimental effort has been expended by 
a small number of individuals in an attempt 
to better understand the wounding power of 
missiles, particularly during the last 50 years. 
This knowledge is of paramount importance 
before one can gain a full appreciation of the 
various etiologies of arterial trauma and 
the resultant degree of damage. As stated in 
the aforementioned quote, the temporary cav- 
itational affect of a missile has an extremely 
important adverse effect on tissues, including 




■ FIGURE 1-34 

A 16-grain sphere traveling at 1000 feet per 
second through the suspended hindlimb of a 
canine model demonstrates that there is even a 
small temporary cavity formed in muscle by a 
low velocity missile. (From Amato JJ, Billy LJ, 
Lawson NS, Rich NM: High velocity missile 
injury. An experimental study of the retentive 
forces of tissue. Am J Surg 1974;127:454- 
459.) ■ 



chOl.qxd 4/16/04 3:20PM Page 56 



56 I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 





■ FIGURE 1-35 

These angiograms of a canine model, started 10 minutes after missile wounding (the entrance 
wound is marked), show the marked increase in arterial flow in the injured leg on the dog's left side, 
compared to the contralateral side, as judged by the rapid transit of contrast media. Note in C the 
earlier venous filling. (From Rybeck B: Acta Chir Scand Suppl 1974;450:1.) ■ 



wound tract expands after the high-velocity 
missile passes through it. 

Under the auspices of the International 
Commission of the Red Cross, a series of meet- 
ings have been held by many interested 
nations to discuss the possibility of prohibi- 
tion of certain weapons used in warfare (Rich, 
1975). Included in the weapons systems that 
have been criticized are those that fire high- 
velocity bullets. In 1974 in Sweden, Rybeck 
conducted an interesting series of five exper- 
iments to determine the hemodynamic effects 
if energy absorption following missile wound- 
ing. Among their results is a graphic demon- 
stration of the increased arterial flow in the 
injured limb compared to that in the oppo- 
site uninjured limb (Fig. 1-35). 

Experimental vascular trauma continues to 
challenge the interested investigator. Some 
might say this is only a problem for military 
medicine. However, with the increasing 
number of gunshot wounds in our cities, 
including those caused by high-velocity mis- 
siles, this information also has practical value 
in our civilian community. Despite the inter- 
national prohibition at the turn of the century, 
the "dum-dum" bullet is again being used. It 
is no longer used on the battlefield; however, 
numerous law enforcement agencies in the 
United States have reinstituted or are con- 
sidering reinstitution of its use. Yet very little 
is understood regarding either the experi- 
mental or the clinical aspects of the wound- 
ing power of this missile. 



Additional international symposia on 
wound ballistics has been conducted. The 
interested reader is referred to the 1988 and 
1996 supplements from the Journal of Trauma, 
where additional detailed studies are reported 
from numerous investigators around the 
world. Additional material from individuals 
investigators is available from the Sixth Inter- 
national Symposium in Wound Ballistics held 
in November 1988 in Chongqing, Peoples 
Republic of China and in St. Petersburg, Russia 
in September 2 through 7, 1994. 

With the rapidly expanding increase in the 
management of civilian vascular trauma, it is 
equally important that long-term follow-up be 
obtained to evaluate the true success of 
various acceptable procedures and to develop 
new techniques that will help continue to 
improve the results of managing patients with 
vascular trauma. 



HISTORICAL REFLECTIONS 
AND PROJECTIONS 



We should not rest content with the work of our 
predecessors, or assume that it has proved 
everything conclusively, on the contrary it 
should serve only as a stimulus to further 
investigation. Ambroise Pare (sixteenth 
century) 



chOl.qxd 4/16/04 3:20PM Page 57 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



57 



This quote from the translated review by Bill- 
roth (1931) on studies on the nature and treat- 
ment of gunshot wounds emphasizes the 
historical development and current status of 
vascular surgery. Although some might say that 
all the principles of vascular surgery are estab- 
lished and accepted by the vast majority of 
surgeons, we must not lose sight of the need 
for continued analysis of results and contin- 
ued investigation to solve the problems that 
remain. 

As a prime example, the "ideal conduit" still 
has not been discovered, appreciating the pio- 
neering work of Voohrees reported first in 
1952. A substitute for both the arterial and 
the venous system is greatly needed. A conduit 
of varied size in diameter and length that 
would be an acceptable biologic substitute will 
always be needed in the repair of traumatized 
arteries and veins. Although some might 
argue that the ravages of atherosclerosis can 
be greatly helped by drug therapy and other 
conservative medical regimens in the future, 
the increasing incidence of injured arteries 
and veins in civil life (urban violence) and on 
the battlefield accentuates the importance of 
the need for a substitute vascular conduit. 
Many materials have been investigated, both 
clinically and under laboratory conditions. At 
Walter Reed Army Institute of Research, an 
assortment of grafts and prostheses have been 
used in the venous system without universal 
success. The problem remains more signifi- 
cant in the repair of injured veins than of 
injured arteries. Considering that the Nobel 
Prize in Medicine was awarded to Carrel in 
1912 based in part on his contributions to vas- 
cular surgery, including the reconstruction of 
arteries and veins, this might be an additional 
stimulus to the serious investigator in search 
of the "ideal conduit." 

The first vascular surgery procedures were 
described in patients with vascular trauma. For 
almost two centuries, the treatment of vascu- 
lar conditions was basically a very early history 
of the evaluation and treatment of injured 
vessels. The observations in the patients with 
injured arteries and veins led to innumerable 
concepts and laboratory experiments that 
made significant contributions to the field of 
surgery. It was the concomitant availability of 
angiography, antibiotics, plastics, vascular 



instruments developed through metallurgy, 
and synthetic monofilament suture that 
allowed the explosion of vascular surgery 
development during the 1950s and 1960s. 
Ironically, the principles established in treat- 
ment of wounded arteries and veins were 
immediately available and adaptable to the 
receptive new vascular surgeons eager to 
develop new imaginative horizons. Con- 
comitantwith this new composite technology 
and surgical vision, three major campaigns 
allowed for the field testing of many of the 
emerging concepts and instrumentation. 
Ironically, the types of lessons learned in Korea 
and Vietnam became adaptable to the third 
major warfare in the urban hospitals of 
America. There was a paradigm shift from the 
military vascular injury where over 90% were 
in the extremities to the civilian vascular 
trauma arena where over 60% were in the 
trunk. It is also ironic that this epidemic con- 
tinued through the writing of the second 
edition of this book. It is further ironic that 
during the military conflicts in Grenada 
(1983), Panama (1989), and the Persian 
Gulf (1991) and Somalia (1992), as well as 
"peace keeping" in Haiti (1994 to 1996) and 
Bosnia (1996 to 1999), that no single surgeon 
handled more than two or three vascular 
injuries. 

Continuing technology, at the time of the 
writing of the second edition of this book, 
innumerable controversies, and advance- 
ments are dynamically evolving. These include 
the areas of imaging, noninvasive evaluation, 
use of the intravascular technology, changing 
roles of the surgeon and interventional radi- 
ologists, improvements in substitute conduits 
and suture material, and changing adjuncts 
in autotransfusion and extracorporeal bypass 
and various shunts. There are also changes in 
hospital and specialty credentialing and 
recognition, as well as an explosion of spe- 
cialty organizations with interests in vascular 
surgery and in trauma. The areas of infection 
and thrombosis continue to plague the vas- 
cular surgical investigator. In the interim 
between the writing of the first two editions 
of this book, the viral infections of hepatitis 
B and C, human immunodeficiency virus, and 
others create challenges, paranoia, and ethical 
issues for the patient and the clinician alike. 



chOl.qxd 4/16/04 3:20PM Page 58 



58 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



Finally, the issue of training, role models, and 
the practice of vascular trauma has consider- 
able problems in the arena of managed care, 
health maintenance organizations, trauma 
center development, and specialty drift. The 
push for increased numbers of primary care 
physicians and primary surgeons will increase 
to the detriment to not only the vascular 
surgery specialists, but individuals who choose 
to enter the field of trauma care. Furthermore, 
the current regulatory and medical/legal 
climate and the perceptions of potential liti- 
gation involving the patient with a vascular 
injury may cause the patient who literally 
needs the care of an advanced specialist the 
most to not be able to find one during their 
moment of greatest need. 

A major factor in this remaining problem 
in vascular surgery has been the obvious 
paucity of long-term follow-up studies of vas- 
cular repairs. The importance of long-term 
follow-up of patients with vascular injuries 
cannot be overemphasized. This would be 
true of both patients who have had success- 
ful repair and those in whom repair failed or 
was not possible. In the former group, peri- 
odic evaluation of the function of the repair 
should be carried out. There is early docu- 
mentation of the value of providing details of 
vascular cases with appropriate follow-up 
information. Although arterial aneurysms 
previously had been treated by proximal lig- 
ation, excision, or the Matas repair from within 
the sac, Pringle (1913) developed a modifi- 
cation of the method used by Carrel and 
Guthrie in excising an aneurysm of the 
popliteal artery and reestablishing continu- 
ity with an autogenous saphenous vein graft. 
Both the resected popliteal aneurysm and the 
vein graft specimen were obtained years later 
postmortem, and the findings are shown in 
Figure 1-36. 

Goodman (1918), in describing his expe- 
rience at the number 1 (Presbyterian U.S.A.) 
General Hospital in France during World War 
I, reported a successful closure with continu- 
ous silk suture of 5-mm longitudinal openings 
in both the popliteal artery and vein in one 
patient with a shall fragment. However, the 
patient was followed only 9 days before being 
transferred to the Base Hospital. Goodman 
reported that "an attempt to obtain further 




■ FIGURE 1-36 

The value of adequate documentation and 
follow-up of vascular cases was demonstrated 
early by Pringle. A, An excised popliteal 
aneurysm is shown, and B, the vein specimen 
obtained years later at postmortem. Pringle 
reported his work in 1913, when he modified 
the method of Carrel and Guthrie in excising an 
aneurysm of the popliteal artery by 
reestablishing continuity with an autogenous 
saphenous vein graft. (Photograph obtained 
from and used with permission of the Royal 
College of Surgeons of Edinburgh.) ■ 



information covering the case is now under- 
way and will be embodied in a subsequent 
report." 

If there was any further follow-up infor- 
mation obtained or reported, it became 
obscured in the available literature. At least 
this military surgeon recognized the impor- 
tance of obtaining long-term follow-up infor- 
mation to thoroughly evaluate his method of 
managing vascular trauma. 



chOl.qxd 4/16/04 3:20PM Page 59 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



59 



In the classic report by DeBakey and 
Simeone (1946) from the U.S. experience in 
World War II, early patency of venous graft in 
the arterial system was demonstrated angio- 
graphically (Fig. 1-37). Although consider- 
able time and effort were expended following 
World War II in an attempt to provide addi- 
tional long-term follow-up information, the 
results of this effort are not generally avail- 
able. Individual follow-up has been possible 
in a random way for some patients, such as in 
the case of an acute, femoral arteriovenous 
fistula, mentioned earlier, which was repaired 
shortly after D-Day in Normandy on June 16, 
1944 (Boyden, personal communication, 
1970). Rob (1985) had a 10-year follow-up of 
a patient from World War II (Fig. 1-38). 

The importance of a well-documented past 
medical history covering previous vascular 
trauma was emphasized in the long-term 
follow-up of a 50-year-old former United 
States Army Officer who entered the Periph- 
eral Vascular Surgery Clinic at Walter Reed 
General Hospital for evaluation to rule out 
cerebrovascular ischemia. The patient had 
complained of several episodes of visual dis- 
turbance and weakness of his left hand during 
the past year. The patient knew that he had a 



ligation of "some of the arteries in his neck" 
during World War II. An angiogram of the 
aortic arch and its major branches was 
obtained to determine the amount of arter- 
ial flow to the brain. The study demonstrated 
no identifiable right common carotid artery 
or its branches, and there was no late retro- 
grade filling of the right internal carotid 
artery. A copy of part of his old military medical 
records was finally obtained and it was revealed 
that he had sustained a fragment wound on 
the right side of the neck on January 19, 1945, 
on Saipan when an ammunition dump 
exploded. Although only debridement was 
necessary at first, approximately 6 months later 
ligation of the right common internal and 
external carotid arteries was necessary. In sub- 
sequent follow-up through the Vascular Clinic, 
the patient had no significant problems. 
Jackson, Brengman, and Rich (1997) have 
added the latest long-term delayed vascular 
injury from Walter Reed Hospital. The patient 
was a World War II casualty who developed a 
false aneurysm of the brachial arterial branch 
about 50 years after injury (Fig. 1-39) . 

Murray (1952) stated approximately 45 
years ago that the fate of venous grafts in the 
arterial system had been under considerable 








m 



m FIGURE 1-37 

A, This arteriogram was performed 
there and one-half weeks after a 
nonsuture anastomosis of the 
superficial femoral artery. There is 
patency of the anastomosis and no 
evidence of undue ballooning of the 
vein segment. The operation was 
performed at the 8th Evacuation 
Hospital during World War II. B, This 
roentgenogram of a successful 
nonsuture anastomosis of the 
superficial femoral artery shows the 
extent of the defect bridged by the 
position of the Vitallium tubes. (From 
DeBakey ME, Simeone FA: Ann 
Surg 1946;123:534-579.) ■ 



chOl.qxd 4/16/04 3:20PM Page S 



60 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 




■ FIGURE 1-38 

This follow-up angiogram was obtained by 
Professor Charles G. Rob at St. Mary's Hospital 
in London in 1954, which was 10 years after 
successful repair of a popliteal arteriovenous 
fistula during World War II. Although patency 
was maintained, aneurysmal dilation occurred 
at the site where a portion of the sac had been 
included in the repair. This was replaced with a 
vein graph. (From Rob CG: JR Army Med 
Corps 1986;132:11-15.) ■ 



discussion. However, he felt that the saphe- 
nous grafts would continue to function 
without complications for a long period of 
time. He reported that he had removed a 
venous graft that had functioned in the 
carotid artery of a dog for nine years. Although 
the graft was slightly larger than the adjacent 
artery and there was some arteriosclerotic 
change in one area, it continued to function 
well. There are some reports of good results 
with aneurysmal formation in utilizing an 
adjacent vein, such as the femoral vein next 
to the common femoral artery as documented 
by Murray (1952) , but the greater saphenous 



vein still appears to be the best arterial sub- 
stitute, particularly for major arteries of the 
extremities. 

The outstanding documentation of the U.S. 
experience during the Korean Conflict by 
Hughes, Jahnke, Spencer, and others has pro- 
vided an opportunity for long-term follow-up. 
Figure 1-40 is an angiogram of a patient fol- 
lowed up after 19 years with a patent inter- 
position greater saphenous vein in the 
proximal right superficial femoral artery. 
The establishment of a Vascular Registry and 
Blood How Laboratory at Walter Reed General 
Hospital in 1966 provided an opportunity for 
long-term follow-up of former combat casu- 
alties who sustained vascular injuries. In the 
early efforts of the Vietnam Vascular Registry, 
the problems of obtaining long-term follow- 
up of patients who had vascular injuries in 
Vietnam were illustrated by an attempt to 
follow those listed in Fisher's report (1967) 
of 108 vascular injuries. After intensive inves- 
tigation at the time of organization of statis- 
tics for the preliminary report for the Vietnam 
Vascular Registry, it was possible to find only 
60 of his patients whose postoperative period 
and convalescence could be completely eval- 
uated. This represents slightly more than 50% 
of the patients of the original study. In sub- 
sequent years, however, the long-term follow- 
up percentage continued to improve. 

What is the long-term fate of the autoge- 
nous greater saphenous vein used as an inter- 
posed segmental graft in the arterial system? 
Most surgeons continue to believe that the 
long-term patency is excellent. However, few 
recognize the development of aneurysmal 
changes in these grafts. The long-term follow- 
up effort in the Vietnam Vascular Registry has 
continued to demonstrate an increasing 
number of patients with these changes. The 
true significance and the actual percentage 
of these changes remain unknown. This does, 
however, emphasize the great need for con- 
tinued long-term follow-up studies. This com- 
plication of fusiform aneurysmal dilation of 
an autogenous greater saphenous interposi- 
tion segment used for repair of an injured 
artery was first brought to our attention by 
Carrasquilla and Weaver (1972) when they 
reported on the follow-up of a 22-year-old 
Marine who had originally been wounded and 



chOl.qxd 4/16/04 3:20PM Page SI 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



61 




■ FIGURE 1-39 

Angiogram demonstrates a false aneurysm of a branch of the brachial artery diagnosed 49 years 
after the original injury in World War II. Successful treatment was carried out at Walter Reed Army 
Medical Center. (From Jackson MR, Brengman ML, Rich NM: J Trauma 1997;43:159-616.) ■ 



chOl.qxd 4/16/04 3:20PM Page S 2 



62 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 




■ FIGURE 1-40 

This long-term follow-up femoral angiogram 
demonstrates patency of an interposition 
autogenous greater saphenous vein used to 
repair the proximal right superficial femoral 
artery. The follow-up period extended from 
1953 until the patient was evaluated at Walter 
Reed General Hospital in 1972 (19 years). This 
is one of the longest known follow-ups and 
represents the continued effort to provide this 
type of data for former combat casualties from 
the Korean Conflict. (From Rich WR: General 
Hospital, 1978.) ■ 



this manner. B-mode ultrasonography has 
been tested to augment this information; 
however, data remain fragmentary and unsat- 
isfactory. Color-flow duplex offers good eval- 
uations of many arteries and veins, although 
the equipment is expensive (Fig. 1^13). 

The continuing challenge that remains in 
the management of patients with vascular 
injuries is exemplified by the questions and 
problems involving the search for the "ideal 
conduit" for segmental replacement of injured 
arteries and veins. There are also many other 
aspects of the management of injured patients 
with vascular injuries that could be expanded. 
The management of concomitant fractures 
associated with vascular injuries, the use of 
fasciotomy, the use of fasciotomy in extremi- 
ties with vascular injuries, and other associ- 
ated considerations are among these factors. 
Moreover, there are a multitude of profes- 
sional challenges that exist in unusual situa- 
tions involving vascular trauma. The following 
is cited as an example. Kapp, Gielchinsky, 
and Jelsma (1973) stated that they could 
find only four cases of intravascular metallic 
fragment embolization to the cerebral circu- 
lation. To their surgical review they added the 
report of two patients who were treated by the 
24th Evacuation Hospital in the Republic of 
South Vietnam. Because of the unusual 
problem, some details of one of these cases 
follow: 



treated in Vietnam. Figure 1-41 demonstrates 
the findings. With the accumulation of 
approximately 250 follow-up angiograms of 
Vietnam casualties, with the range in time from 
months to years, the number of recognized 
aneurysmal dilation of these venous interpo- 
sition grafts is in the range of 6%. 

It is often not practical or economically fea- 
sible to routinely obtain follow-up angiograms, 
particularly in asymptomatic patients. The 
long-term follow-up through the Vietnam Vas- 
cular Registry relies to a great extent on the 
noninvasive approach through the Blood 
Flow Laboratory. Figure 1-42 demonstrates 
this type of follow-up, using the measurement 
of wrist pressures and obtaining tracings with 
the Doppler ultrasound method. Unfortu- 
nately, aneurysmal changes in venous grafts 
in the arterial system cannot be detected in 



A 19-year-old American soldier received a 
fragment wound of the right side of the neck 
from a grenade explosion, associated with 
immediate onset of weakness of the left side 
of his body. An exploration of his neck was 
carried out and showed no evidence of 
vascular trauma. Three days after 
wounding, the patient was transferred to the 
24th Evacuation Hospital where he showed 
slight improvement in his left-sided 
weakness. 

Roentgenograms of the skull showed a 
small, jagged, metallic fragment, and an arte- 
riogram revealed that the fragment was lodged 
at the origin of the middle cerebral artery, 
completely occluding the middle cerebral 
artery and projecting into the carotid artery 
(Fig. 1-44). 



chOl.qxd 4/16/04 3:20PM Page S3 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



63 



Rights were not granted to include this figure in electronic media. 
Please refer to the printed publication. 



■ FIGURE 1-41 

A, Fusiform aneurysmal dilation of an autogenous greater saphenous vein segment used as an 
interposition graft in the right common carotid artery of a Vietnam casualty is demonstrated 
angiographically. B, The operative photograph demonstrates the dilated segment of saphenous 
vein. C, Arterial reconstruction was completed with a Dacron prosthesis. (From Carrasquilla C, 
Weaver AW: Aneurysm of the saphenous graft to the common carotid artery. Vase Surg 1 972;6:66- 
68.) ■ 



It was also thought that there was throm- 
bus formation around the fragment. Six days 
following injury, the right internal carotid 
artery, the anterior cerebral artery and the 
middle cerebral artery were exposed through 
a right front temporal craniotomy. After 
applying temporary vascular clamps, the frag- 
ment was removed without difficulty through 
a longitudinal arteriotomy in the internal 
carotid artery. A thrombus was also extracted 



from the middle cerebral artery. An arterior- 
rhaphy was performed, with some initial 
spasm at the repair site. In the postoperative 
period the patient's neurologic status 
improved. An arteriogram performed on the 
25th postoperative day revealed that the 
carotid artery was patent and without steno- 
sis or aneurysmal formation (Fig. 1-45) . The 
middle cerebral artery was thrombosed at 
its origin, but its branches filled readily via 



chOl.qxd 4/16/04 3:20PM Page S 4 



64 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



■ FIGURE 1-42 

The long-term follow-up through 
the Vietnam Vascular Registry 
includes the recording of wrist 
pressures and Doppler 
ultrasonicgraphic tracings. The 
two tracings at the top show the 
comparison of the right side and 
the abnormal left side, where 
occlusion of the repair of the left 
brachial artery with a saphenous 
vein graft had occurred in 1969. 
The two lower tracings show the 
change after reconstruction of 
the left brachial artery with a new 
segment autogenous greater 
saphenous vein. There is 
considerable improvement in the 
wrist pressure and Doppler 
tracing on the left. (From NM. 
Walter Reed General Hospital, 
1978.) ■ 




Left 



mm 

Wrist Pressure 150 



Pre-op 






Wrist Pressure 88 



Post-op 




Wrist Pressure 150 



Jh 


m 


:;:: 












■ '■■■■ 














a ■ 
m : 


: :: 
















: 


II 












: SL 


- ::: 


: 












IfeiJi 




\, 










•MS 


" 














liMi!! 


! - 








. 1 i 



Wrist Pressure 140 




■ FIGURE 1-43 

Color-flow duplex sonogram of axillary vein valve transfer 1 year postoperatively. No venous reflux is 
found when the patient performs a Valsalva maneuver, indicating that the valve remains competent. 
(From Goff JM, Gillespie DL, Rich NM: J Trauma 1998;44:209-211.) ■ 



chOl.qxd 4/16/04 3:21PM Page 6 5 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



65 





■ FIGURE 1-44 

A, Reports of migration of intravascular metallic 
fragments have been rare. This right carotid 
angiogram demonstrates an intra-arterial metal 
fragment at the intracranial bifurcation of the 
carotid explosion. B, This operative photograph 
taken at the 24th Evacuation Hospital in 
Vietnam shows an intra-arterial fragment with 
extreme thinning of the wall of the artery 
overlying the fragment at the origin of the 
middle cerebral artery. (From Kapp JP, 
Gielchinsky I, Jelssma R: J Trauma 
1973;13:256-261.) ■ 

collateral channels with a large patent right 
anterior cerebral artery. 

The authors outlined the possible problems 
that might occur when a metallic fragment 
lodges in a cerebral vessel: 

1. Neurologic defect secondary to arterial 
occlusion and infarction 




■ FIGURE 1-45 

This angiogram performed 25 days after 
removal of the intra-arterial metallic fragment 
shown in Figure 1-44 A and B demonstrates 
patency of the carotid artery, a patent anterior 
cerebral artery that is larger than normal, and 
thrombosis of the origin of the middle cerebral 
artery. (From Kapp JP, Gielchinsky I, Jelssma 
R: J Trauma 1973;13:256-261.) ■ 



2. Proximal and distal propagation of throm- 
bus, which could extend the infarcted area 

3. Erosion with hemorrhage 

4. Infection, arteritis, and then abscess for- 
mation or meningitis 

5. Infection with mycotic aneurysm formation 
and probable subsequent rupture 

They emphasized that one of their main 
concerns was the possibility of erosion through 
the small, thin-walled artery caused by pul- 
satile motion of the fragment. They also 
stressed the importance of maintaining a high 
index of suspicion in patients with neurologic 
symptoms who have wounds of the neck and 
chest. 

International exchange of information is 
important in the treatment of patients with 
vascular trauma. In some parts of the world, 



chOl.qxd 4/16/04 3:21PM Page 6 6 



66 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 




■ FIGURE 1-46 

Temporary intraluminal arterial shunts have been used in Munich, Germany, to reduce the ischemic 
time, to diminish thrombosis in the peripheral venous system, and to allow repair of concomitant 
lacerated veins for major arterial repair. This use of the temporary intraluminal arterial shunt in the 
management of acute arterial injuries is somewhat unique in that it was not documented in the 
United States during the same period between 1965 and 1970. (From Mack D, Scherer H, Maurer P: 
Mschr Unfallheilk 1973;76:217-224.) ■ 



specific vascular trauma, such as avulsion of 
the femoral vessels by a bull's horn in the bull- 
ring in Mexico or in Spain, may be unique to 
a certain region or country. Nevertheless, the 
common goal of all surgeons to provide the 
best medical care possible creates a desirable 
situation for exchange of data and experience 
among surgeons in all parts of the world who 
have an interest in the management of vas- 
cular injuries. Language difficulties can often 
be overcome through personal exchange and 
translation of scientific articles. An example 
of this is the personal exchange that took place 
with Doctor Peter Mauer in Munich in 1973. 
Mack, Scherer, and Maurer (1973) described 
the treatment of 154 patients with vascular 
injuries in Munich betweenjanuary 1965 and 
December 1971. They found that 80% of 129 
of their patients had suffered additional 
trauma, including fractures, trauma to the 
head, and rupture abdominal organs. Also, 
60% of their patients had vascular injuries in 
association with concomitant fractures. They 
found angiography to be of great value in diag- 
nosing the vascular injury. One relatively 
unique aspect of their management, in con- 
trast to the management of arterial injuries 
in the United States during the same time, 
was reestablishing arterial flow by temporary 
intraluminal shunts (Fig. 1-46) . They felt that 



this reduced ischemic time, diminished throm- 
bosis in the peripheral venous system, and 
allowed repair of lacerated veins before arte- 
rial repair was instituted. They were success- 
ful in restoring circulation in 75.8% of their 
patients; 12.6% showed remaining symptoms 
secondary to complications associated with 
vascular injuries, and their amputation rate 
was4.2%.BarrosD'Sa (1990) champions intra- 
luminal shunts in northern Ireland. 

In the United States, Weinstein and Golding 
(1975) used temporary external Silastic arte- 
rial and venous shunts in replanting a trau- 
matically amputated upper extremity in a 
10-year-old boy who was involved in an auto- 
mobile accident (Fig. 1-47). The level of the 
incomplete traumatic amputation was at the 
upper third of the arm, with only a posterior 
skin bridge intact. These authors emphasized 
that early arterial perfusion decreased the total 
anoxic time. The challenge persists with 
unusual and complex injuries such as this, and 
the varied and unique additions to the 
surgeon's armamentarium that might assist 
in obtaining satisfactory results should be 
known and understood. 

The following quote by Carleton Mathew- 
son, Jr., (Fig. 1-48) in the discussion of the 
paper by Morris, Creech, andDeBakey (1957) 
is most apropos: 



chOl.qxd 4/16/04 3:21PM Page 6 7 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



67 



Venous shunt 
Arterial shunt- 




■ FIGURE 1-47 

This diagrammatic drawing demonstrates the use 
of temporary external Silastic arterial an venous 
shunts during replantation of a traumatically 
amputated upper extremity in a 10-year-old boy 
involved in an automobile accident. The shunts, 
which were 20cm long and 2.5mm in internal 
diameter, reduced the anoxic time during 
replantation of the upper extremity. (From 
Weinstein MH, Golding AL: J Trauma 
1975:15:912-915.) ■ 



MILITARY SURGICAL HERITAGE 

DEPARTMENT OF SURGERY, USUHS 

CarMon Malhawtoit, Jr. 

__ 




Chlnf of Surgery (COL) 59th Evac Hoso 1942 - 1946 

Organized first Military Surgery Residency Program 1040 

Consultant Le tier man Army Hospital 1046 - 

visiting Board of Surgery, USUHS 1977 - 



■ FIGURE 1-48 

Doctor Carleton Mathewson (1902- 
1 989) as professor of surgery at 
Stanford, and subsequently at the 
University of California, provided 
leadership in establishing residency 
programs in surgery in the military 
following World War II. He served on 
the Visiting Board at the Uniformed 
Services University of the Health 
Sciences, (Bethesda, Maryland) with 
other senior surgeons identified in 
this manuscript. ■ 



chOl.qxd 4/16/04 3:21PM Page 6 8 



68 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



Unfortunately in many quarters these lessons 
so well emphasized during the stress of 
world conflict have been neglected in the 
complacency of civilian life. It is important, 
therefore, that we re-emphasize the 
seriousness of vascular injury and, where 
possible, stress the favorable 
circumstances that present themselves in 
civilian life with the successful primary 
repair of injured vessels. 

Endovascular procedures offer a new and 
alternative consideration to the more tradi- 
tional suture repair of injured arteries and 
veins in the 1990s. Parodi (1990) in Buenos 
Aires championed this approach. In collabo- 
ration with Marin and Veith in New York City 
(1994) endovascular approaches have been 
used for primary repairs of arteriovenous fis- 
tulas and false aneurysms in most cases (Fig. 
1-49) . Durability and ultimate success await 
needed follow-up. 

Exciting challenges remain. From the 
microbiology research, we know of a con- 





* * 1 

BAU.OMV I 

01 



■ FIGURE 1-49 

Angiographic images of a patient who 
sustained a gunshot wound to the right chest. 
A, Prograde arteriogram showing subclavian 
artery and active bleeding in the region. B, 
Image after proximal balloon occlusion, which 
stopped the bleeding. C, Retrograde brachial 
arteriogram showing extent of injury. D, 
Completion arteriogram after successful repair. 
(From Patel AV, Marin MD, Veith FJ, et al. J 
EndovascSurg 1996;3:382-388.) ■ 



nection between P-55 receptor site alterations 
and complications of vascular manipulation 
to include thrombosis and possible stenosis. 
Nevertheless, while basic research continues 
so does daily examples of man's inhumanity 
to man resulting in vascular trauma now in 
regional conflicts and in urban violence. 



REFERENCES 

Abbott WM, Darling RC: Axillary artery aneurysms 
secondary to crutch trauma. Am J Surg 1973; 
125:515-520. 

Adar R, Bass A, Walden R: Five years' experience 
in general and vascular surgery in a university 
hospital. Ann Surg 1982;196:725-729. 

Agnew D II: The Principles and Practice of Surgery. 
Philadelphia, JB Lippincott Co, 1978. 

Alpert J, O'Donnell JA, Parsonnet V, et al: Clini- 
cally recognized limb ischemia in the neonate 
after umbilical artery catheterization. Am J Surg 
1980;140:413-418. 

AustJC, Bredenberg CE, Murray DG: Mechanisms 
of arterial injuries associated with total hip 
replacement. Arch Surg 1981;116:345-349. 

Barker NW, Hines EAJr: Arterial occlusion in the 
hands and fingers associated with repeated 
occupational trauma. Proc Staff Meet Mayo Clin 
1944;19:345-349. 

Barros D'Sa AAB, Hassard TH, Livingston RH, 
Irwin JW: Missile-induced vascular trauma. Injury 
1980;12:13-30. 

Bell J: Principles of surgery. Discourse 1801;9:4. 

Bernheim BM: Bloodvessel surgery in the war. Surg 
Gynecol Obstet 1920;30:564-567. 

Billroth T: Historical studies on the nature and treat- 
ment of gunshot wounds from the 15th century 
to the present time. Yale J Biol Med 1931;4:16- 
36. 

Bolasny BL, Killen DA: Surgical management of 
arterial injuries secondary to angiography. Ann 
Surg 1971;174:962-964. 

Bole PV, Purdy RT, Munda RT, et al: Civilian arte- 
rial injuries. Ann Surg 1976;183:13-23. 

Boyden AM: Personal communication, 1970. 

Brener BJ, Couch N: Peripheral arterial compli- 
cations of left heart catheterization and their 
management. Am J Surg 1973;125:521-526. 

Carrasquilla C, Weaver AW: Aneurysm of the saphe- 
nous graft to the common carotid artery. Vase 
Surg 1972;6:66-68. 



chOl.qxd 4/16/04 3:21PM Page S 9 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



69 



Carrel A: La technique operatoriesdes anastomoses 
vasculaires e t la tran splan tation de s viscere s. Lyon 
Medical 1902;98:859. 

Carrel A, Guthrie CC: Uniterminal and bitermi- 
nal venous transplantations. Surg Gynecol Obstet 
1906;2:266-286. 

Chandler JG, RnappRW: Early definitive treatment 
of vascular injuries in the Vietnam conflict. JAMA 
1967;202:960-966. 

Cheek RC, Pope JC, Smith HF, etal: Diagnosis and 
management of major vascular injuries: A review 
of 200 operative cases. Am Surg 1 975;41 :755-760. 

Clermont G: Suture laterale et circulaire des 
veines. Presse Med 1901;1:229. 

Coley RW. Case of rupture of the carotid artery 
and wound of several of its branches successfully 
treated by tying off the common trunk of the 
carotid itself. Med Chir J (Lond) 1817;3:2. 

Collins HA, Jacobs JK Acute arterial injuries due 
to blunt trauma. J Bone Joint Surg 1 961 ;43A: 193- 
197. 

CronenwettJL, Walsh DB, Garrett HE: Tibial artery 
pseudoaneurysms: delayed complication of 
balloon catheter embolectomy. J Vase Surg 
1988;8:483-488. 

DeBakey ME, Simeone FA: Battle injuries of the 
arteries in World War II: An analysis of 2,471 cases. 
Ann Surg 1946;123:534-579. 

De Takats G: Vascular Surgery. Philadelphia, WB 
Saunders, 1959. 

Dillard BM, Nelson DL, Norman HG: Review of 85 
major traumatic arterial injuries. Surgery 
1968;63:391-395. 

Dorfler J: Uber arteriennaht. Beitr Klin Chir 
1889;25:781. 

Drapanas T, Hewitt RL, WeichertRF III, Smith AD: 
Civilian vascular injuries: A critical appraisal of 
three decades of management. Ann Surg 
1970;172:351-360. 

Eck NV: K. voprosu o perevyazkie vorotnois veni. 
Predvaritelnoye soobshtshjenye (Ligature of the 
portal vein). Voen Med J 2001;130:1-2. 

ElliotJA: Acute arterial occlusion: an unusual cause. 
Surgery 1956;39:825-827. 

Ellis J: Case of gunshot wound, attended with sec- 
ondary hemorrhage in which both carotid arter- 
ies were tied at an interval of four and a half 
days. N YJ Med 1845;5:187. 

Ettien JT: Crutch-induced aneurysms of the axil- 
lary artery. Am Surg 1980;46:267-269. 

Feliciano DV, Bitondo CG, Mattox KL, et al: Civil- 
ian trauma in 1 980s: A one-year experience with 



456 vascular and cardiac injuries. Ann Surg 
1984;199:717-724. 

Ferguson IA, Byrd WM, McAfee DK: Experiences 
in the management of arterial injuries. Ann Surg 
1961;153:980-986. 

Fisher GW: Acute arterial injuries treated by the 
United States Army Medical Service in Vietnam, 
1965-1966. J Trauma 1967;7:844-855. 

Flanigan DP, Keifer TJ, Schuler JJ, et al: Experi- 
ence with iatrogenic pediatric vascular injuries: 
Incidence, etiology, management, and results. 
Ann Surg 1983;198:430-442. 

Caspar MR, Treiman RL: The management of 
injuries to major veins. AmJ Surg 1960;100:l7l- 

175. 

Gensoul: Note sur les Bless es recus a l'Hotel Dieu 
de Lyon, pendant les troubles de 1831 . Gaz Med 
Paris 1883;297. 

Gibson JMC: Rupture of axillary artery. J Bone Joint 
Surgl962;44B:114-115. 

Gliick T: Uber zwei falle von aortenaneurysmen 
nebst bemerkungen uber die naht der blutge- 
fasse. Arch Klin Chir 1883;28:548. 

Goodman C: Suture of blood vessel from projec- 
tiles of war. Surg Gynecol Obstet 1918;27:528. 

Goyanes J: Neuvos trabajos de chirurgial vascular: 
substitution plastica de las the arterias por las 
venas, o arterio-plastia venosa, aplicado, como 
neuvo metodo, al traitamiento de los aneuris- 
mas. El Siglo Med 1906;53:546-561. 

GurriJA, Johnson G Jr: Management of brachial 
artery occlusion after cardiac catheterization. Am 
Surg 1980;46:233-235. 

Guthrie GJ: Disease and injuries of arteries. 
1830. 

Haimovici H: History of arterial grafting. J Cardiac 
Surg 1963;4:152-174. 

Hallowell. Extract of a letter from Mr. Lambert, 
surgeon at Newcastle upon Tyne, to Dr. Hunter, 
giving an account of new method of treating an 
aneurysm. Med Obser Inq 1762;30:360. 

Halsted WS: The effect of ligation of the common 
iliac artery on the circulation and function of 
lower extremity. Report of a cure of iliofemoral 
aneurysm by their application of an aluminum 
band to the vessel. Bull Johns Hopkins Hosp 
1912;23:191-220. 

Hardy JD, Raju S, Neely WA, Berry DW: Aortic and 
other arterial injuries. Ann Surg 1975;181:640- 
653. 

Heaton LD, Hughes CW, Rosegay H, et al: Military 
surgical practices of the United States Army in 



chOl.qxd 4/16/04 3:21PM Page 70 



70 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



Vietnam. In Current Problems in Surgery. 
Chicago, Year Book Medical Publishers, 
1966. 

Herget CH: Wound ballistics. In Bowers WB (ed) : 
Surgery of Trauma. Philadelphia, JB Lippincott 
Co, 1956. 

Hobson RW II, Rich NM: Traumatismes veineux 
des membres inferieurs. In Kieffer E (ed) : Trau- 
matismes Arteriels. Paris, AERCV, 1995. 

Hobson RW II, Rich NM, Wright CB: Venous 
Trauma: Pathophysiology, Diagnosis and Surgi- 
cal Management. Mt. Kisco, NY, Futura Pub- 
lishing, 1983. 

Hoover NW: Injuries of the popliteal artery asso- 
ciated with fractures and dislocations. Surg Clin 
North Am 1961;41:1099-1112. 

Hughes CW: Acute vascular trauma in Korean War 
casualties: An analysis of 180 cases. Surg Gynec 
Obstet 1954;99:91-100. 

Hughes CW: The primary repair ofwounds of major 
arteries: An analysis of experience in Korea in 
1953. Ann Surg 1955;141:297-303. 

Hughes CW: Arterial repair during the Korean War. 
Ann Surg 1958;147:555-561. 

Hughes CW: Vascular surgery in the armed forces. 
Milit Med 1959;124:30-46. 

Hunter W: The history of an aneurysm of the aorta, 
with some remarks on aneurysms in general. Med 
Obs Soc Phys Lond 1757;1:323. 

Inui FK, Shannon J, Howard JM: Arterial injuries 
in the Korean conflict: Experiences with 111 con- 
secutive injuries. Surgery 1955;37:850-857. 

Jackson MR, Brengman ML, Rich NM: Delayed pre- 
sentation of 50 years after a World War 2 vascu- 
lar injury with intraoperative localization by 
duplex ultrasound of a traumatic false aneurysm. 
J Trauma 1 997;43: 1 59-1 61 . 

Jahnke EJJr: Late structural and functional results 
of arterial injuries primarily repaired. Surgery 
1958;43:175-183. 

Jahnke EJJr, Seeley SF: Acute vascular injuries in 
the Korean War: An analysis of 77 consecutive 
cases. Ann Surg 1953;138:158-177. 

Jarstfer BS, Rich NM: The challenge of arteriove- 
nous fistula formation following disk surgery: A 
collective review. J Trauma 1976;16:726-733. 

Jassinowsky A: Die arteriennhat: Eine experi- 
mentelle studie. Inaug Diss Dorpat 1889. 

Jensen: Ueber circulare gefassutur. Arch Klin Chir 
1903;69:938-998. 

Jimenez F, Utrilla A, Cuesta C, etal: Popliteal artery 
and venous aneurysm as a complication of 



arthroscopic meniscectomy. J Trauma 1988;28: 
1404-1405. 

Kapp JP, Gielchinsky I, Jelsma R: Metallic fragment 
embolization to the cerebral circulation. J 
Trauma 1973;13:256-261. 

Kay JK Dykstra PC, Tsuji HK: Retrograde ilioaor- 
tic dissection. A complication of common 
femoral artery perfusion during open-heart 
surgery. Am J Surgery 1966;111:464-468. 

Kelly GL, Eiseman B: Civilian vascular injuries. 
J Trauma 1975;15:507-514. 

Kennedy JC: Complete dislocations of the knee. 
J Bone Joint Surg 1959;41:878. 

Kozloff L, Rich NM, Brott WH, et al: Vascular 
trauma secondary to diagnostic and therapeu- 
tic procedures: Cardiopulmonary bypass and 
intra-aortic balloon assist. Am Surg 

1980;140:302-305. 

Kummel: Uber circulare naht der gefasse. Munch 
Med Wschr 1899;46:1398. 

KirkupJR: Major arterial injury complicating frac- 
ture of the femoral shaft. J Bone Joint Surg 
1963;45:337-343. 

Kurtoglu M, Ertekin C,BulutT, etal: Management 
of vascular injuries of the extremities. One 
hundred and fifteen cases. Int Angiol 1991; 
10:95. 

Langenbeck B: Beitrage zur chirurgischen patholo- 
gie der venen. Arch Klin Chir 1861 ;1:1. 

Learmonth J: Vascular injuries in war. Royal Soc 
Med 1946;39:488. 

Lexer E: Die ideale operation des arteriellen und 
des arte riell-venosen aneurysma. Arch Klin Chir 
1907;83:459-477. 

Linton R: Injuries to major arteries and their treat- 
ment. N YJ Med 1949;49:2039. 

Lord JW, Stone P, Clouthier W, Breidenbach L: 
Major blood vessel injury during elective surgery. 
Arch Surg 1958;77:282. 

Luetic V, Sosa T, Tonkovic I, et al: Military vascu- 
lar injuries in Croatia. Cardiovasc Surg 1993;1 :3- 
6. 

Makins G: Gunshot injuries to the blood vessels. 
Bristol, England, John Wright and Sons, 
1919. 

Makins G, Howard JM, Green R: Arterial injuries 
complicating fractures and dislocations: The 
necessity for a more aggressive approach. Surgery 
1966;59:203-209. 

Matas R: An operation for radical cure of aneurysm 
based on arteriography. Ann Surg 1903;37:161- 
196. 



chOl.qxd 4/16/04 3:21PM Page 71 



1 • HISTORICAL AND MILITARY ASPECTS OF VASCULAR TRAUMA 



71 



Matas R: Military Surgery of the Vascular System. 
Philadelphia, WB Saunders, 1921. 

Mattox KL: Approaches to trauma involving the 
major vessels of the thorax. Surg Clin North Am 
1989;69:77-92. 

Mattox KL, Feliciano DV, Burch J, et al: Five thou- 
sand seven hundred sixty cardiovascular injuries 
in 4459 patients: epidemiologic evolution 1958 
to 1987. Ann Surg 1989;209:698-707. 

McKenzie A, Sinclair A: Axillary artery occlusion 
complicating shoulder dislocation: a report of 
two cases. Ann Surg 1958;148:139-144. 

Meagher DJ, Defore W, Mattox KL, Harberg F: Vas- 
cular trauma in infants and children. J Trauma 
1979;19:532-536. 

Miller D: Gangrene from arterial injuries associ- 
ated with fractures and dislocations of the leg in 
the young and in adults with normal circulation. 
Am J Surg 1957;93:367-375. 

Moore T: Acute arterial obstruction due to the trau- 
matic circumferential intimal fracture. Ann Surg 
1958;148:111-114. 

Moore C, Wolma F, Brown R, Derrick J: Vascular 
trauma: A review of 250 cases. Am J Surg 
1971;122:576-578. 

Morris GCJr, Creech OJr, DeBakey ME: Acute arte- 
rial injuries in civilian practice. Am J Surg 
1957;93:565-572. 

Mullins R, Lucas C, Ledgerwood A: The natural 
history following venous ligation for civilian 
injuries. J Trauma 1980;20:737-743. 

Murphy J: Resection of arteries and veins injured 
in continuity end-to-end suture. Exp Clin Res 
Med Rec 1897;51:73-104. 

Murray G: Surgical repair of injuries to main arter- 
ies. Am J Surg 1952;83:480. 

Natali J, Benhemori AC: Iatrogenic vascular 
injuries. J Cardiovasc Surg 1979;20:169-176. 

Ngu V, Konstam PG: Traumatic dissecting 
aneurysm of the abdominal aorta. Br J Surg 
1965;52:981-982. 

Nolan B: Vascular injuries. J Royal Coll Surg 
1968;13:72. 

Oiler D, Rutledge R, Clancey T, et al: Vascular 
injuries in a rural state: A review of 978 patients 
from a state trauma registry. J Trauma 1992;32: 
740-746. 

Orcutt M, Levine B, Gaskill H III, Sirinek K Iatro- 
genic vascular injury: A reducible problem. Arch 
Surg 1985;120:384-385. 

Pate J, Wilson H: Arterial injuries of the base of 
the neck. Arch Surg 1964;89:1106-1110. 



Patman R, Poulos E, Shires G: The management 
of civilian arterial injuries. Surg Gynecol Obstet 
1964;118:725-738. 

Perdue GJ, Smith RI: Intra-abdominal vascular 
injury Surgery 1968;64:562-568. 

Perler B, McCabe C, Abbott WM, Buckley MJ: Vas- 
cular complications of in tra-aortic balloon coun- 
terpulsation . Arch Surg 1983;118:957-962. 

Perry MO, Thai E, Shires G: Management of arte- 
rial injuries. Ann Surg 1971;173:403-408. 

Postempski P: La sutura dei vasi sanguigni. Arch 
Soc Ital Chir Roma 1886;3:391. 

Pritchard D, MaloneyJ, Barnhorst DA, Spittel JJ: 
Traumatic popliteal arteriovenous fistulas. Arch 
Surg 1977;112:849-852. 

Reynolds R, McDowell H, Diethelm A: The surgi- 
cal treatment of arterial injuries in the civilian 
population. Am Surg 1979;189:700-708. 

Rich NM: Vietnam missile wounds evaluated in 750 
patients. Milit Med 1968;133:9-22. 

Rich NM: Vascular trauma in Vietnam. J Cardio- 
vasc Surg 1970;11:368-377. 

Rich NM: Venous injuries. In Sabiston DJ (ed) : Text- 
book of Surgery. Philadelphia, WB Saunders, 
1995. 

Rich NM, Hobson RW II, Collins GJ Jr, Anderson 
CA: The effect of acute popliteal venous inter- 
ruption. Ann Surg 1976;183:365-368. 

Rich NM, Hobson RW II, Fedde C: Vascular trauma 
secondary to diagnostic and therapeutic proce- 
dures. Am J Surg 1974;128:715-721. 

Rich NM, Hughes CW: Vietnam Vascular Registry: 
A preliminary report. Surgery 1969;65:218-226. 

Rich NM, Spencer FC: Vascular Trauma. Philadel- 
phia, WB Saunders, 1978. 

Rich NM, Sullivan W: Clinical recanalization of an 
autogenous vein graft in the popliteal vein. J 
Trauma 1972;12:919-920. 

Richardson JD, Fallat M, Nagaraj H, et al: Arterial 
injuries in children. Arch Surg 1981 ;1 16:685-690. 

Rob CG: A history of arterial surgery. Arch Surg 
1972;105:821-823. 

Rob CG, Standeven A: Closed traumatic lesions of 
the axillary and brachial arteries. Lancet 
1956;1:597-599. 

Rybeck B: Missile wounding and hemodynamic 
effects of energy absorption. Acta Chir Scand 
1974;450:1. 

Salander J, YoukeyJ, Rich NM, e t al: Vascular injury 
related to lumbar disk surgery. J Trauma 
1984;24:628-631. 



chOl.qxd 4/16/04 3:21PM Page 72 



72 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



Saletta J, Freeark R: The partially severed artery. 
Arch Surg 1968;97:198-205. 

Saletta J, Freeark R: Injuries to the profunda femoris 
artery. J Trauma 1972;1 2:778. 

Schede M: Zur frage von der jodoformvergiftung. 
Zentralb Chir Beil 1882;9:33-38. 

Schramek A, Hashmonai M: Distal arteriovenous 
fistula for the prevention of occlusion of venous 
interposition grafts to veins. J Cardiovasc Surg 
1974;15:392-395. 

Schramek A, Hashmonai M, Farbstein J, Adler O: 
Reconstructive surgery in major vein injuries in 
the extremities. J Trauma 1975;15:816-822. 

Schwartz A: The historical development of methods 
of hemostasis. Surgery 1958;44:604. 

Seeley S: Vascular surgery at Walter Reed Army Hos- 
pital. US Armed Forces Med J 1954;8. 

Shumacker HBJr: Incisions in surgery of aneurysm: 
With special reference to explorations in the 
antecubital and popliteal fossae. Ann Surg 
1946;124:586-598. 

Shumacker HB Jr: Surgical cure of innominate 
aneurysm; report of a case with comments on 
applicability of surgical measures. Surgery 
1947a;22:729-739. 

Shumacker HBJr: Resection of the clavicle with 
particular reference to the use of bone chips in 
the periosteal bed. Surg Gynecol Obstet 
1947b;84:245-248. 

Shumacker HBJr: Problem of maintaining the con- 
tinuity of artery in surgery of aneurysms and arte- 
riovenous fistulae; notes on the development and 
clinical application of methods of arterial suture. 
Ann Surg 1948a;l 27:207-230. 

Shumacker HBJr: Operative exposure of the blood 
vessels in the superior anterior mediastinum. Am 
Surg 1948b;l 27:464-475. 

Shumacker HBJr: Arterial suture techniques and 
grafts: Past, present and future. Surgery 
1969;66:419-433. 

Simeone F, Grillo H, Rundle F: On the question 
of ligation of the concomitant vein when a major 
artery is interrupted. Surgery 1951;29:932-951. 



Smith R, Elliot J, Hageman J: Acute penetrating 
arterial injuries of the neck and limbs. Arch Surg 
1974;109:198-205. 

Smith L, Foran R, Caspar MR: Acute arterial injuries 
of the upper extremity. Am J Surg 1963;106:144- 

151. 

Soubbotitch V: Military experiences of traumatic 
aneurysms. Lancet 1913;2:720-721. 

Spencer FC, Grewe R: The management of acute 
arterial injuries in battle casualties. Ann Surg 
1955;141:304-313. 

Stich R: Ueber gefaess und organ transplantatio- 
nen mittelst gefaessnaht. Ergeon Chir Orth 
1910;1:1. 

Swan KG, Hobson RWII, Reynolds D, et al: Venous 
surgery in the lower extremities. In Swan KG, 
Hobson R, Reynolds D, et al (eds): St Louis, 
Warren H Green Publishers, 1975. 

Todd G, Bregman D, Voorhees A, Reemtsma K Vas- 
cular complications associated with percuta- 
neous intra-aortic balloon pumping. Arch Surg 
1983;118:963-964. 

Vollmar J: Surgical experience with 197 traumatic 
arterial lesions (1953-66) . In Hiertonn T,Rybeck 
B (eds) : Traumatic Arterial Lesions. Stockholm, 
Forsvarets Forskningsanstalt, 1968. 

Warren R: Report to the Surgeon General. Wash- 
ington, DC, Department of the Army, 1952. 

Weinstein M, Golding A: Temporary external 
shunt bypass in the traumatically amputated 
upper extremity. J Trauma 1975;15:912-915. 

Whelan TJ, Baugh JH: Non-Atherosclerotic Arter- 
ial Lesions and Their Management. Current 
Problems in Surgery. Chicago, "Vfear Book Medical 
Publishers, 1967. 

\bukeyj, Clagett GP, Rich NM, et al: Vascular trauma 
secondary to diagnostic and therapeutic proce- 
dures: 1974 through 1982. Am J Surg 1983;146: 
788-791. 



ch02.qxd 4/16/04 3:19 PM Page 73 




Ischemia and 
Reperfusion Injury 



IRSHAD H. CHAUDRY 
PING WANG 
DORAID JARRAR 



O INTRODUCTION 

O ISCHEMIA-REPERFUSION INJURY 

O ROLE OF ENDOTHELIAL CELLS FOLLOWING ISCHEMIA-REPERFUSION 

O LEUKOCYTE-ENDOTHELIAL CELL INTERACTION FOLLOWING 
ISCHEMIA-REPERFUSION 

O ROLE OF PLATELETS AND COMPLEMENT SYSTEM 

O DIVERGENT EFFECTS OF NITRIC OXIDE AND SUPEROXIDE 
FOLLOWING ISCHEMIA-REPERFUSION 

O CONFOUNDINGCOMORBIDITIESFOLLOWING ISCHEMIA-REPERFUSION 

O SYSTEMIC LEVELS OF INFLAMMATORY MEDIATORS AND REMOTE 
ORGAN INJURY 

O SIGNIFICANCE OF ISCHEMIC PRECONDITIONING 

O MODIFYING FACTORS DETERMINING ISCHEMIA-REPERFUSION INJURY 

O TREATMENT OF ISCHEMIA-REPERFUSION INJURY: ROLE OF 
PHARMACOLOGIC ADJUNCTS 

O SUMMARY 



73 



ch02.qxd 4/16/04 3:19 PM Page 74 



74 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



INTRODUCTION 



The original Greek words ischein and haima 
mean to hold and blood. In the modern 
medical field, this means the lack of circula- 
tion or perfusion. In this chapter, the patho- 
physiology of ischemia and the clinical 
implications for the clinician are outlined. 
Specific attention is devoted to ischemia 
caused by vascular injury, as well as the 
potential therapeutic options under those 
conditions (Bickwell and colleagues, 1989; 
Burch and colleagues, 1990; Bickell and 
colleagues, 1994; de Guzman and colleagues, 
1999). Vascular injury is present in approxi- 
mately 20% of all trauma admissions to a ter- 
tiary care center. Besides obvious vascular 
trauma with the threat of immediate exsan- 
guination, vascular compromise with subtle 
changes in perfusion and subsequent 
ischemia is an important issue in the sec- 
ondary survey of the traumatized host. A thor- 
ough physical examination of the patient is, 
therefore, mandatory to identify vascular 
compromise and possible ischemia of the 
dependent organ or system. 

With the evolution of multicellular organ- 
isms, the development of the cardiovascular 
system was essential, because the exchange of 
nutrients and oxygen is limited by diffusion 
and, therefore, is feasible only for single-cell 
organisms. In an adult human, thousands 
of miles of vessels of various size, shape, and 
capacity provide the body with oxygen and 
nutrients. This complex system also is respon- 
sible for the clearance of waste products and 
serves as the transport medium for hormones 
to reach their target sites. With the depen- 
dence of virtually all organs on aerobic metab- 
olism, a stasis or even a reduction of blood 
flow will inevitably result in tissue damage or 
death, unless the collateral blood flow is 
sufficient to meet the metabolic demands of 
the affected organ bed. 

The terms ischemia and hypoxia have been 
used indiscriminately. In view of this, differen- 
tiation between these two terms is important. 
In this chapter, ischemia refers to a total lack of 
oxygen, whereas hypoxia refers to decreased 
oxygen availability. Furthermore, whereas 
ischemia, that is, stasis of blood flow, inevitably 
leads to hypoxia of the dependent organ or 



organ system, hypoxia can occur in the pres- 
ence of normal blood flow. Moreover, it 
appears that the detrimental effects of 
ischemia are not only due to the lack of oxygen, 
but also to the role of blood both to preserve 
tissue homeostasis and to deliver oxygen. 

Clinical decision making is highly depen- 
dent on whether the ischemic event occurred 
acutely or has evolved over a prolonged time. 
If there has been a slow onset of decreased 
blood flow, leading ultimately to ischemia, col- 
lateral blood supply may have developed. 
However, in the case of traumatic vascular 
injury to an extremity, revascularization must 
be accomplished within 6 hours, because 
warm ischemia time for striated muscle results 
in irreversible damage after 6 to 8 hours. 

With the re-establishment of blood flow to 
a previous ischemic organ or limb, reperfu- 
sion is initiated, that is, ischemia-reperfusion 
(I/R) . This event also marks the onset of 
reperfusion injury, a complex event that 
involves many cellular and hormonal com- 
ponents including oxygen radicals, neu- 
trophils, and complement activation. 
Reperfusion of an ischemic vascular bed not 
only produces local injury but also could 
produce distant organ injury. 

The clinical hallmarks of ischemia are the 
five Ps: pain, pulselessness, paresthesia, pallor, 
and paralysis. In the event of acute ischemia, 
pain and pulselessness are the leading clini- 
cal symptoms. Nevertheless, it should be kept 
in mind that in the multi-injured or uncon- 
scious patients, the clinical diagnosis may be 
difficult, and an appropriate vascular exami- 
nation including Doppler flow ultrasound is 
mandated if vascular injury or compromise is 
suspected. If vascular trauma is present, the 
therapeutic goal should be restoration of func- 
tion to the preinjury level (Burch and col- 
leagues, 1990; Bickell and colleagues, 1994). 
In case of extremity trauma, warm ischemia 
time should not exceed 6 hours to ensure com- 
plete recovery of the extremity. 



ISCHEMIA-REPERFUSION 
INJURY 

Ischemia caused by conditions such as hem- 
orrhagic shock, vascular trauma, and cardiac 



ch02.qxd 4/16/04 3:19 PM Page 75 



2 • ISCHEMIA AND REPERFUSION INJURY 



75 



arrest, followed by reperfusion of the tissues 
with oxygenated blood, can compromise 
microvascular and cellular integrity (Massberg 
and Messmer, 1998; Ikeda and colleagues, 
2000; Lefer, 1999; Lefer, 1994). The patho- 
physiologic mechanisms causing the so-called 
I/R injury are quite complex and involve a 
variety of cell populations including endothe- 
lial cells, leukocytes, hormones, up-regulation 
of cell surface proteins, and activation of the 
complement system. Part of the I/R injury is 
attributable to the phenomenon of slow reflow 
or no reflow, which is characterized by reduced 
blood flow despite the restoration of adequate 
perfusion pressure (Menger and colleagues, 
1992). Although this phenomenon is still 
poorly understood, apparently, leukocytes, at 
least partially, mediate postischemic microvas- 
cular compromise (Schlag and colleagues, 
2001; Kadambi and Skalak, 2000; Menger and 
colleagues, 1997; Waxman, 1996; Nolte and 
colleagues, 1991; Lehr and colleagues, 1991) . 
Leukocyte adhesion to the endothelium is 
significantly enhanced following I/R injury, 
which is mediated by several adhesion mole- 
cules on the surface of leukocytes and/or 
endothelial cells such as immunoglobulin-like 
receptors (intercellular adhesion molecule-1 
[ICAM-1], platelet endothelial cell adhesion 
molecule-1 [PECAM-1 ] , vascular cell adhesion 
molecule-1 [VCAM-1]), integrins (CD11/ 
CD18) , and selectins (E-, P-, L-selectin) (Nolte 
and colleagues, 1994; Thorlacius and col- 
leagues, 1998; Jaeschke, 1998; Weiser and col- 
leagues, 1996; Farhood and colleagues, 1995) . 
The endothelium now has been recognized 
not just to be a lining of the vascular conduit, 
but also to play a key role in the multistep 
process of leukocyte accumulation and emi- 
gration (Ikeda and colleagues, 2000). More- 
over, soluble mediators, which are released 
after reperfusion, such as proinflammatory 
cytokines (tumor necrosis factor-a [TNF-a], 
interleukins, platelet-activating factor [PAF] ) , 
and leukotrienes, contribute to postischemic 
endothelial edema formation and perfusion 
dysfunction (Lefer, 1999; Jarrar and col- 
leagues, 1999; Wang and colleagues, 1995; 
Jarras and colleagues, 2001; Linden, 2001). 
Moreover, radicals generated during reper- 
fusion with oxygen-rich blood are factors that 
cause membrane damage and microvascular 
dysfunction. 



The depletion of energy-rich phosphates 
such as adenosine triphosphate (ATP) dimin- 
ishes the ability of the endothelial cell to main- 
tain a transmembrane gradient of cations and 
anions as during normal homeostasis and 
leads to cell swelling and impairs cell integrity, 
causing extravasation of macromolecules, the 
so-called leakage (Wang and colleagues, 1999; 
Chaudry, 1983; Chaudry, 1990; Wang and col- 
leagues, 1995; Wang and colleagues, 1994; 
Clemens and colleagues, 1985; Chaudry, 
1989). The bioavailability of nitric oxide 
(NO) under those conditions is also markedly 
reduced (Ikeda and colleagues, 2000; Kim and 
Hwan, 2001; Hierholzer and colleagues, 2001; 
Uhlmann and colleagues, 2000; Traber, 2000) . 
NO plays an important role in maintaining 
vascular tone and has antiadhesive properties 
(Wang and colleagues, 1995; Carden and 
Granger, 2000; Zhou and colleagues, 1997, 
Wang and colleagues, 1995; Wang and col- 
leagues, 1994) . With the advances in molec- 
ular biology techniques and knowledge, it has 
been shown that the proinflammatory milieu 
and generation of oxygen radicals trigger the 
activation of intracellular signaling pathways, 
leading to translocation of nuclear transcrip- 
tion factors and induction of stress genes and 
de novo protein synthesis (Okubo and col- 
leagues, 2000; McDonald and colleagues, 
2001; Jarrar and colleagues, 2000; Massberg 
and colleagues). 

The key role of leukocytes in the manifesta- 
tion of I/R injury has been documented using 
anti-adhesion molecule strategies, for example, 
monoclonal antibodies or antineutrophil 
serum. Through polymorphonuclear (PMN) 
leukocytes accumulation and leukocyte-capil- 
lary plugging, reperfusion is further impaired, 
enhancing the vicious cycle of no- reflow under 
those conditions (Massberg and Messmer, 
1998; Lefer, 1999; Schlag and colleagues, 2001; 
Yamaguchi and colleagues, 1999). 

The gut has been proposed as the motor 
for initiating multiorgan dysfunction follow- 
ing trauma and I/R. Although endotoxin and 
bacteria translocation may play a role in induc- 
ing cell and organ dysfunction following I/R, 
mediators that are released to the portal blood 
or lymph can activate neutrophils and Kupffer 
cells to release proinflammatory mediators, 
causing organ dysfunction. Characterization 
of the role of gut-derived mediators and/or 



ch02.qxd 4/16/04 3:19 PM Page 76 



76 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



factors during I/Rwill provide further insight 
into the mechanism responsible for cell and 
organ dysfunction following I/R. 



ROLE OF ENDOTHELIAL CELLS 
FOLLOWING ISCHEMIA- 
REPERFUSION 



Unstressed endothelial cells express a distinct 
set of genes that produce a nonthrombogenic 
lining of the blood vessels (Linden, 2001 ; Boyle 
and colleagues, 1999) . This minimizes inter- 
action between the endothelial lining and cir- 
culating blood cells and platelets. Moreover, 
during homeostasis, antithrombotic and pro- 
coagulatory mechanisms are balanced. This 
includes the production of thrombomodulin 
and vasoactive molecules such as NO and 
prostacyclin, which promote vasodilation and 
inhibition of smooth muscle cell contraction 
(Boyle and colleagues, 1999; Massberg and 
colleagues, 1999; Lefer and Lefer, 1993). Fol- 
lowing I/R, genes favoring an inflammatory 
milieu are preferentially induced. This leads 
to the increased production of E-selectin, 
ICAM, VCAM, and in terleukin-8 (IL-8) , which 
promote leukocyte rolling, aggregation, sub- 
sequent adhesion and trans-endothelial cell 
migration (Massberg and Messmer, 1998; 
Schlag and colleagues, 2001; Massberg and 
colleagues, 1999; Boyle and colleagues, 1999) . 
Moreover, the production of the vasoactive 
molecule NO by constitutive NO synthetase 
(NOS) is diminished, whereas superoxide pro- 
duction is increased. Experimental data have 
shown that following I/R, endothelium- 
dependent vasodilation in arterioles is 
reduced because NO is not available in 
sufficient quantities to serve as a second mes- 
senger in response to endogenous vasodila- 
tors such as acetylcholine (Farhood and 
colleagues, 1995; Lefer and Leafer, 1993). 
Arteriolar smooth muscle cell responsiveness, 
however, is maintained under those condi- 
tions. At a molecular level, studies have shown 
that the transcription factor NF-kB plays a key 
in the phenotypic changes of the endothelial 
cell lining toward an inflammatory phenotype. 
NF-kB is activated by oxidative stress, and upon 
degradation of its inhibitory molecule IkBoc, 
NF-kB is translocated to the nucleus where it 



binds to specific deoxyribonucleic acid (DNA) 
binding sequences, commonly the promoter 
region of inflammatory proteins and adhe- 
sion molecules, thereby increasing their rate 
of gene transcription. This includes E-selectin, 
VCAM, ICAM, IL-8, TNF-a, and the inter- 
leukins. Moreover, PAF receptor, tissue factor, 
and plasminogen activator are regulated by 
NF-kB and are induced following I/R, leading 
to microthrombosis, reduced blood flow, and 
leukocyte activation (McDonald and col- 
leagues, 2001). 



LEUKOCYTE-ENDOTHELIAL 
CELL INTERACTION 
FOLLOWING ISCHEMIA- 
REPERFUSION 



Leukocyte trafficking through the microcir- 
culation of tissues is essential for immune sur- 
veillance of tissues and early detection of 
pathologic conditions. Leukocyte recruit- 
ment is tightly regulated not only by the neu- 
trophils, but also by the endothelial cell and 
adjacent tissue cells such as monocytes and 
mast cells. Several distinct steps regulate the 
recruitment of leukocytes into the extravas- 
cular space following inflammatory stimuli as 
observed after I/R (Fig. 2-1). The first step 
in this process is the rolling of leukocytes 
along the microvascular endothelium. Under 
normal flow conditions, leukocytes travel 
along an axial stream, whereas rolling allows 
contact of the blood cells with the endothe- 
lium. P-selectin, an adhesion glycoprotein, pri- 
marily regulates this process. In the second 
step, which is mediated via the expression of 
CD11/CD18, a member of the (3 2 -integrins, 
on the surface of leukocytes and ICAM-1, 
a member of the immunoglobulin super- 
family, on the apical site of endothelial cells, 
the rolling leukocyte adheres firmly to the 
endothelium (Massberg and colleagues, 1998; 
Becker and colleagues, 1994; Menger and col- 
leagues, 1994; Menger and colleagues, 1997; 
Pickelmann and colleagues, 1998; Steinbauer 
and colleagues, 1998; Kaeffer and colleagues, 
1997). Finally, transendothelial migration 
occurs, requiring PECAM-1. The primary 
target sites for the aforementioned process 



ch02.qxd 4/16/04 3:19 PM Page 77 



Proinflammatory milieu: E-selectin, P-selectin, IL-6, IL-8 



2 • ISCHEMIA AND REPERFUSION INJURY 77 
Oxidative stress Procoagulant 



Free flowing 




C3a, C5aa, Histamine, 
Bradykinin, Serotonin 



■ FIGURE 2-1 

This figure shows the leukocyte-endothelium cell interactions following ischemia-reperfusion. Under 
normal conditions, the cellular components of the blood are separated from the endothelium by a 
rim of plasma. Following adverse circulatory conditions such as ischemia-reperfusion, the leukocyte 
gets in proximity to the endothelial wall by rolling along the wall. Up-regulation of adhesion 
molecules such as intercellular adhesion molecule-1 on the endothelial surface and the CD1 1/CD18 
complex on polymorphonuclear (PMN) leukocytes then promotes sticking of the leukocytes to the 
venules. Subsequently, transendothelial migration of PMN is enhanced by tissue mast cells and the 
release of inflammatory meditators. Together with activation of platelets, this leads to the no-reflow 
phenomenon in postcapillary venules following ischemia-reperfusion. ■ 



are the postcapillary venules. This multistep 
process is markedly enhanced by local oxida- 
tive stress. Although initially the endothelium 
via xanthine oxidase serves as a production 
site of superoxide and hydrogen peroxide, the 
adherent leukocyte then amplifies this event 
and accounts for the substantially greater 
amount of radicals produced. Reperfusion 
and reintroduction of molecular oxygen add 
significantly to oxidative stress in the post- 
capillary venules. Furthermore, this results in 
an imbalance in the production of NO and 
superoxide, accounting for microvascular 
dysfunction and the no-reflow phenomenon. 
The lack of sufficient NO by the endothelial 
NOS leads to the unavailability of NO to serve 
as a second messenger and to effectively 
scavenge superoxide. Studies by Massberg 
and Messmer (1998) have demonstrated that 



leukocyte-endothelial cell interaction pre- 
cedes capillary perfusion failure and is the 
primary step responsible for the pathophysi- 
ologic sequelae following I/R (Fig. 2-2). 



ROLE OF PLATELETS AND 
COMPLEMENT SYSTEM 

Other than PMN cells and the endothelium, 
other blood components such as the anuclear 
platelets play an important role in I/R injury. 
Platelets are a source of oxygen radicals, 
release inflammatory mediators including 
thromboxane A 2 , leukotrienes, serotonin, 
and platelet factor-4. Recruitment of platelets 
early following I/R to the postischemic vas- 
culature leads to luminal narrowing and local 



ch02.qxd 4/16/04 3:19 PM Page 78 



78 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



-| Ischemia/Reperfusion [ 



Leukocytes | 



NF-kB, MAPK 



Calpain/HSP/Antisense 
oligonucleotides 



CD11/CD18 



Endothelium 



| Inflammatory milieu 



NF-kB, MAPK 



Adhesion molecules: 
ICAM-1, ICAM-2 



] P-selectin 



I 



Anti-selectin Ab , 



\ Decreased ATP 



L 



ATP-MgCl2 , 



^Leukotrienes 



I 



NF-kB, MAPK 



TNF-a, IL-6, Reactive oxygen species 



-^Scavengers, Receptor antagonists 



Decreased nitric oxide 



L-Arginine/Allopurinol r 



■ Increased superoxide 



^ Platelet-activating factor receptor 



| Tissue/plasminogen factor 



] Activation of complement system | 



Prostaglandin E1 , 



-i Calcium blocker: Diltiazem . 



m FIGURE 2-2 

This schema shows the pathophysiologic changes occurring following ischemia-reperfusion and 
possible therapeutic interventions. Activation of intracellular stress signaling pathways are the key 
event on a molecular level. The inflammatory milieu then results in a vicious cycle on the level of the 
microcirculation. The potential therapeutic strategies are highlighted in dashed boxes. ■ 



thrombosis. Concomitantly, fibrinogen is 
deposited at the endothelium, which displays 
a procoagulant phenotype under those 
conditions (Massberg and colleagues, 1997; 
Massberg and colleagues, 1999). This is 
dependent on the expression of ICAM-1. 



DIVERGENT EFFECTS OF 
NITRIC OXIDE AND 
SUPEROXIDE FOLLOWING 
ISCHEMIA-REPERFUSION 

NO plays a critical role in I/R injury. As men- 
tioned earlier in this chapter, this is partially 
due to the role of NO to scavenge superox- 
ide, which is produced by xanthine oxidase. 
The substrate of xanthine oxidase, hypoxan- 
thine, accumulates as the ATP stores are 
depleted. The imbalance between reduced 
availability of NO and the enhanced produc- 
tion of toxic radicals increases leukocyte 



accumulation and impairs microvascular per- 
fusion. The role of xanthine oxidase as a con- 
tributor to I/R injury has been demonstrated 
using inhibitors of this enzyme such as allop- 
urinol. Experimental data support the notion 
that xanthine oxidase-derived oxidants act as 
a chemoattractant that regulates leukocyte 
trafficking in the microvasculature. The 
studies of Suzuki and colleagues (1991) have 
shown that exposure of postcapillary venules 
to oxidative stress increases leukocyte accu- 
mulation. The precise contribution of NO to 
I/R injury, however, is complicated because 
depending on the enzymatic source, NO may 
have beneficial or deleterious effects on tissue 
perfusion. Both constitutive and inducible 
forms of the enzyme NOS accountfor the pro- 
duction of NO. Constitutive Ca 2+ -dependent 
production by constitutive NOS (cNOS) 
(endothelial and neuronal NOS) is present 
before the injury and is thought to be 
beneficial. The Ca 2+ -independent, inducible 
isoform (inducible NOS [iNOS], NOS-2) 



ch02.qxd 4/16/04 3:19 PM Page 79 



2 • ISCHEMIA AND REPERFUSION INJURY 



79 



accounts for the detrimental effects of NO, 
including the production of peroxynitrate. 
Insight into the complicated role of NOS fol- 
lowing I/R has been gained using mice with 
targeted disruption of the iNOS (NOS-2) 
isoform (Kozlov and colleagues, 2001). 
Deficiency in iNOS-derived NO resulted in a 
significant reduction of skeletal muscle necro- 
sis following hind-limb ischemia and reper- 
fusion. Moreover, L-arginine supplementation 
during I/R prevents microvascular perfusion 
dysfunction by providing substrate for cNOS 
and maintaining tissue NO levels while con- 
comitantly decreasing superoxide production 
(Ikeda and colleagues, 2000; Uhlmann and 
colleagues, 2000; Traber, 2000; Carden and 
Granger, 2000; Suzuki and colleagues, 1991; 
Rahat and colleagues, 2001). These findings 
not only underscore the significance of NO 
in maintaining microvascular integrity and 
perfusion under normal and pathologic con- 
ditions but also imply that L-arginine may be 
a useful therapeutic agent under those 
conditions. 

CONFOUNDING 
COMORBIDITIES FOLLOWING 
ISCHEMIA-REPERFUSION 

Although most trauma patients are in the 
younger population with no prior medical 
history, the consequences of I/R injury sec- 
ondary to vascular trauma and/or compro- 
mise require special attention in comorbid 
patients. Comorbidities such as diabetes niel- 
li tus, hypertension, and hypercholesterolemia 
are all associated with arteriosclerotic disease 
and preexisting microvascular compromise 
(Huk and colleagues, 2000; Tailor and Granger, 
2000; Hoshida and colleagues, 2000; Salas and 
colleagues, 1999; Bouchard and Lamontagne, 
1998; Panes and colleagues, 1996). It is very 
conceivable and proven experimentally using 
animal strains expressing the aforementioned 
diseases that these confounding factors 
further aggravate I/R injury following vascu- 
lar trauma and successful revascularization. 
This should be kept in mind when taking care 
of elderly patients, because the window for 
successful therapeutic interventions is even 
narrower under those conditions. 



SYSTEMIC LEVELS OF 
INFLAMMATORY MEDIATORS 
AND REMOTE ORGAN INJURY 

Following ischemia and reperfusion, local pro- 
duction and release of inflammatory media- 
tors such cytokines, oxygen radicals, and 
vasoactive peptides are markedly enhanced. 
As discussed earlier in this chapter, this local 
inflammatory milieu contributes to PMN 
and platelet adhesion and ultimately to the 
no-reflow phenomenon. However, following 
I/R, this is not contained to the affected organ 
or organ system and leads to significant 
increased levels of mediator in the systemic 
circulation. For example, even simple laparo- 
tomy increased TNF-a messenger ribonucleic 
acid (mRNA) production in lung tissues, 
which was even further enhanced after 
remote, that is, intestinal, I/R. Circulating 
levels of xanthine oxidase are markedly ele- 
vated following I/R and accounted for acti- 
vation of Kupffer cells and elevated liver 
enzyme release following hind-limb ischemia. 
Moreover, similar results were obtained 
by the administration of exogenous xanthine 
oxidase. From experimental and clinical data, 
it appears that the lungs are the most sus- 
ceptible organ to low-flow conditions includ- 
ing ischemic events. The acute respiratory 
distress syndrome (ARDS) is a threaded event 
following adverse circulatory conditions, with 
a high mortality once fully developed. Appar- 
ently, once the initial I/R insult has been severe 
enough to lead to the systemic inflammatory 
response syndrome, circulating neutrophils 
are activated, leading to leukocyte- 
endothelial cell interaction in multiple vas- 
cular beds. The radiologic hallmark of ARDS, 
bilateral infiltrates, are caused by leukocyte 
influx, interstitial edema, and alveolar wall 
thickening. The development of respiratory 
insufficiency usually begins within 24 to 72 
hours after the initiating ischemic event. As 
outlined earlier, the risk of developing ARDS 
corresponds to the length of the ischemic time 
and is markedly increased in the elderly 
comorbid patient. 



ch02.qxd 4/16/04 3:19 PM Page 80 



80 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



SIGNIFICANCE OF ISCHEMIC 
PRECONDITIONING 



Preconditioning refers to the phenomenon in 
which the exposure of cells, tissues, or organ 
systems to brief periods of ischemia protects 
them from the deleterious effects of sub- 
sequent prolonged ischemia. Although pre- 
conditioning, for obvious reasons, is not 
available in the acute setting of vascular 
trauma and because of the need for subse- 
quent immediate revascularization, it should 
be discussed because important insights into 
the mechanism ofl/R injury have been gained 
from this phenomenon. Preconditioning 
blunts the impairment of endothelium- 
dependent relaxation to acetylcholine, 
capillary plugging, leukocyte adhesion, and 
the no-reflow phenomenon usually observed 
following I/R. Among other mechanisms, a 
distinct set of genes the family of heat shock 
proteins (HSPs) confers protection against 
adverse circulatory events. HSPs maintain cel- 
lular survivability by preserving metabolic and 
structural integrity of cells. Recent data suggest 
that via the adenosine A[ receptor and acti- 
vation of protein kinase C, as well as tyrosine 
kinases, HSP27 is phosphorylated, thereby 
conferring protection against a subsequent 
lethal insult (Davies and Hagen, 1993; 
Speechly-Dick and colleagues, 1995) . Sum- 
marizing ongoing efforts, it appears that 
selective induction of the HSPs might become 
a therapeutic modality. 



MODIFYING FACTORS 
DETERMINING ISCHEMIA- 
REPERFUSION INJURY 

Several factors modify the consequences, that 
is, the extent of tissue injury following 
I/R. Ambient temperature is well known to 
modulate the extent of necrosis. These lessons 
are learned from transplantation of solid 
organs, a classic example of I/R injury. 
Decreasing the temperature of the storage 
solution to 4°C can accomplish later re- 
establishment of the venous and arterial 
blood flow with complete return of organ func- 



tion. In the case of extremity trauma with com- 
plete amputation, the recommendations are 
that the severed limb be stored in ice water 
until the patient is transported to an appro- 
priate center with expertise in vascular surgery. 



TREATMENT OF ISCHEMIA- 
REPERFUSION INJURY: ROLE 
OF PHARMACOLOGIC 
ADJUNCTS 

A key event during ischemia is the decrease 
in the cellular levels of energy-rich phosphates, 
such as ATP. In view of this, studies have used 
the approach of administrating ATP-MgCl 2 
after I/R, and such studies have shown that 
endothelial cell function can be restored fol- 
lowing adverse circulatory conditions (Wang 
and colleagues, 1999; Wang and colleagues, 
1995; Dana and colleagues, 2000; Gaudio and 
colleagues, 1982; Chaudry and colleagues, 
1983; Ohkawa and colleagues, 1983; Chaudry 
and colleagues, 1984) . Moreover, monoclonal 
antibodies against adhesion molecules such 
as CD 11 /CD 18 or the P-selectin family have 
also been beneficial in preventing I/R injury 
(Ohkawa and colleagues, 1984). 

Improvement of blood flow using rheologic 
agents such as dextran are also effective in 
improving capillary reflow following I/R 
(Sharar and colleagues, 1991; Schott and 
colleagues, 1998). Administration of pro- 
staglandin Ex following I/R normalized NO 
and superoxide release and thus improved 
microvascular blood flowwith reduced adher- 
ence of leukocytes (Berglund and colleagues, 
1981). 

Although all the aforementioned agents 
have been shown to reduce I/R injury in 
experimental models, they have not been used 
clinically to improve outcome in patients 
following vascular trauma and subsequent 
ischemic events. Only the intraoperative 
administration of heparin following vascular 
extremity injury has been shown to signifi- 
cantly improve the rate of limb salvage (Forrest 
and colleagues, 1991; Melton and colleagues, 
1997; Wang and colleagues, 1990; Rana and 
colleagues, 1992; Wang and colleagues, 1993; 
Wang and colleagues, 1994; Zellweger, 1995) . 



ch02.qxd 4/16/04 3:19 PM Page 81 



2 • ISCHEMIA AND REPERFUSION INJURY 



81 



SUMMARY 



This chapter has covered pathophysiology of 
ischemia and reperfusion injury, including the 
consequences of re-establishment of blood 
flow following a variable period of cessation 
of perfusion with oxygenated blood (Wang 
and colleagues, 1996; Wall and colleagues, 
1996) . Apparently, I/R injury is due not only 
to the lack of oxygen as seen during ischemia, 
but also to the absence of whole blood with 
its scavenging properties. Generally speaking, 
I/Rinjury is the most common cause of death 
in the Western Hemisphere because of the 
prevalence of arteriosclerotic disease includ- 
ing the coronary arteries (Shin and col- 
leagues, 2000; Beebe and colleagues, 1996; 
Brengman and colleagues, 2000; Eifert and 
colleagues, 2000). The key pathophysiologic 
events following I/R have been investigated 
in depth. In this regard, the leukocyte- 
endothelial cell interactions appear to be a 
key element, which results in the phenome- 
non of no reflow following adverse circula- 
tory conditions, secondary to enhanced PMN 
leukocyte recruitment to the postcapillary 
venules. Using intravital microscopy, it has 
been shown that an orderly sequence of events 
takes place including leukocyte rolling, stick- 
ing, and adherence, followed by transendothe- 
lial migration. These processes are modified 
by the inflammatory milieu generated fol- 
lowing I/R injury. Reactive oxygen species, 
proinflammatory cytokines, superoxide, and 
other paracrine messengers all intensify 
the leukocyte-endothelial cell interaction, 
leading to microvascular perfusion failure. 
Paradoxically, most of the damage occurs 
during the reperfusion period and to a lesser 
extent during the actual ischemic time. 

The probably most significant independent 
factor determining injury following vascular 
injury and subsequent ischemia of an organ 
or limb is time until re-establishment of blood 
flow. Modifying factors, however, include 
ambient temperature and preexisting comor- 
bidities. Although a variety of therapeutic 
agents have been effectively used in experi- 
mental studies, this has notyet been translated 
into an accepted modality in clinical practice. 
Of note, the decrease in hematocrit during 



vascular injury caused by blood loss has bene- 
ficial effects on the rheology of the microvas- 
culature but does mandate immediate blood 
transfusion. 

With the use of molecular biology tools, the 
changes on the cellular mRNA and protein 
level have also been characterized following 
I/R. A distinct set of stress response genes and 
signaling pathways appear activated, leading 
to a change in the phenotype of leukocytes 
and endothelial cells. This propagates the local 
inflammatory milieu, resulting in the up- 
regulation of adhesion molecules and the no 
reflow following ischemia and reperfusion. 
With the advances in our understanding of 
the molecular mechanisms of I/R injury, selec- 
tive modulation of the aforementioned phe- 
notypic changes in leukocytes and endothelial 
cells should be forthcoming. 



REFERENCES 

Barker JE, Knight KR, Romeo R, et al: Targeted 
disruption of the nitric oxide synthase 2 gene 
protects against ischemia/ reperfusion injury to 
skeletal muscle. J Pathol 2001;194:10-115. 

Becker M, Menger MD, Lehr HA: Heparin-re leased 
superoxide dismutase inhibits postischemic 
leukocyte adhesion to venular endothelium. Am 
J Physiol 1994;267:H925-H930. 

Beebe HG, Bergan JJ, Bergqvist D, et al: 
Classification and grading of chronic venous 
disease in the lower limbs. A consensus statement. 
EurJ Vase Endovasc Surg, 1996;12:487-492. 

Berglund B, Swanbeck G, Hedin H: Low molecu- 
lar weight dextran therapy for digital ischemia 
due to collagen vascular disease. Dermatologica 
1981;163:353-357. 

Bickell WH, Shaftan GW, Mattox KL: Intravenous 
fluid administration and uncontrolled hemor- 
rhage . J Trauma 1989;29:409. 

Bickell WH, Wall MJ Jr, Pepe PE, et al: Immediate 
versus delayed fluid resuscitation for hypoten- 
sive patients with penetrating torso injuries. N 
Engl Med 1994;331:1105-1109. 

Bouchard JF, Lamontagne D: Protection afforded 
by preconditioning to the diabetic heart against 
ischemic injury. Cardiovasc Res 1998;37:82- 
90. 

Boyle EM Jr, Canty TG Jr, Morgan EN, et al: Treat- 
ing myocardial ischemia-reperfusion injury by 



ch02.qxd 4/16/04 3:19 PM Page 82 



82 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



targeting endothelial cell transcription. Ann 
Thorac Surg 1999;68:1949-1953. 

Brengman ML, O'Donnell SD, Mullenix P, et al: 
The fate of a patent carotid artery contralateral 
to an occlusion. Ann Vase Surg 2000;14:77-81. 

Burch JM, Richardson RJ, Martin RR, Mattox KL: 
Penetrating iliac vascular injuries: recent expe- 
rience with 233 consecutive patients. J Trauma 
1990;30:1450-1459. 

Carden DL, Granger DN: Pathophysiology of 
ischemia-reperfusion injury. J Pathol 2000;190: 
255-266. 

Chaudry IH: Cellular mechanisms in shock and 
ischemia and their correction. Am J Physiol 
1983;245:R117-R134. 

Chaudry IH: ATP-MgCL and liver blood flow fol- 
lowing shock and ischemia. Prog Clin Biol Res 
1989;299:19-31. 

Chaudry IH: The use of ATP following shock and 
ischemia. Ann N Y Acad Sci 1990;603: 
130-141. 

Chaudry IH, Ohkawa M, Clemens MG: Improved 
mitochondrial function following ischemia and 
reflow by ATP-MgCl,. Am J Physiol 1984;246: 
R799-R804. 

Chaudry IH, Ohkawa M, Clemens MG, Baue AE: 
Alterations in electron transport and cellular 
metabolism with shock and trauma. Prog Clin 
Biol Res 1983;111:67-88. 

Clemens MG, McDonah PF, Chaudry IH, Baue AE: 
Hepatic microcirculatory failure after ischemia 
and reperfusion: Improvement with ATP-MgCL 
treatment. AmJPhysiol 1985;248:H804-H811. 

Dana A, Skarli M, Papakrivopoulou J, Yellon DM: 
Adenosine A(l) receptor induced delayed 
preconditioning in rabbits: Induction of p38 
mitogen-activated protein kinase activation and 
Hsp27 phosphorylation via a tyrosine kinase — 
and protein kinase C-dependent mechanism. 
Circ Res 2000;86:989-997. 

Davies MG, Hagen PO: The vascular endothelium. 
A new horizon. Ann Surg 1993;218:593-609. 

de Guzman E, Shankar MN, Mattox KL: Limited 
volume resuscitation in penetrating thoracoab- 
dominal trauma. AACN Clin Issues 1999;10:61- 
68. 

Eifert S, VillavicencioJL, Kao TC, et al: Prevalence 
of deep venous anomalies in congenital vascu- 
lar malformations of venous predominance. 
J Vase Surg 2000;31 :462-471 . 

Farhood A, McGuire GM, Manning AM, et al: 
Intercellular adhesion molecule 1 (ICAM-1) 
expression and its role in neutrophil-induced 



ischemia-reperfusion injury in rat liver. J Leukoc 
Biol 1995;57:368-374. 

Forrest CR, Pang CY, Zhong AG, Kreidstein ML: 
Efficacy of intravenous infusion of prostacyclin 
(PG12) or prostaglandin Ei (PGE]) in augmen- 
tation of skin flap blood flow and viability in the 
pig. Prostaglandins 1991;41:537-558. 

Gaudio KM, Taylor MR, Chaudry IH, et al: Accel- 
erated recovery of single nephron function by 
the postischemic infusion of ATP-MgCL. Kidney 
Int 1982;22:13-20. 

Geller DA, Chia SH, Takahashi Y, et al: Protective 
role of the L-arginine-nitric oxide synthase 
pathway on preservation injury after rat liver 
transplantation. J Patenter Enter Nutr 2001; 
25:142-147. 

Hierholzer C, Harbrecht BG, Billiar TR, Tweardy 
DJ: Hypoxia-inducible factor-1 activation 
and cyclo-oxygenase-2 induction are early 
reperfusion-independent inflammatory events 
in hemorrhagic shock. Arch Orthop Trauma 
Surg 2001;121:219-222. 

Hoshida S, Yamashita N, Otsu K, et al: Cholesterol 
feeding exacerbates myocardial injury in Zucker 
diabe tic fatty rats. Am J Physiol Heart Circ Physiol 
2000;278:H256-H262. 

Huk I, Brovkovych V, Nanobashvili J, et al: 
Prostaglandin E] reduces ischemia/reperfusion 
injury by normalizing nitric oxide and super- 
oxide release. Shock 2000;14:234-242. 

Ikeda Y, Young LH, Scalia R, Lefer AM: Cardio- 
protective effects of citrulline in ischemia/ 
reperfusion injury via a non-nitric oxide- 
mediated mechanism. Methods Find Exp Clin 
Pharmacol 2000;22:563-571. 

Jaeschke H: Mechanisms of reperfusion injury after 
warm ischemia of the liver. J Hepatobiliary 
Pancreat Surg 1998;5:402-408. 

Jarrar D, Chaudry IH, Wang P: Organ dysfunction 
following hemorrhage and sepsis: Mechanisms 
and therapeutic approaches [Review] . Int J Mol 
Med 1999;4:575-583. 

Jarrar D, Wang P, Chaudry IH: Hepatocellular 
dysfunction: Basic considerations. In Holzheimer 
RG, Mannick JA (eds): Surgical Treatment: 
Evidence-Based and Problem-Oriented. Munich, 
Germany, Zuckschwerdt Verglad, 2001 : 763-767. 

Jarrar D, Wang P, Song GY, et al: Inhibition of tyro- 
sine kinase signaling after trauma-hemorrhage: 
A novel approach for improving organ function 
and decreasing susceptibility to subsequent 
sepsis. Ann Surg 2000;231:399-407. 



ch02.qxd 4/16/04 3:19 PM Page 83 



2 • ISCHEMIA AND REPERFUSION INJURY 



83 



Kadambi A, Shalak TC: Role of leukocytes and 
tissue-derived oxidants in short-term skeletal 
muscle ischemia-reperfusion injury. Am J Physiol 
Heart Circ Physiol 2000;278:H435-H443. 

Kaeffer N, Richard V, Thuillez C: Delayed 
coronary endothelial protection 24 hours after 
preconditioning: Role of free radicals. Circula- 
tion 1997;96:2311-2316. 

Kim H, Hwan KK: Role of nitric oxide and mucus 
in ischemia/ reperfusion-induced gastric muco- 
sal injury in rats. Pharmacology 2001;62:200-207. 

Kozlov AV, Sobhian B, Duvigneau C, et al: Organ 
specific formation of nitrosyl complexes under 
intestinal ischemia/reperfusion in rats involves 
NOS-independent mechanism (s). Shock 2001; 
15:366-371. 

Lefer AM: Endotoxin, cytokines, and nitric oxide 
in shock. Shock 1994;1:79-80. 

Lefer AM: Role of the beta2-integrins and 
immunoglobulin superfamily members in 
myocardial ischemia-reperfusion. Ann Thorac 
Surg 1999;68:1920-1923. 

Lefer AM, Lefer DJ: Pharmacology of the endothe- 
lium in ischemia-reperfusion and circulatory 
shock. ArmRev Pharmacol Toxicol 1993;33:71-90. 

Lehr HA, Guhlman A, Nolte D, et al: Leukotrienes 
as mediators in ischemic-reperfusion injury in 
a microcirculation model in the hamster. J Clin 
Invest 1991;87:2036-2041. 

Linden J: Molecular approach to adenosine recep- 
tors: Receptor-mediated mechanisms of tissue 
protection. Ann Rev Pharmacol Toxicol 2001; 
41:775-787. 

Massberg S, Enders G, Matos FC, et al: Fibrinogen 
deposition at the postischemic vessel wall pro- 
motes platelet adhesion during ischemia-reper- 
fusion in vivo. Blood 1999;94:3829-3838. 

Massberg S, Messmer K: The nature of ischemia/ 
reperfusion injury. Transplant Proc 1998;30: 
4217-4223. 

Massberg S, Sausbier M, Klatt P, et al: Increased 
adhesion and aggregation of platelets lacking 
cyclic guanosine 3',5'-monophosphate kinase I. 
J Exp Med 1999;189:1255-1264. 

McDonald MC, Mota-Filipe H, Paul A, et al: 
Calpain inhibitor I reduces the activation of 
nuclear factor-kappaB and organ injury/ 
dysfunction in hemorrhagic shock. FASEB J 
2001;15:171-186. 

Melton SM, Croce MA, Patton JHJr, et al: Popliteal 
artery trauma. Systemic anticoagulation and 
intraoperative thrombolysis improves limb 
salvage. Ann Surg 1997;225:518-527. 



Menger MD, Kerger H, Geisweid A, et al: 
Leukocyte-endothelium interaction in the 
microvasculature of postischemic striated 
muscle. Adv Exp Med Biol 1994;361:541-545. 

Menger MD, Pelikan S, Steiner D, Messmer K; 
Microvascular ischemia-reperfusion injury in stri- 
ated muscle: significance of reflow paradox. Am 
J Physiol 1992;263:H1901-H1906. 

Menger MD, Rucker M, Vollmar B: Capillary dys- 
function in striated muscle ischemia/ 
reperfusion: on the mechanisms of capillary 
no-reflow. Shock 1997;8:2-7. 

Menger MD, Volmmar B: In vivo analysis of 
microvascular reperfusion injury in striated 
muscle and skin. Microsurgery 1994;15:383-389. 

Nolte D, Hecht R, Schmid P, et al: Role of Mac-1 
and ICAM-1 in ischemia-reperfusion injury in a 
microcirculation model of BALB/C mice. Am J 
Physiol 1994;267:H1320-H1328. 

Nolte D, Lehr HA, Messmer K: Adenosine inhibits 
postischemic leukocyte-endothelium interac- 
tion in postcapillary venules of the hamster. Am 
J Physiol 1991;261:H651-H655. 

Ohkawa M, Chaudry IH, Clemens MG, Baue AE: 
ATP-MgCL produces sustained improvement in 
hepatic mitochondrial function and blood flow 
after hepatic ischemia. J Surg Res 1984;37:226- 
234. 

Ohkawa M, Clemens MG, Chaudry IH: Studies on 
the mechanism of beneficial effects of ATP-MgCL 
following hepatic ischemia. Am J Physiol 1983; 
244:R695-R702. 

Okubo S, Bernardo NL, Elliott GT, et al: Tyrosine 
kinase signaling in action potential shortening 
and expression of HSP72 in late precondition- 
ing. Am J Physiol Heart Circ Physiol 2000;279: 
H2269-H2276. 

PanesJ, Kurose I, Rodriguez-Vaca D, et al: Diabetes 
exacerbates inflammatory responses to ischemia- 
reperfusion. Circulation 1996;93:161-167. 

Pickelmann S, Nolte D, Leiderer R, et al: Attenu- 
ation of postischemic reperfusion injury in stri- 
ated skin muscle by diaspirin-cross-linked 
Hb. Am J Physiol 1998;275:H361-H368. 

Rahat MA, Lahat N, Smollar J, et al: Divergent 
effects of ischemia/reperfusion and nitric oxide 
donor on TNF-alpha mRNA accumulation in rat 
organs. Shock 2001;15:312-317. 

RanaMW, Singh G, WangP, etal: Protective effects 
of preheparinization on the microvasculature 
during and after hemorrhagic shock. J Trauma 
1992;32:420-426. 

Salas A, PanesJ, Rosenbloom CL, et al: Differen- 
tial effects of a nitric oxide donor on 



ch02.qxd 4/16/04 3:19 PM Page 84 



84 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



reperfusion-induced microvascular dysfunction 
in diabetic and non-diabetic rats. Diabetologia 
1999;42:1350-1358. 

Schlag MG, Harris KA, Potter RF: Role of leuko- 
cyte accumulation and oxygen radicals in 
ischemia-reperfusion-induced injury in skeletal 
muscle. Am J Physiol Heart Circ Physiol 2001; 
280:H1716-H1721. 

Schott U, Lindbom LO, Sjostrand U: Hemody- 
namic effects of colloid concentration in exper- 
imental hemorrhage: A comparison of Ringer's 
acetate, 3% dextran-60, and 6% dextran-70. Crit 
Care Med 1998;16:346-352. 

Sharar SR, Winn RK, Murry CE, e t al: A CD1 8 mon- 
oclonal antibody increases the incidence and 
severity of subcutaneous abscess formation after 
high-dose Staphylococcus aureus injection in 
rabbits. Surgery 1991;110:213-220. 

Shin DD, Wall MJ Jr, Mattox KL: Combined pene- 
trating injury of the innominate artery, left 
common carotid artery, trachea, and esophagus. 
J Trauma 2000;49:780-783. 

Speechly-Dick ME, Grover GJ, Yellon DM: Does 
ischemic preconditioning in the human involve 
protein kinase C and the ATP-dependent K + 
channel? Studies of contractile function after 
simulated ischemia in an atrial in vitro model. 
Circ Res 1995;77:1030-1035. 

Steinbauer M, Harris AG, Leiderer R, et al: 
Impact of dextran on microvascular distur- 
bances and tissue injury following ischemia/ 
reperfusion in striated muscle. Shock 1998;9: 
345-351. 

Tailor A, Granger DN: Role of adhesion molecules 
in vascular regulation and damage. Curr Hyper- 
tens Rep 2000;2:78-83. 

Thorlacius H, Vollmar B, Westermann S, et al: 
Effects of local cooling on microvascular hemo- 
dynamics and leukocyte adhesion in the striated 
muscle of hamsters. J Trauma 1998;45:715- 
719. 

Traber DL: Nitric oxide synthase and tissue injury. 
Shock 2000;14:243-244. 

Uhlmann D, Uhlmann S, Spiegel HU: Endothe- 
lin/nitric oxide balance influences hepatic 
ischemia-reperfusion injury. J Cardiovasc Phar- 
macol 2000;36:S212-S214. 

Wall MJ Jr, Granchi T, Liscum K, Mattox KL: Pen- 
etrating thoracic vascular injuries. Surg Clin 
North Am 1996;76:749-761. 

Wang P, Ba ZF, Chaudry IH: Endothelial cell dys- 
function occurs after hemorrhage in nonhep- 
arinized bvit not in preheparinized models. J Surg 
Res 1993;54:499-506. 



Wang P, Ba ZF, Chaudry IH: Chemically modified 
heparin improves hepatocellular function, 
cardiac output, and microcirculation after 
trauma-hemorrhage and resuscitation. Surgery 
1994;116:169-175. 

Wang P, Ba ZF, Chaudry IH: Nitric oxide. To block 
or enhance its production during sepsis? Arch 
Surg 1994;129:1137-1142. 

Wang P, Ba ZF, Chaudry IH: ATP-MgCL restores 
depressed endothelial cell function after hem- 
orrhagic shock and resuscitation. Am J Physiol 
1995;268:H1390-H1396. 

Wang P, Ba ZF, Chaudry IH: Endothelium- 
dependent relaxation is depressed at the macro- 
and microcirculatory levels during sepsis. 
Am J Physiol 1995;269:R988-R994. 

Wang P, Ba ZF, Reich SS, et al: Effects of nonanti- 
coagulant heparin on cardiovascular and hepa- 
tocellular function after hemorrhagic shock. Am 
J Physiol 1996;270:HI294-HI302. 

Wang P, Ba ZF, Stepp KJ, Chaudry IH: Pentoxify- 
line attenuates the depressed endothelial cell 
function and vascular muscle contractility fol- 
lowing trauma and hemorrhagic shock. J Trauma 
1995;39:121-126. 

Wang P, Singh G, Rana MW, et al: Prehepariniza- 
tion improves organ function after hemorrhage 
and resuscitation. Am J Physiol 1990;259:R645- 
R650. 

Wang P, Tait SM, Ba ZF, Chaudry IH: ATP-MgCL 
administration normalizes macrophage cAMP 
and beta-adrenergic receptors after hemorrhage 
and resuscitation. Am J Physiol 1994;267:G52- 
G58. 

Waxman K: Shock: ischemia, reperfusion, and 
inflammation. New Horiz 1996;4:153-160. 

Weiser MR, Gibbs SA, Valeri CR, et al: An ti-se lectin 
therapy modifies skeletal muscle ischemia and 
reperfusion injury. Shock 1996;5:402-407. 

Yamaguchi Y, Matsumura F, Liang J, et al: Neu- 
trophil elastase and oxygen radicals enhance 
monocyte chemoattractant protein-expression 
after ischemia/ reperfusion in rat liver. Trans- 
plantation 1999;68:1459-1468. 

Zellweger R, Ayala A, Zhu XL, e t al: A novel nonan- 
ticoagulant heparin improves splenocyte and 
peritoneal macrophage immune function after 
trauma-hemorrhage and resuscitation.J Surg Res 
1995;59:211-218. 

Zhou M, Wang P, Chaudry IH: Endothelial nitric 
oxide synthase is downregulated during hyper- 
dynamic sepsis. Biochim Biophys Acta 1997; 
1335:182-190. 



ch03.qxd 4/16/04 3:21PM Page 85 




Minimal Vascular Injuries 



JAMES W. DENNIS 



O DEFINITION 

O HISTORY OF THE MANAGEMENT OF MINIMAL INJURIES 

O DEFINING THE NATURAL HISTORY OF MINIMAL INJURIES 

O APPLICATION TO PENETRATING PROXIMITY EXTREMITY TRAUMA 

O APPLICATION TO PENETRATING NECK INJURIES 

O APPLICATION TO HIGH-RISK ORTHOPEDIC INJURIES 

O SUMMARY 



Arterial injuries come in various 
shapes and sizes. Regardless of the 
etiology, complete transections, 
occlusions, bleeding lacerations, and large 
pseudoaneurysms almost universally require 
immediate surgical intervention or the patient 
faces the loss of life or limb. These types of 
arterial injuries make up the vast majority 
(80% to 90%) of cases (Hardy and colleagues, 
1975) . Since the earliest times, surgeons have 
recognized that the type of injury can have a 
profound effect on the ultimate outcome. A 
special class of injuries has been recognized 
over the past 15 years that can be called 
"minimal injuries." This small select group 
appears to have a unique natural history and 



must be considered in this light. By under- 
standing this natural history, management 
plans for patients presenting with certain types 
of injuries can be formulated so that they 
ensure proper and safe treatment in the acute 
setting. 



DEFINITION 

Minimal injuries are generally defined as 
identifiable damage to a blood vessel, usually 
by arteriography or ultrasound, with no clin- 
ical signs of that injury. By definition, hard 
signs of vascular trauma including pulse 

85 



ch03.qxd 4/16/04 3:21PM Page 86 



86 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 




■ FIGURE 3-1 

A, Short segmental narrowing and intimal irregularity after a gunshot wound to the right brachial 
artery. B, Complete resolution of injury 6 weeks later. (From Dennis JW, Frykberg ER, Crump JM, et 
al: New perspectives on the management of penetrating trauma in proximity to major limb arteries. J 
Vase Surg 1990;11:84-93.) ■ 



deficit, active hemorrhage, expanding 
hematoma, distal ischemia, and bruit or thrill 
are absent. Soft signs such as a history of bleed- 
ing, stable hematoma, associated nerve deficit, 
or unexplained hypotension may or may not 
be present and have no direct relationship to 
these injuries. Minimal injuries will also reg- 
ularly demonstrate prograde flow of contrast 
on arteriography or flow on ultrasound. In 
addition, no gross, uncontained extravasation 
of blood or contrast is seen outside the 
normal lumen of the vessel involved. 

Studies have identified four basic types of 
minimal injuries, of which two dominate. The 
first major type is focal segmental narrowing 
or constriction that is characteristically smooth 
in nature with tapering at both ends (Figs. 3- 
1A and 3-2A). Arteries can demonstrate this 
abnormality secondary to external compres- 
sion, intramural hematoma (contusion), or 



reactive spasm. Spasm is due to the myogenic 
response of blood vessels to the blast effect of 
penetrating missiles or direct effects of blunt 
forces. Blood flow can be demonstrated 
throughout this narrowed segment, which can 
vary in length from just a few millimeters to 
several centimeters. 

The second predominant type of minimal 
injury is that of the intimal flap or irregular- 
ity (Fig. 3-3A) . This is usually seen as a luminal 
surface abnormality in which the intimal layer 
has a raised portion extending into the lumen. 
Flaps may be lifted in either a proximal or a 
distal orientation. It may also appear as a focal, 
roughened area of the luminal surface. In both 
forms, flow is present within the lumen and 
there is no extravasation outside of it. 

Small pseudoaneurysms and arteriovenous 
(AV) fistulas make up the other two 
smaller categories of minimal injuries. 



ch03.qxd 4/16/04 3:21PM Page 87 



3 • MINIMAL VASCULAR INJURIES 



87 




■ FIGURE 3-2 

A, Long smooth area of narrowing of the left brachial artery after a gunshot wound. B, Complete 
resolution after 1 week. (From Dennis JW, Frykberg ER, Crump JM, et al: New perspectives on 
the management of penetrating trauma in proximity to major limb arteries. J Vase Surg 1 990; 
11:84-93.) ■ 



Pseudoaneurysms areformed when there is an 
incomplete laceration of an artery with the 
resultant hemorrhage contained by the sur- 
rounding tissue (Fig. 3-4A) . These lesions are 
easily seen by arteriography or ultrasound and 
will appear as contained extravasation of con- 
trast or blood outside the normal arterial 
lumen. AVfistulas develop when an arterial lac- 
eration and simultaneous laceration or tear 
occurs in the adjacentvein causing flow to enter 
into the low-pressure venous channel. This is 
also clearly identified by arteriography or ultra- 
sound as a direct passage of contrast or blood 
from an artery into a vein without passing 
through a capillary system. 

In each of these minimal injuries, distal 
pulses usually remain intact. In patients with 
a pseudoaneurysm or AV fistula, an audible 
bruit or thrill may be present on physical exam- 
ination, indicating the need for further 



evaluation to determine the nature and extent 
of these injuries. 



HISTORY OF THE 
MANAGEMENT OF MINIMAL 
INJURIES 

Early information concerning vascular 
trauma was the result of military experience 
inWorldWarsIandll (DeBakeyandSimeone, 
1946). Direct vascular repair of injuries was 
not performed on a widespread basis, however, 
until the 1950s in both the Korean conflict 
and civilian settings (Hughes, 1958; Ferguson, 
Byrd, and McAfee, 1961). The decision to 
operate was initially based on physical exam- 
ination alone, as there was no other available 
or reliable means to diagnose vascular injuries. 



ch03.qxd 4/16/04 3:21PM Page 88 



88 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 




■ FIGURE 3-3 

A, Intimal irregularity or flap in the superficial femoral artery after a through-and-through gunshot 
wound of the right thigh. 6, Complete resolution of the injury after 1 week. (From Dennis JW, 
Frykberg ER, Crump JM, et al: New perspectives on the management of penetrating trauma in 
proximity to major limb arteries. J Vase Surg 1990;11:84-93.) ■ 



Penetrating wounds in proximity to major 
arteries began to be routinely explored to 
determine the presence or absence of a 
vascular injury. This policy continued into the 
1970s when arteriography began to be widely 
used to evaluate patients for vascular trauma 
and avoid unnecessary surgery. 

Multiple studies were published in the late 
1970s and early 1980s that showed arteriog- 
raphy to be as accurate as surgical exploration 
for detecting any type of vascular injury fol- 
lowing penetrating trauma to the extremities 
(Synder and colleagues, 1978; Sirinek and 
colleagues, 1981). These studies consistently 
showed a more than 95% chance of having a 
significant arterial injury when hard signs were 
present on physical examination. In addition, 
there was a 10% to 20% risk of an arterio- 
graphic abnormality found even in the face 
of normal physical examination results (Table 
3-1). This new use of arteriography first 
demonstrated the presence of these minimal 
arterial injuries that had not been seen before 



its use. Because no data existed about their 
clinical significance, surgeons erred on the 
side of operating on any abnormalities found. 
This approach was based on the fear of 
missing an injury that needed repair even in 
the absence of any clinical findings. Univer- 
sal recommendations resulting from these 
series were to either surgically explore or 
obtain an arteriogram on every patient with 
penetrating trauma to the extremities. If any 
abnormalities were seen on arteriography, 
they required immediate exploration and 
repair if needed. 



DEFINING THE NATURAL 
HISTORY OF MINIMAL 
INJURIES 



The first evidence that minimal vascular 
injuries might have the potential to 



ch03.qxd 4/16/04 3:21PM Page 89 



3 • MINIMAL VASCULAR INJURIES 



89 




■ FIGURE 3-4 

A, Small pseudoaneurysm of 
the left distal axillary artery 
after a gunshot wound to the 
shoulder. B, Pseudoaneurysm 
10 months later — essentially 
unchanged with possibly slight 
improvement. (From Dennis 
JW, Frykberg ER, Crump JM, et 
al: New perspectives on the 
management of penetrating 
trauma in proximity to major 
limb arteries. J Vase Surg 
1990;11:84-93.) ■ 



TABLE 3-1 








MECHANISM OF PENETRATING PROXIMITY EXTREMITY TRAUMA AND ULTIMATE 


OUTCOME 








Injury 


Total (No.) 


No. Arterial Injuries 


No. Requiring Surgery (%) 


Gunshot 


247 


24(9.7) 


2 (0.8)* 


Stab 


54 


5 (9.3) 


2(3.7) 


Shotgun 


17 


3(17.6) 


2(11.8)* 


Total 


318 


32(10.0) 


6(1.8) 



*One operated on immediately. 

From Dennis JW, Frykberg ER, Crump JM, et al: New perspectives on the management of penetrating trauma in proximity to 

major limb arteries. J Vase Surg 1990;11:84-93. 



ch03.qxd 4/16/04 3:21PM Page 90 



90 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



TABLE 3-2 

RESULTS OF NONOPERATIVE OBSERVATION OF 29 CLINICALLY OCCULT ARTERIAL 
INJURIES ACCORDING TO MORPHOLOGY 



Total No. 



Repeat Arteriogram (No.) 
RES IMP UNC 



WOR 



Clinical 
Follow-up (No.) 



UNC 



WOR 



Narrowing 


12 


7 





1 





4 





Intimal flap 


12 


6 


2 





1* 


2 


1 


Pseudoaneurysm 


5 


2 


1 





2* 








Total 


29 


15 


3 


1 


3 


6 


1 



'Underwent surgical repair. 

IMP, improved; RES, resolved; UNC, unchanged; WOR, worsened. 

From Dennis JW, Frykberg ER, Crump JM, et al: New perspectives on the management of penetrating trauma in proximity to 

major limb arteries. J Vase Surg 1990;11:84-93. 



spontaneously heal was work done by Glover 
in 1986. He induced intimal tears in rat arter- 
ies and harvested them for up to 1 year later. 
All vessels remained patent and the endothe- 
lial injury was consistently healed by 8 weeks 
with no long-term sequelae (Glover, 1986) . 
Clinical studies suggesting this might hold true 
in humans began to appear soon (Stain and 
colleagues, 1989; Kestenberg, 1990). These 
scientific studies began to look at the natural 
history of this unique class of injuries. The 
first prospective series on penetrating extrem- 
ity injuries was performed by Frykberg and 
colleagues in 1989. This landmark article 
detailed the potential healing properties of 
this new class of minimal injuries when 
followed with nonoperative management and 
serial arteriograms. This new approach 
revealed the natural history to be somewhat 
benign in that the vast majority (up to 89%) 
would either resolve spontaneously or remain 
unchanged. Approximately 11% would dete- 
riorate and require surgical repair, but this 
could be done safely and with no increase in 
morbidity or limb loss when performed on a 
delayed basis. Alarger series published almost 
2 years later confirmed the earlier results, by 
showing 87% of minimal injuries would 
heal if treated conservatively (Dennis and 
colleagues, 1990) (Table 3-2). Other trauma 
centers soon reported similar experiences 
when nonoperative management of these 
minimal injuries was employed (Francis and 
colleagues, 1991; Itani, 1991; Trooskin, 1993; 



Gahtan, 1994). The ability to resolve these 
minimal injuries in nonextremity arteries was 
also first demonstrated by Frykberg and 
colleagues (1991) in a series that included 
injuries of the torso and neck. 

The data consistently illustrated that dif- 
ferent types of minimal injuries behaved dif- 
ferently over time. Smooth arterial narrowings 
would almost uniformly resolve on their own 
and are the most benign of the minimal 
injuries (Figs. 3-l_B and 3-2-B). Intimal flaps 
or irregularities would deteriorate into pseu- 
doaneurysms approximately 10% of the time. 
The morphology of the intimal disruption did 
not reliably predict the possibility of it wors- 
ening. Even the fact that a flap might be large 
or directed "upstream " into the prograde flow 
of blood did not seem to be a particularly 
ominous sign (Fig. 3-3_B). Also, cases in which 
there appeared to be little intimal damage 
were later found to worsen into pseudo- 
aneurysms. The long held view that these types 
of injuries would soon lead to acute arterial 
occlusions was also proven to be false. To date, 
there have been no documented cases of these 
types of minimal injuries ever deteriorating 
in such a fashion. Of note, no heparin or any 
type of antiplatelet agent was ever used in these 
studies. 

Small (<2-cm) pseudoaneurysms are 
much less common, and as a result, a much 
smaller number of these lesions have been 
followed. Older series had demonstrated 
their potential to thrombose, embolize, 



ch03.qxd 4/16/04 3:21PM Page 91 



3 • MINIMAL VASCULAR INJURIES 



91 



become infected, and even rupture (Linde- 
nauer, Thompson, and Kraft, 1969; Bole, 
Munda, and Purdy, 1976). More current 
studies have also shown them to be more 
likely to worsen over time if watched expec- 
tantly than smooth narrowings or intimal 
flaps (Dennis and colleagues, 1990). 
Approximately 40% will eventually require 
surgical repair, and the remaining 60% will 
either remain stable or improve (Fig. 3-4U) . 
Even those pseudoaneurysms diagnosed on 
a delayed diagnosis basis carry an amputa- 
tion rate reported to be zero (Feliciano and 
colleagues, 1987; Richardson, Vitale, and 
Flint, 1987) . This indicates a more flexible 
approach is possible than mandatory repair. 

Small AV fistulas are the rarest of the 
minimal injuries and similarly may or may not 
resolve over time. Smaller AV fistulas tend to 
close spontaneously, and larger ones will 
more often remain patent and become symp- 
tomatic (Shumacker and Waysson, 1950; Fryk- 
berg and colleagues, 1991). Due to the small 
numbers involved, the exact chance of 
resolution is difficult to determine. Initial 
observation of small fistulas has proven to be 
benign and late repair can also be undertaken 
with no increase in morbidity. 

Over the past decade, no center has been 
able to demonstrate any conclusive data con- 
trary to these studies. Anecdotal reports have 
described cases of delayed presentations of 
arterial injuries (Perry, 1993; Tufaro, 1994). 
These reports generally lack detailed accounts 
of the initial presentation, initial management, 
overall incidence, and consistent follow-up by 
an experienced surgeon. Despite this lack of 
any substantial conflicting evidence, many 
surgeons expressed concern that over the 
long-term follow-up of these patients, some 
of these minimal injuries would eventually lead 
to vascular-related problems. This argument 
was put to rest with a 5- to 1 0-year study showing 
no negative long-term sequelae (Dennis and 
colleagues, 1998) . Two groups of patients were 
studied. The first group of 39 patients had doc- 
umented minimal injuries on arteriograms 
between the years 1986 and 1989. Twenty- 
three of these patients (58%) were re- 
evaluated by history, physical examination, 
and ultrasound at a mean follow-up interval 
of 9.1 years (range, 8.6 to 11.1 years). All were 



asymptomatic, all had normal physical exam- 
ination results, and only one had a residual 
mild narrowing by ultrasound. A second 
much larger group of 287 patients with 
penetrating proximity injuries who were 
seen between the years 1989 and 1991 and 
not evaluated by arteriography was also con- 
sidered. Four had required delayed surgery, 
all within the first week of the injury. Seventy- 
eight patients representing 90 injuries (29%) 
could be contacted. All patients within this 
group reported no long-term complications 
from any missed injury that later developed 
into a significant vascular problem. No patient 
at this institution (including those outside this 
study) has been found to have any deteriora- 
tion after 3 months following the initial 
trauma if they are compliant with follow-up. 
Based on these long-term data, it appears that 
nonoperative management can now be con- 
sidered the standard of care of these minimal 
injuries when identified by arteriography or 
ultrasound. Follow-up of these patients is 
extremely important both within the initial 
hospitalization and for up to 3 months 
after the injury. This component of the man- 
agement may limit the application of this 
approach in small nondesignated trauma 
centers with limited personnel and resources. 



APPLICATION TO 
PENETRATING PROXIMITY 
EXTREMITY TRAUMA 



The rationale behind obtaining arteri- 
ograms in all penetrating trauma to the 
extremities in which the penetrating agent or 
missile trajectory was determined to be in 
proximity to major arteries was the 10% to 
20% chance of a clinically occult injury that 
would otherwise escape detection by physical 
examination. The new emerging data that 
documented these minimal injuries had a 
benign clinical course led to the next step of 
no longer obtaining arteriograms, because 
their detection would not alter any manage- 
ment decisions. A summary of 1 5 studies using 
this approach documents the overall missed 
injury rate to be 1.4% (Table 3-3). This rate 
is not significantly different than the 0.3% to 



ch03.qxd 4/16/04 3:21PM Page 92 



92 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



TABLE 3-3 

PROFILE OF ASYMPTOMATIC PENETRATING INJURIES IN PROXIMITY TO 
EXTREMITY ARTERIES* 





No. of 


No. of Occult 


No. of Occult 




Proximity 


Vascular Injuries 


Vascular Injuries 


Author 


Wounds 


(%) 


Requiring Surgery (%)* 


Dennis and colleagues, 1990* 


254 


25(10) 


2 (0.8) 


= rancis and colleagues, 1991* 


160 


17(11) 


7 (4.4) 


Gahtan, 1994 


394 


37 (9.4) 


7(1.8) 


Gomez and colleagues, 1986 


72 


17(24) 


1 (1.4) 


Hartling and colleagues, 1987 


36 


5(14) 





Itani and colleagues, 1992 


1712 


216(14) 


28(1.6) 


Kauffman, 1992* 


92 


22 (24) 





Lipchik, 1987 


59 


3(5) 


1(1.7) 


McCorkell and colleagues, 1985 


57 


7(12) 





McDonald, Goodman, and Weinstock, 1975 


85 


5(6) 





Rose and Moore, 1988 


97 


Not Given 





Smyth, 1991 


65 


2(3) 


1(1.5) 


Tohmeh, 1990 


58 


1(1.7) 





Trooskin, 1993* 


153 


7 (4.6) 


1(1.3) 


Weaver and colleagues, 1990* 


157 


17(11) 


1 (0.6) 


Total 


3451 


381 (11) 


50(1.4) 



'Including only published cases in the extremity proper, excluding shotgun and thoracic outlet injuries. 

♦Percentage of all proximity wounds, excluding negative explorations. 

♦Prospective study. 

From Dennis JW, Frykberg ER, Veldenz HC, et al: Validation of nonoperative management of occult vascular injuries and 

accuracy of physical examination alone in penetrating extremity trauma: 5-10 year follow-up. J Trauma 1998;44:243-253. 



6% missed injury rate reported for arteriog- 
raphy (Sclafani and colleagues, 1986; Feli- 
ciano, 1987). In addition, arteriography 
carries the small but real risk of contrast allergy 
and local complication such as hemorrhage, 
pseudoaneurysms, and thrombosis. Depend- 
ing on the location and extent of the exami- 
nation, the cost of arteriography will approach 
$2000 to $3000 per patient. The time involved 
in obtaining any imaging study may also delay 
the definitive treatment of other serious asso- 
ciated injuries. 

Some trauma centers, however, have been 
reluctant to base treatment on physical exam- 
ination results alone and have advocated 
duplex ultrasound or Doppler pressure mea- 
surements as alternatives (Bynoe, 1991; 
Johansen and colleagues, 1991; Knudson and 
colleagues, 1993). Though accurate, these 
tests require time, equipment, and skilled per- 
sonnel. In addition, no study has ever demon- 
strated them to be significantly more accurate 
than physical examination alone. Again, 
emphasis must be placed on the importance 



of close observation of these patients to iden- 
tify the small group that will eventually dete- 
riorate and require surgery. This should be 
done for the first 24 hours after the injury and 
for the first 3 months after discharge. Careful 
instructions must be given to patients con- 
cerning the possible development of 
significant vascular symptoms and the need 
to return immediately to the hospital should 
they develop at home. The current manage- 
ment algorithm for penetrating extremity 
injuries at the University of Florida, Jack- 
sonville, is shown in Figure 3-5 (Dennis and 
colleagues, 1998). 



APPLICATION TO 
PENETRATING NECK INJURIES 



Early military and civilian experience first 
led surgeons to adopt the practice of 
mandatory neck exploration for any pene- 
trating injury deep to the platysma (Hughes, 



ch03.qxd 4/16/04 3:21PM Page 93 



3 • MINIMAL VASCULAR INJURIES 



93 



Penetrating Mechanism 
INJURED EXTREMITY 



Resuscitation 



PHYSICAL EXAMINATION 



Hard Signs 



Severe Bone Fracture 
Chronic Vascular Disease 
Soft Tissue Injury 
Shotgun Wound 
Thoracic Outlet Location 
Missile Parallels Vessel 



No 



SURGICAL «- 
EXPLORATION 



No Hard Signs 



Yes 



Positive (Occlusion 
or Extravasation) 



Arteriography 



Negative or "Minimal" — 
Nonocclusive Arterial Injury 



NONOPERATIVE 
OBSERVATION 



■ FIGURE 3-5 

Algorithm for evaluation and 
management of penetrating 
extremity trauma used at 
University of Florida, 
Jacksonville. (From Dennis JW, 
Frykberg ER, Veldenz HC, et al: 
Validation of nonoperative 
management of occult vascular 
injuries and accuracy of 
physical examination alone in 
penetrating extremity trauma: 
5-10 year follow-up. J Trauma 
1998:44:243-253.) ■ 



1954; Fogelman, 1956). Penetrating neck 
injuries were later divided and classified in the 
1960s according to anatomic zones, and man- 
agement was based on the zone in which the 
injury occurred (Monson, Saletta, and 
Freeark, 1969). Patients with penetrating 
injuries to zones 1 and 3 were recommended 
to undergo arteriography because of their 
difficult exposure, and patients with zone 2 
injuries would continue to be explored, 
regardless of the physical findings. 

Similar to the history of managing pene- 
trating extremity trauma, the high number of 
negative explorations led surgeons to consider 
arteriography as an alternative. An extended 
review by Merion in 1981 analyzed 27 articles 
in the literature concerning this topic and 
found no significant difference in the mor- 
bidity or mortality rates between the two treat- 
ment groups (Merion, 1981). These results 
subsequently led most trauma centers to 
replace routine exploration with arteriogra- 
phy (Massac, 1983; Hiatt, Busuttil, andWilson, 
1984; Carducci, 1986) . As experience grew, it 
was soon noted that similar to extremity 
arteriography, cervical arteriography began 
to identify minimal injuries in the carotid and 
vertebral arteries. Initially, surgical dogma 
stated that any abnormality seen on arteri- 
ogram required surgical repair. In the late 
1980s and early 1990s, however, having 
observed the benign natural history of these 
minimal injuries in the extremities, some 



centers began challenging the need to obtain 
any imaging study (Rivers, 1988; Menawatand 
colleagues, 1992). Careful analysis revealed 
that minimal injuries of the cervical arteries 
occurred in approximately 5% of the patients 
in this situation and could be safely observed. 
This was particularly helpful in zone 3 and 
vertebral arteries, because of their difficult sur- 
gical access. Also noted were other abnor- 
malities such as asymptomatic vertebral artery 
occlusions, which did not need to be treated 
either. Mean follow-up time was 6 months for 
the entire study group. 

These retrospective studies formed the 
basis on which prospective studies could be 
performed (Atteberry and colleagues, 1994; 
Biffl and colleagues, 1997; Sekharan and 
colleagues, 2000) . The rationale was similar 
to that in the extremities: If minimal injuries 
do not have to be treated, then arteriography 
is not needed to identify them. It then follows 
that physical examination alone is adequate 
to determine whether a patient needs surgi- 
cal repair after penetrating trauma to zone 2, 
which is amenable to it. Prospective studies 
appear to support this hypothesis (Table 3- 
4). The combined false-negative or missed 
injury rate using this approach is 0.6%, which 
is equal to or better than that of arteriogra- 
phy or ultrasound without the added time and 
cost (Table 3-5) . Some reluctance still appears 
to linger by surgeons to accept this manage- 
ment approach to zone 2 neck injuries. Many 



ch03.qxd 4/16/04 3:21PM Page 94 



94 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



TABLE 3-4 

PENETRATING NECK TRAUMA: MANAGEMENT BY MECHANISM AND LOCATION OF 
INJURY 









Observed 






Explored 








- 


+ 


Missed 
Injury (%) 


- 


+ 


Negative 
Exploration (%) 


Significant 
Injury (%) 


Zone 1 


GSW 


6 











8 





57 




SW 


23 








1 


3 


25 


11 


Zone 2 


GSW 


19 








3 


20 


13 


55 




SW 


109 


1 


0.9 


12 


44 


21 


27 


Zone 3 


GSW 


12 








1 


5 


17 


18 


Total 


SW 


37 
206 




1 



0.5 



17 


8 
88 



16 


29 
29 



GSW, gunshot wound; SW, stab wound. 

From Biff I WL, Moore EE, Rehse DH, et al: Selective management of penetrating neck trauma based on cervical level of injury. 

Am J Surg 1997;174:678-682. 



trauma centers still obtain arteriograms or 
ultrasounds in these patients (Demetriades, 
1995; Ginzburg, 1996). This is probably 
because of a persistentyet unfounded concern 
over causing a stroke by missing a significant 
injury, a complication considered more 
serious than a missed injury of the extremity. 
Published long-term follow-up data are some- 
what lacking at this time also. 



APPLICATION TO HIGH-RISK 
ORTHOPEDIC INJURIES 



Generally, much less evidence is known con- 
cerning minimal arterial injuries and blunt 
trauma. Although the overall risk of arterial 
injury with bone fractures is between 0.3% 
and 6.4% (depending on the definition), 



TABLE 3-5 

STUDIES RECOMMENDING PHYSICAL EXAMINATION ALONE IN THE MANAGEMENT 
OF PENETRATING ZONE 2 NECK INJURIES 





No. of 


Penetrating Zone 2 Injuries 










With Hard Signs 


With No 


No. of Missed 


Study 


Total 


or Explored 


Hard Signs 


Injuries (%) 


Biffl and colleagues, 1997* 


208 


80 


128 


1 (0.9) 


Beitsch, 1994 


178 


42 


136 


1 (0.7) 


Jarvik, 1995 


111 


45 


66 





Demetriades, 1993* 


335 


66 


269 


2 (0.7) 


Gerst, 1990 


110 


52 


58 





Byers, 1990 


106 


62 


44 





Rivers, 1988 


23 


1 


22 





Sekharan and colleagues, 2000* 


145 


31 


114 


1 (0.8) 


Totals 


1216 


379 


837 


5 (0.6) 



'Prospective study. 

From Sekharan J, Dennis JW, Veldenz JC, et al: Continued experience with physical examination alone for evaluation and 

management of penetrating zone 2 neck injuries: Results of 145 cases. J Vase Surg 2000;32:483-489. 



ch03.qxd 4/16/04 3:21PM Page 95 



3 • MINIMAL VASCULAR INJURIES 



95 



several orthopedic injuries have been well 
documented to carry an increased risk up to 
20% of the time (Lange and colleagues, 1985; 
Bassett, 1986; Cone, 1989). These include 
posterior knee dislocations, supracondylar 
humerus fractures, first rib fractures, and prox- 
imal tibia and distal femur fractures. The 
devastating consequences of a missed arter- 
ial injury in these cases (limb loss risk up to 
60%) have led many surgeons to obtain 
arteriograms in all patients with these frac- 
tures or dislocations. This practice would iden- 
tify minimal arterial injuries on approximately 
15% to 30% of the arteriograms obtained 
in the patients with no clinical signs, a rate 
somewhat higher than that seen in pene- 
trating proximity injuries (Dennis, 1993; 
Atteberry and colleagues, 1996). Further- 
more, despite having a different etiology of 
these minimal injuries, the little information 
to date indicates these to have a similar benign 
natural history. 

Particularly ominous has been arterial 
injuries associated with knee dislocations. 
This is due to results of early series in which 
the incidence of limb loss was over 50%, 
although more recently this risk has been 
reduced to less than 5% (Bishara and 
colleagues, 1986). The question whether 
minimal injuries found in the popliteal 
artery following knee dislocation could be 
watched was answered in two studies in 1992 
and 1993 (Treiman and colleagues, 1992; 
Dennis, 1993). These are the two largest 
studies reported, and when their data are 
combined, 16 minimal injuries (all intimal 
irregularities and smooth narrowings) were 
observed nonoperatively without a single 
adverse outcome. Strict follow-up immedi- 
ately after any orthopedic manipulation and 
for several weeks after an injury is essential 
for this type of management to be successful. 

The use of physical examination alone to 
determine whether immediate significantvas- 
cular injury has occurred with these high-risk 
types of orthopedic injuries has been advo- 
cated (Treiman and colleagues, 1992; 
Atteberry and colleagues, 1996) . The presence 
of hard signs of vascular injury mandates 
surgery or arteriography, depending on the 
exact type of clinical picture. Patients with 
a bruit or thrill, distal ischemia, active 



hemorrhage, or absent pulses are generally 
treated with immediate exploration. Those with 
non-life-threatening bleeding or expanding 
hematomas should undergo arteriography, 
because up to 70% of these cases will not have 
significant arterial injury requiring repair. 
Often, single hand-injected arteriograms in 
the operating room before the orthopedic 
manipulation are the simplest and most expe- 
ditious means to evaluate the arterial status. 
Arteriograms are essential if there are multi- 
ple sites of potential arterial injury because 
of more than one orthopedic injury. If 
minimal injuries are detected, they may be 
safely observed with careful follow-up. Recent 
analysis has shown this approach to be both 
safe and accurate when evaluating patients 
with knee dislocations (Miranda, 2001). 



SUMMARY 

Information gathered to date indicates 
minimal injuries tend to follow a similar 
natural history regardless of the trauma 
etiology. Smooth narrowings are extremely 
benign and can be watched with the assur- 
ance that almost all will resolve spontaneously. 
Intimal irregularities or flaps will also gener- 
ally heal, although it must be recognized that 
10% to 15% will deteriorate and require 
definitive treatment. This change will almost 
always occur within the first 3 months after 
an injury, and patients should always be fol- 
lowed for this amount of time as a minimum. 
Those injuries that deteriorate may be 
repaired on a delayed basis with no proven 
adverse effect on morbidity or mortality. 
Careful instructions must be given to patients 
upon discharge, to help them recognize if 
their particular arterial injury is worsening. 
Small pseudoaneurysms (<2cm) may also be 
safely watched buthave a far greater tendency 
to progress to needing direct repair. This prob- 
ably happens 40% to 50% of the time. In addi- 
tion, small AV fistulas need not always be fixed 
immediately unless symptomatic. Evidence 
shows that clinical follow-up is adequate in 
most instances; however, duplex ultrasound 
can be an adjunct in some difficult or partic- 
ularly worrisome injuries. 



ch03.qxd 4/16/04 3:21PM Page 96 



96 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



Knowing this natural history allows physi- 
cians treating trauma patients to use physical 
examination as the definitive basis on which 
to manage penetrating injuries to the extrem- 
ities and neck, as well as knee dislocations from 
blunt trauma. The algorithm in Figure 3-4 
may be used in these situations. The only dif- 
ference in using this approach with neck 
injuries is that distal ischemia may be mani- 
fested by focal neurologic deficits such as a 
stroke or transient ischemic attack. 

Once patients are identified as to having a 
deteriorating minimal injury, they can usually 
be repaired on an urgent, elective basis 
dependent on the presenting symptoms. Stan- 
dard surgical techniques may be used in almost 
all cases. Depending on the experience and 
expertise of the treating surgeons, endovas- 
cular techniques are also proving to be very 
useful in these situations (Marin, Veith, and 
Panetta, 1994; Weiss and Chaikoff, 1999) . The 
placement of a stent graft across these lesions 
on a planned basis either in the operating 
room or in the endovascular suite has become 
standard treatment in some trauma centers. 
Long-term studies will be needed to ensure 
the durability of endovascular repair in these 
situations and compare their outcome with 
proven surgical techniques. 



REFERENCES 

Penetrating extremity trauma and minimal 
arterial injuries 

Dennis JW, Frykberg ER, Crump JM, et al: New 
perspectives on the management of penetrating 
trauma in proximity to major limb arteries. J Vase 
Surg 1990;11:84-93. 

Dennis JW, Frykberg ER, Veldenz HC, et al: Vali- 
dation of nonoperative management of occult 



vascular injuries and accuracy of physical exam- 
ination alone in penetrating extremity trauma: 
5-10 year follow-up. J Trauma 1998;44:243-253. 

Frykberg ER, Crump JM, Dennis JW, et al: Non- 
operative observation of clinically occult arter- 
ial injuries: A prospective evaluation. Surgery 
1991;109:85-96. 

Frykberg ER, Crump JM, Vines FS, et al: A reassess- 
ment of the role of arteriography in penetrat- 
ing extremity trauma: A prospective study. 
J Trauma 1989;29:1041-1052. 

Weiss VJ, Chaikof EL: Endovascular treatment of 
arterial injuries. Surg Clin North Am 
1999;79:653-665. 

Penetrating neck injuries and minimal 
arterial injuries 

Biffl WL, Moore EE, Rehse DH, et al: Selective man- 
agement of penetrating neck trauma based on 
cervical level of injury. AmJ Surg 1997;174:678- 
682. 

MenawatSS,DennisJW, Laneve LM, etal: Are arte- 
riograms necessary in penetrating zone II neck 
injuries? J Vase Surg 1992;16:397-401. 

Sekharan J, Dennis JW, Veldenz JC, et al: Contin- 
ued experience with physical examination alone 
for evaluation and management of penetrating 
zone 2 neck injuries: Results of 145 cases. J Vase 
Surg 2000;32:483-489. 

Orthopedic injuries and minimal 
arterial injuries 

Atte berry LR, Dennis JW, Russo-Alesi F, et al: Chang- 
ing patterns of arterial injuries associated with 
fractures and dislocations. J Am Coll Surg 
1996;183:377-383. 

Bishara RA, Pasch AR, Lim LT, et al: Improved 
results in the treatment of civilian vascular 
injuries associated with fractures and disloca- 
tions. J Vase Surg 1986;3:707-711. 



ch04.qxd 4/16/04 3:22 PM Page 97 





Initial Care, Operative Care 
and Postoperative Care 



DAVID B. HOYT 




RAU 


L COIMBRA 








O 


INTRODUCTION 






n 


GENERAL GUIDELINES- INITIAL CARE -C 








Primary Survey 








Secondary Survey 






O 


NECK VASCULAR INJURIES 






O 


THORACIC VASCULAR INJURIES 






o 


ABDOMINAL VASCULAR INJURIES 






o 


EXTREMITY INJURIES 






o 


COMPLEX ISSUES WITH CONCOMITANT INJURIES 

Blunt Trauma 
Penetrating Trauma 






o 


PERIOPERATIVE CARE 

Initial Anesthesia and Intraoperative Monitoring 
Volume Therapy 
Hypothermia 
Damage Control 






o 


POSTOPERATIVE PRIORITIES 



Hemodynamic Monitoring and Transfusion in the Postoperative 
Phase 

Coagulation Monitoring 

Complications Following Mass Blood Transfusion 



97 



ch04.qxd 4/16/04 3:22 PM Page 98 



98 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



Mechanical Ventilation 

Antibiotics 

Assessment and Determination of Take Back 

Bleeding 

Vascular patency 

Abdominal compartment syndrome 

Extremity compartment syndrome following 
prolonged ischemia 

O SUMMARY 



INTRODUCTION 



The prehospital and the initial in-hospital 
management of trauma patients with vascu- 
lar injuries remain a challenge. Specific 
maneuvers or techniques can be used in the 
prehospital setting to control external 
hemorrhage, but rapid transport to a trauma 
center is of utmost importance. 

Vascular injuries following blunt trauma are 
considered a marker of severe trauma and 
as such should be treated in the context of 
multisystem trauma. Penetrating mechanisms 
cause vascular injuries more often than blunt 
trauma, and depending on the injury loca- 
tion, patients may present with external 
hemorrhage, internal hemorrhage, ischemia, 
or more rarely, a pulsating hematoma or a trau- 
matic arterio-venous fistula. 



GENERAL GUIDELINES: 
INITIAL CARE 

The initial evaluation and management of 
patients with vascular injuries follow the 
guidelines established by the advanced trauma 
life support (ATLS) course of the American 
College of Surgeons-Committee on Trauma 
(ACS-COT) . 

The history should include details about the 
mechanism of injury (e.g., blunt vs. pene- 
trating, position of the patient at the time of 
injury, position of the extremity observed by 
prehospital providers, blood loss at the scene, 
and previous injuries) . 



Primary Survey 

The assessment of airway is the first priority 
even with evidence of obvious hemorrhage. 
Penetrating wounds of the face, neck, and 
chest may be accompanied by airway obstruc- 
tion from bleeding or hematomas. The airway 
should be controlled as soon as possible in 
this circumstance and may require direct trans- 
port to the operating room for definitive 
control with access to complete instrumen- 
tation, excellent light, and anesthesia. 

Many penetrating vascular wounds may 
present with entrance wounds at the lower 
neck or upper abdomen. The wound trajec- 
tory may be such that the chest is involved. 
There may be a pneumothorax, hemathorax, 
or tension pneumothorax, which will present 
as difficulty with breathing and will require 
appropriate diagnosis and decompression 
with a chest tube. This may often be difficult 
to distinguish from airway obstruction or may 
present with airway obstruction in the same 
patient. Systematic evaluation and treatment 
is the best course. 

The occasional patient will present with 
difficulty breathing caused by hypovolemic 
shock. Control of the airway before assessment 
and treatment of the circulation remains the 
priority, even in this circumstance. 

After assessment of the airway and breath- 
ing and definitive airway control, the hemo- 
dynamic status is assessed. Initially, palpation 
of pulse gives a rapid assessment. The pres- 
ence of a radial pulse correlates with a blood 
pressure of at least 90mmHg. The absence 
of radial pulse with the pressure of a carotid 



ch04.qxd 4/16/04 3:22 PM Page 99 



4 • INITIAL CARE, OPERATIVE CARE AND POSTOPERATIVE CARE 



99 



pulse suggests a blood pressure of 60 mm Hg. 
Overall hemodynamic assessment should 
include direct blood pressure measurement, 
but caution should be exercised in the patient 
with a "stable blood pressure." Systolic blood 
pressure can be maintained in the normal 
range until almost 30% of circulating blood 
volume is lost (class I and II hemorrhage). 
Reliance on blood pressure alone can over- 
look a patient with significant hypovolemia. 
As such, measurement of the base deficit will 
also give an initial estimate of the total volume 
of hemorrhage and guide subsequent volume 
resuscitation and assessment of response to 
resuscitation (Table 4-1). 

Recent changes in the ATLS protocols have 
suggested that bleeding control is a priority 
when evaluating the circulation, before fluid 
resuscitation. This is important in patients with 
vascular injuries, because they may present 
with external hemorrhage, for which external 
compression should suffice to control bleed- 
ing. They may also present with intracavitary 
hemorrhage in the chest or abdomen, requir- 
ing an operation to control active hemorrhage, 
as part of the resuscitation phase of care. Always 
keeping control of bleeding as an early pri- 
ority will shift priorities to early operation. This 
is best for vascular injuries. 

Although the ideal fluid therapy (type of 
solution, volume given, and timing of infu- 
sion) for the bleeding patient still remains a 
matter of controversy, it seems reasonable to 
avoid over-resuscitation, particularly in the 
subgroup of patients in whom the index of 
suspicion for the presence of a major vascu- 
lar injury is high. 



The two major goals in the management of 
traumatic shock during initial assessment and 
resuscitation are to arrest hemorrhage and to 
restore blood volume to provide adequate 
tissue oxygen delivery. Delayed resuscitation 
has been proposed to avoid rapid increases 
in blood pressure, clot dislodgement, and con- 
sequently, increased hemorrhage. Avoidance 
of over-resuscitation before surgical control 
is obtained is certainly prudent. However, 
whether all trauma victims would benefit from 
delayed fluid resuscitation is not clear. 

Rapid cannulation of large veins is essen- 
tial for adequate fluid therapy. Care should 
be taken when cannulating upper or lower 
extremity veins in patients with proximal 
penetrating injuries. To achieve adequate fluid 
resuscitation, one should use large-bore 
tubing. Warmed fluids should be infused to 
prevent or minimize heat loss and subsequent 
hypothermia. Rapid infusion systems (such as 
the level I) have the ability to infuse large 
volumes of warmed solutions per minute. Most 
practitioners agree that the initial resuscita- 
tion should be with crystalloids (Ringer's 
lactate or normal saline) ; however, a small but 
very important subset of patients will also 
require blood transfusion. This will be true if 
the estimated blood loss is greater than 30% 
of the total circulating blood volume. Avail- 
ability of type-specific or O-negative blood is 
an essential component of the resuscitation 
of the severely injured patient. 

Resuscitative or emergency department 
(ED) thoracotomy can be used as an adjunct 
to resuscitation in the severely injured patient. 
However, not all patients are candidates for 



TABLE 4-1 










QUANTIFICATION OF BLOOD LOSS (ATLS, 1993) 








Class I 


Class II 


Class III 


Class IV 


Blood loss (ml_) 


<750 


750-1500 


1500-2000 


>2000 


Blood loss (%) 


<15 


15-30 


30-40 


>40 


Heart rate 


<100 


>100 


>120 


>140 


Respiratory rate 


14-20 


20-30 


30-40 


>35 


Urinary output 


>30 


20-30 


5-15 


Absent 


Level of consciousness 


Anxious 


Agitated 


Confused 


Confused/lethargic 


Blood pressure 


Normal 


Normal 


Decreased 


Decreased 



ch04.qxd 4/16/04 3:22PM Page 100 



100 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



this procedure. In general, survival rates are 
higher for patients presenting with vital signs 
than for patients presenting only with signs 
of life. Victims of penetrating trauma benefit 
more than patients with blunt trauma. 

In general, ED thoracotomy is indicated in 
patients with penetrating wounds to the chest 
who develop sudden cardiac arrest or loss 
of vital signs during transport, persistent 
hypotension with signs of cardiac tamponade, 
or intrathoracic hemorrhage. Patients with 
penetrating injuries to the abdomen and 
refractory hypotension may benefit from ED 
thoracotomy and aortic cross clamping before 
exploratory laparotomy is performed; 
however, this is often a matter of individual 
preference. Blunt trauma victims with car- 
diopulmonary resuscitation (CPR) in progress 
and no cardiac electrical activity upon arrival 
are not candidates for this procedure. 

A rapid neurologic assessment should be 
done. A depressed level of consciousness may 
be due to shock, associated blunt head injury, 
drugs or alcohol, or occasionally direct injury 
to the carotid artery. Abnormal or asymmet- 
rical motor function should raise suspicion of 
an intracranial mass lesion and consideration 
of evaluation with a computed tomographic 
(CT) scan should be an early priority. 

Particularly in victims of penetrating 
trauma, it is also important to examine the 
whole body surface area, undressing the 
patient completely because small gunshot or 
stab wounds may be hidden between the but- 
tocks, gluteal folds, in the back, in the axilla, 
or in the folds of the neck. With complete 
exposure, the ongoing concern for hypother- 
mia should be initiated and the patient ade- 
quately covered with warm blankets while 
keeping the ambient temperature warm as 
well. 



Secondary Survey 

The secondary survey should include a 
detailed examination of the vascular system 
in the extremities. The documentation of 
distal pulses is important and will guide 
further investigations and the use of specific 
diagnostic tools. The presence of a distal pulse, 



TABLE 4-2 

HARD AND SOFT SIGNS OF 
ARTERIAL INJURY 



Hard Signs 



Signs of ischemia 



Pallor 



Pain 



Pulselessness 

Paresthesia 

Paralysis 
Poikilothermia 
Pulsatile bleeding 
Palpable thrill/audible bruit 
Expanding hematoma 



Soft Signs 

Diminished distal 

pulses 
Penetrating injury in the 

proximity of major 

artery 
Fracture in the 

proximity of major 

artery 
History of external 

bleeding at the scene 
Peripheral neurologic 

deficit 



however, does not rule out a proximal arter- 
ial injury. On the other hand, bilateral absence 
of distal pulses in a patient in shock with poor 
tissue perfusion does not indicate an arterial 
injury. 

Clinical signs of arterial injury are divided 
into "hard" and "soft" (Table 4-2). According 
to the physical examination findings, patients 
can be stratified according to the risk of 
having an arterial injury. Patients with hard 
signs have high-risk injuries, those with soft 
signs have intermediate-risk injuries, and 
those with no soft or hard signs have low-risk 
injuries. Accuracy of this classification system 
for the lower extremities is improved when the 
ankle-brachial index (ABI) is added. 

A thorough and ongoing neurologic 
evaluation of the victim with penetrating 
extremity trauma is mandatory. Changes in 
the neurologic examination results may indi- 
cate aggravation of ischemia or a developing 
compartment syndrome, and changes in pri- 
orities and management might be necessary 
in these circumstances. 

The diagnosis of a vascular injury in the 
multi-injured patient depends on the mech- 
anism, clinical signs at presentation, and the 
type of arterial injury (Fig. 4-1). 



ch04.qxd 4/17/04 2:48 PM Page 101 



4 • INITIAL CARE, OPERATIVE CARE AND POSTOPERATIVE CARE 101 

ETIOLOGY, INCIDENCE AND CLINICAL PATHOLOGY 



I 



Laceration 





Transection 



• 




Incomplete 
Transection 




Contusion and 
Segmental Spasm 




^;m.<^-' 



I 



Contusion and 
Thrombosis 



Pulsating Hematoma 
or False Aneurysm 




Arteriovenous Fistula 




Contusion and 
True Aneurysm 




External 
Compression 



■ FIGURE 4-1 

Common types of arterial injury. (From Rich NM, Spencer FC: Vascular Trauma. Philadelphia, WB 
Saunders, 1979.) ■ 



NECK VASCULAR INJURIES 

Penetrating neck injuries generally pre- 
sent with external bleeding, a significant 
hematoma, or airway obstruction. Surgical 
access is limited and determined by surface 
landmarks that correlate with surgical acces- 
sibility. Monson's zones define the limits of 
surgical exposure. Zone III injuries (above the 
angle of the mandible) may not be surgically 
accessible and angiography can help define 
this possibility with a suspicious wound or 
hematoma. Zone II wounds (between the 
angle of the mandible and the cricoid carti- 
lage) are directly accessible through the stan- 
dard sternocleidomastoid approach. Zone II 
wounds (below the cricoid cartilage or adja- 
cent to the thoracic inlet) may require tho- 
racic exposure, and hemodynamically stable 



patients should undergo angiography to allow 
surgical planning. 

Blunt carotid or vertebral injuries may be 
suspected because of neurologic abnormali- 
ties detected, but not confirmed, on CT scan. 
Generally a mechanism of extension and 
external rotation can be elicited. Any suspi- 
cion or evidence of blunt neck trauma should 
raise the possibility of carotid injury and 
duplex scanning will screen for this 
possibility. 



THORACIC VASCULAR 
INJURIES 

Penetrating thoracic vascular injuries usually 
present with hemothorax or ischemia. Chest 
tube output will determine whether a 



ch04.qxd 4/16/04 3:22PM Page 102 



102 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



thoracotomy is necessary for bleeding control 
and definitive repair of the arterial injury. If 
upper extremity or cerebral ischemia is the 
predominant clinical sign, a preoperative 
angiogram will help operative planning in the 
hemodynamically stable patient. Recent expe- 
rience with thoracic wounds that traverse the 
chest has used fine-cut CT scans to define 
superficial wounds in hemodynamically stable 
patients. Further experience is needed to 
better define the indications. 

The aorta or its thoracic branches may be 
injured after blunt thoracic injuries. Most 
patients who bleed from these injuries die at 
the scene or during transport. The majority 
of patients with blunt thoracic aortic injury 
will present to the ED hemodynamically stable 
and will have a widened mediastinum on initial 
chest x-ray films. The predominant sign 
accompanying injuries to the thoracic aortic 
main branches is upper extremity or cerebral 
ischemia. The diagnosis is confirmed by 
angiography or high-quality helical CT scans. 
Patients with isolated bluntinjuries to the tho- 
racic aorta should undergo operative repair. 
However, many of these patients have associ- 
ated closed head injuries, and the manage- 
ment of the aortic tear (operative vs. 
nonoperative) will depend on the severity of 
the head injury. 



ABDOMINAL VASCULAR 
INJURIES 



EXTREMITY INJURIES 



Blunt abdominal vascular injuries are rare. 
The astute physician should suspect a major 
intra-abdominal injury secondary to pene- 
trating trauma when the patient does not 
respond to initial fluid resuscitation. These 
patients should be quickly transported to the 
operating room, and the diagnosis is usually 
made intraoperatively. Line placement in the 
lower extremity should be avoided in patients 
with a high index of suspicion for major intra- 
abdominal vascular, particularly inferior vena 
caval injuries. 



In general, patients presenting with significant 
external hemorrhage or limb ischemia caused 
by an isolated penetrating injury to the 
extremity do not pose any difficulty in the diag- 
nosis and management. These patients 
require no additional diagnostic tests and 
should be promptly transported to the oper- 
ating room. Patients with multiple penetrat- 
ing injuries to the extremity presenting with 
ischemia also should be promptly operated 
on; however, a preoperative angiogram may 
be useful in determining the exact location 
of the most proximal injury, thus helping with 
operative planning. 

Angiography is the "gold standard" test to 
evaluate the arterial tree. In some instances, 
hemodynamic instability, associated life- 
threatening injuries, or the need to perform 
other surgical procedures, moving the patient 
to the angiography suite is not feasible. In cases 
of prolonged ischemia or in which the deci- 
sion to perform "damage-control" surgery on 
the injured extremity by placement of an 
intravascular shunt is necessary, an intraop- 
erative on-table angiogram can be obtained. 

Trauma surgeons should be familiar with 
this procedure, because it may save enormous 
amounts of time and may expedite reperfu- 
sion of an ischemic limb. For the lower 
extremity, an 18-gauge needle is inserted into 
the femoral artery, below the inguinal liga- 
ment. An x-ray plate is placed under the thigh 
and 20 mL of contrast is injected under 
pressure. Compression of the femoral artery 
above the needle site will limit contrast dilu- 
tion, and the dye should be injected as rapidly 
as possible. Flow through the Luer lock con- 
nector will prevent injecting too rapidly. The 
initial film will give the surgeon an idea when 
to expose the film to x-ray after the end of the 
injection to demonstrate the vessels in the area 
of interest. Subsequent films are obtained 
distally. 

For the upper extremity, on-table 
angiograms are useful if one wants to evalu- 
ate the proximal axillary artery and the sub- 
clavian artery. This can be done by inserting 
an 18-gauge needle in the brachial artery and 
inflating the cuff of a blood pressure manome- 



ch04.qxd 4/16/04 3:22 PM Page 103 



4 • INITIAL CARE, OPERATIVE CARE AND POSTOPERATIVE CARE 



103 



ter over the forearm. An x-ray plate is posi- 
tioned under the upper extremity and upper 
chest, and 20 mL of contrast is injected under 
pressure. Further films are obtained depend- 
ing on the area of the arterial tree to be 
studied. 

Blunt arterial injuries are usually caused by 
significant forces applied to the extremities, 
also leading to fractures or dislocations. The 
classic fracture or dislocation sites associated 
with arterial injuries are listed in Box 4—1. 
Alignment and immobilization of fractures is 
mandatory to decrease bleeding, avoid further 
injury to soft tissues, and eventually restore 
distal flow. 

In general, vascular injuries are just one 
component of a multitude of injuries, and 
adherence to the priorities set forth by 
the ATLS will facilitate initial management 
and diagnosis. The management of life- 
threatening injuries takes precedence over 
limb-threatening injuries. In the presence of 
multiple injuries, the management of periph- 
eral vascular injuriesmay be delayed, although 
trauma surgeons should keep in mind that 
duration of ischemia greater than 6 hours is, 
in general, associated with poor functional 
outcome and should be avoided. 



COMPLEX ISSUES WITH 
CONCOMITANT INJURIES 

Blunt Trauma 

In patients with concomitant blunt thoracic 
or abdominal trauma and peripheral vascu- 
lar injuries with ischemia, the initial priority 
is to stop the bleeding in the chest (chest tube 
placement and eventually thoracotomy) or 
abdomen (exploratory laparotomy) . If the 
anticipated ischemia time is greater than 6 
hours, consideration should be given to con- 
comitant operations (exploratory laparotomy 
and/or thoracotomy and vascular explo- 
ration) by two separate surgical teams, as well 
as fasciotomy and use of temporary antra- 
arterial shunts. 

For patients with peripheral vascular 
injuries and associated long bone fractures or 
dislocations, best care is provided by a com- 
bined approach, taking into account the 
duration of ischemia. Usually the vascular 
injury can be approached first and the deci- 
sion to use an antra-arterial shunt and perform 
orthopedic fixation followed by definitive 
repair of the vascular injury versus primary 











NLY ASSOCIATED WITH 




■ 


ORTHOPEDIC INJURIES COMMO 
VASCULAR TRAUMA 


1 








UPPER EXTREMITIES 
FRACTURE 


ASSOCIATED VASCULAR 
INJURIES 




O Supracondylar fracture of 

the humerus 
O Clavicular fracture 
O Shoulder dislocation 
O First rib fracture 


O Brachial artery 
O Subclavian artery 
O Axillary artery 
O Aorta, carotid 




LOWER EXTREMITIES 
FRACTURE 






O Posterior knee dislocation 


O Popliteal artery 




O Distal femur 


O Femoral artery 




O Proximal tibia 


O Popliteal trifurcation 



ch04.qxd 4/16/04 3:22PM Page 104 



104 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



repair of the vascular injury followed by 
orthopedic fixation is made intraoperatively 
based on objective assessment of the duration 
of ischemia and the degree of bony instabil- 
ity. Adequate communication between the 
surgeon and orthopedist is key to a success- 
ful management of these complex patients. 



Penetrating Trauma 

The decision-making process in patients with 
penetrating injuries in multiple body areas is 
far less complicated than in blunt trauma. The 
principles of management, however, remain 
the same. 

In patients with penetrating thoracic and 
abdominal injuries, the priority is to treat tho- 
racic conditions first (hemothorax or pneu- 
mothorax), because in the ABCs breathing 
(orU) comes before circulation (or G). As with 
blunt chest trauma, most patients with pene- 
trating chest injuries will not require a tho- 
racotomy and tube thoracentesis will suffice. 
Once pleural problems have been addressed, 
abdominal bleeding should then be addressed 
by means of an exploratory laparotomy. 

In patients with penetrating neck and 
abdominal wounds, the initial priority is to 
obtain a patent airway. If there is active bleed- 
ing from the neck wound, a two-team 
approach should be considered in the 
hypotensive patient, and a concomitant neck 
and abdominal exploration should be per- 
formed. If that is not feasible, then applying 
gentle pressure to the neck wound, opening 
the abdomen, and packing to control major 
bleeding should be done before the formal 
neck exploration is performed, because the 
likelihood of one dying from exsanguination 
is higher with abdominal bleeding than cer- 
vical bleeding. The same principles (ABCs) 
apply to penetrating neck, thoracic, and 
abdominal injuries. 

In patients with chest or abdominal injuries 
and extremity vascular injuries, the priority 
is to rule out intrathoracic hemorrhage, 
pneumothorax, and cardiac tamponade. 
Then, attention is paid to the abdomen. Most 
patients will require an exploratory laparo- 
tomy. Lower extremity vascular injuries are the 
last priority in this scenario. Depending on 



the necessity, an on-table angiogram to deter- 
mine the location of the arterial injury, and 
eventually, intra-arterial shunt placement can 
be done as described previously. Patients with 
multiple penetrating injuries of the extrem- 
ity and signs of ischemia should undergo an 
angiography to help with surgical planning, 
and this can be done in the operating room. 



PERIOPERATIVE CARE 

Initial Anesthesia and 
Intraoperative Monitoring 

The multi-injured patient with a vascular 
injury usually presents with hypovolemia 
caused by hemorrhage. Two factors are of 
utmost importance when initially assessing 
such patients and while in the operating room : 
the evaluation of the circulating blood volume 
deficit and the prediction of additional losses. 

Oxygenation and ventilation, maintenance 
of an adequate perfusion pressure, infusion 
of warm fluids, and serial monitoring of 
urinary output, temperature, hematocrit, 
blood gases, base deficit, and coagulation 
studies are the intraoperative priorities in the 
multi-injured patient. More sophisticated and 
invasive monitoring techniques may not be 
feasible during the resuscitative and opera- 
tive phases of care but should be implemented 
during the critical care phase. 

Ketamine or etomidate are appropriate 
for induction of anesthesia in hypotensive, 
hypovolemic patients. Fentanyl and nitrous 
oxide are also adequate for anesthesia and 
analgesia, but care should be taken when 
hypovolemia is profound. Volatile agents, 
benzodiazepines, and barbiturates should be 
avoided. 

All patients receive antibiotics preopera- 
tively. Antibiotics should be chosen to achieve 
broad coverage but limit toxic side effects, 
particularly when aggravated by associated 
shock. A second-generation cephalosporin is 
ideal. 



ch04.qxd 4/16/04 3:22PM Page 105 



4 • INITIAL CARE, OPERATIVE CARE AND POSTOPERATIVE CARE 



105 



Volume Therapy 

One of the main goals of perioperative care 
is to achieve hemodynamic stability. After trau- 
matic hemorrhage has been controlled, other 
factors may interfere with hemodynamic sta- 
bility. Hemostasis is impaired by the devel- 
opment of hypothermia coagulopathy and 
acidosis. 

Initially, maintenance of adequate intravas- 
cular volume is achieved by fluid resuscitation 
(with crystalloid and blood) during the oper- 
ative phase and extended into the initial crit- 
ical care phase. Although more sophisticated 
devices may be available for intraoperative 
monitoring of the cardiovascular system, 
large-bore intravenous (IV) lines, a central line 
(internal jugular or subclavian), and an arte- 
rial line will generally be adequate. 

Following massive bleeding and shock, pro- 
found acidosis can be concerning because of 
the perceived risk of low pH level. Acidosis 
will resolve with control of bleeding and ade- 
quate volume resuscitation. Use of NaHCO a 
should be limited because rapid formation of 
increased C0 2 can cause precipitation of intra- 
cellular acidosis and make things worse. The 
overuse of bicarbonate can lead to diminished 
oxygen delivery by shifting the oxygen disas- 
sociation curve so that oxygen is more tightly 
bound. Acidosis should never be treated with 
HC0 3 unless the pH level is less than 7.1 to 
7.2. The bicarbonate deficit should be calcu- 
lated and only 50% should be replaced until 
it can be reassessed. Calculation of the HCO a 
deficit is according to the formula: base 
deficit X body weight X 0.2. The space of dis- 
tribution of HC0 3 is considered to be 20% of 
the total body. 

A great deal of confusion and controversy 
regarding the indications for urgent or 
emergent blood transfusion in the severely 
injured patient exists. Several factors should 
be considered before the decision to trans- 
fuse is made, including degree of hemorrhage, 
hemoglobin level, intravascular volume status, 
and chronic diseases. The goal of blood trans- 
fusion is to enhance oxygen delivery to the 
tissues. 

Communication between the surgery team 
and the anesthesiologist is important to avoid 
over-transfusion and under-transfusion intra- 



operatively. Similarly, the use of too much crys- 
talloid can occur particularly if one is not 
watching simple parameters. It can be easy to 
over-resuscitate with crystalloid if the patient 
initially has no urine output because of shock- 
induced acute tubular necrosis (ATN). Con- 
tinuous surveillance of the correction of the 
base deficit should be a reliable guide to 
volume resuscitation in this circumstance, and 
pushing fluid until urine output returns will 
overload the patient. 

Uncross-matched type O blood is immedi- 
ately available for patients with blood loss 
greater than 30% to 40% of total circulating 
blood volume with hypotension. If the 
patient's blood type is known, transfusion 
of type-specific uncross-matched blood is 
appropriate. If time is not a cause for concern, 
type-specific cross-matched blood should be 
used. In an emergency situation, there is often 
not enough time to perform all compatibil- 
ity testing. 

Autotransfusion is an excellent alternative 
or adjunct to massive blood transfusion in the 
hypotensive trauma patient. It is primarily 
useful in patients with a large hemothorax. 
The blood accumulated in the reservoir con- 
nected to the chest tube can be transfused. 
Intraoperative blood salvage using cell-saver 
devices is effective in reducing transfusion of 
stored autologous blood, even in the presence 
of bacterial contamination, because the red 
blood cells are washed before transfusion. 
Most people will not use contaminated blood 
however. Complications of autotransfusion 
include coagulopathy resulting from excessive 
amounts of anticoagulants or infusion of acti- 
vated products of coagulation and fibrinoly- 
sis leading to disseminated intravascular 
coagulation (DIC). 

The use of heparin in the acute setting of 
vascular trauma is controversial. Because of a 
multitude of injuries and in view of massive 
fluid and blood resuscitation, heparin should 
primarily be used locally in vascular trauma, 
and systemic heparinization should be avoided 
until the patient is in the intensive care unit 
(ICU) . After 24 hours when hypothermia and 
coagulopathy have been corrected, and if no 
brain or spinal cord injury has been identified, 
if needed, systemic heparin might be appro- 
priate. 



ch04.qxd 4/16/04 3:22PM Page 106 



106 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



Platelets are used when the platelet count 
drops to less than 50,000 cells/mm 3 associ- 
ated with microvascular bleeding. Prophylactic 
platelet administration after massive transfu- 
sion is not indicated. Fresh frozen plasma 
(FFP) contains all coagulation factors and is 
used in the severely injured patient who has 
continuous bleeding after transfusion of 
approximately one blood volume or when an 
intraoperative coagulopathy is identified as 
measured by a partial thromboplastic time 
(PTT) of more than 1.5. FFP should not be 
used as a volume expander during the resus- 
citation phase and is not indicated for general 
coagulopathy prophylaxis after massive 
transfusion. 

Cryoprecipitate contains fibrinogen, factor 
VIII, factor XIII, and von Willebrand's factor. 
In the acute setting, it is indicated only in 
severe fibrinogen deficiency, or when the 
serum fibrogen level is less than 100 mg%. 



Hypothermia 



metabolism, decreased intestinal motility, 
hyperglycemia, and increased affinity of hemo- 
globin for oxygen. 

Rewarming can be passive or active. Passive 
external rewarming is indicated for mild 
hypothermia and is achieved by increasing 
room temperature and using blanket cover- 
age to prevent further heat loss. Complica- 
tions associated with passive rewarming may 
include metabolic acidosis and increased 
lactic acid production. Active external rewarm- 
ing includes the use of heating or convective 
air blankets and radiant warmers. Active core 
rewarming is indicated for hypothermic 
patients with severe vasoconstriction. Methods 
include warmed IVfluids, body-cavity (pleural, 
peritoneal) lavage with warm fluids, airway 
rewarming, and extracorporeal circulatory 
rewarming (cardiopulmonary bypass). The 
latter is the most effective rewarming method. 
Prevention by avoiding transfusion of 
unwarmed crystalloid and refrigerated blood 
is important, to avoid the problem during the 
first several hours after injury. 



Hypothermia in the severely injured massively 
resuscitated trauma patient is multifactorial 
and may occur at any phase of care. In patients 
experiencing hypoperfusion and shock, heat 
production is decreased. Rapid infusion of 
large amounts of unwarmed crystalloid and 
stored blood also contributes to hypothermia. 

Normal production is 315 kj per day and 
normal loss is about the same. Body temper- 
ature drops by l°Cfor each additional 315 kj 
lost. Each liter of crystalloid at 21 °C can cause 
67.2 kj of heat loss and each unit of 4°C blood 
can cause 30°C heat loss. Even with modest 
resuscitation, this compounded heat loss can 
rapidly cause significant hypothermia. 

Hypothermia affects coagulation by 
decreasing platelet function, altering enzy- 
matic kinetics in the coagulation cascade, and 
increasing fibrinolysis. Oxygen consumption 
and cardiovascular oxygen demand are 
increased in mild hypothermia, and moder- 
ate to severe hypothermia can lead to arrhyth- 
mias, hypotension, and sudden cardiac arrest. 
Other effects of hypothermia include depres- 
sion of the respiratory center, bronchospasm, 
decreased cerebral blood flow, altered level 
of consciousness, fluid shifts, prolonged drug 



Damage Control 

The surgical management of the severely 
injured massively bleeding trauma patient has 
changed dramatically in the last decade. The 
concept of staged laparotomy or damage- 
control operation has emerged from the 
observation that prolonged operations to 
repair all injuries will lead to physiologic 
exhaustion, associated with hypothermia, 
acidosis, coagulopathy, and death. 

By definition, it is a phased approach to the 
critically injured patient. The indications 
include patients with hypothermia (tempera- 
ture <35°C), nonmechanical bleeding, 
pH level less than 7.15, and significant 
retroperitoneal and visceral swelling due to 
massive fluid resuscitation. Eligible patients 
are those with major solid organ injury, pelvic 
fractures, major abdominal vascular injury, 
bleeding injuries in more than one body area, 
or multiple competing injuries. The goal of 
the initial operation is to control bleeding and 
gross contamination of the peritoneal cavity 
with intestinal contents. This can be achieved 
by shunting or ligating injured vessels, packing 



ch04.qxd 4/16/04 3:22PM Page 107 



4 • INITIAL CARE, OPERATIVE CARE AND POSTOPERATIVE CARE 



107 



solid organs (particularly the liver), and 
closing or resecting bowel injuries en bloc, 
using staplers. 

The definitive reconstruction is left to a 
second operation while performing a tempo- 
rary closure of the abdomen. The second 
phase or stage occurs in the critical care unit, 
where the patient will continue to be resusci- 
tated and rewarmed and will receive coagula- 
tion factors to correct coagulopathy. The goal 
of this phase is to restore some of the patient's 
physiologic reserve by correcting the base 
deficit, restoring intravascular volume, and 
achieving adequate oxygenation. Once stable 
(i.e., mechanical bleeding stopped, the base 
deficit corrected, and the body temperature 
near normal) , the patient is taken back to the 
operating room for definitive repair of all 
injuries. This is an approach described for 
abdominal trauma but has been extended to 
chest and pelvic and/or extremity injuries asso- 
ciated with other competing injuries. 

If the abdomen is closed during the first or 
second operation, continuous surveillance is 
necessary to identify an early complication 
associated with this approach abdominal 
hypertension, and its most severe form, the 
abdominal compartment syndrome. 



POSTOPERATIVE PRIORITIES 



Hemodynamic Monitoring and 
Transfusion in the 
Postoperative Phase 

Postoperative placement of a Swan-Ganz 
catheter to monitor cardiac output and pul- 
monary capillary pressure in persistently 
unstable patients or in patients with preex- 
isting illnesses in the ICU is appropriate. 
Nonetheless, it has been ours and others 
experience that analysis of base-deficit trends 
is as helpful as more sophisticated methods 
to monitor effectiveness of resuscitation. If 
used, physiologic parameters should not 
replace the use of metabolic endpoints such 
as base deficit. 

Postoperatively, the decision to transfuse is 
not as simple, because clear guidelines do not 
exist and the "10/30 rule" (lOg/dL of hemo- 



globin or 0.30 hematocrit) is no longer widely 
accepted. The young adult trauma patient 
without comorbid conditions and with a near- 
normal intravascular volume usually tolerates 
a hematocrit level as low as 0.20. Elderly 
patients with limited cardiopulmonary reserve 
may need blood transfusion to maintain a 
hematocrit level of more than 0.25, but no 
good data are available to define this endpoint. 

Recently, signs and symptoms of anemia and 
oxygen delivery measurements have been 
used as transfusion triggers; however, in the 
immediate post-traumatic or postoperative 
period, most trauma patients will be sedated 
and intubated in the ICU, making it difficult 
to evaluate symptoms of anemia. Tachycardia 
and hypotension may reflect anemia but may 
also occur secondarily to inflammatory medi- 
ator release and the systemic inflammatory 
response and are, therefore, not relatable 
transfusion endpoints. 

Measurements of oxygen delivery and con- 
sumption are probably more reliable in pre- 
dicting transfusion requirements. It seems 
reasonable to transfuse blood to patients with 
a hemoglobin concentration of 7 g/dL or less, 
provided the intravascular volume is normal 
and there are no associated chronic illnesses. 
It is also common practice to transfuse blood 
to patients with hemoglobin concentrations 
between 7 and 10 g/dL who have coronary 
artery disease, are older than 60 years, and 
have congestive heart failure. 

Recent National Institutes of Health (NIH) 
recommendations for perioperative blood 
transfusion state that no single criterion for 
transfusion such as a hemoglobin concentra- 
tion less than 1 g/dL should be used and that 
clinical judgment cannot be replaced by any 
single measurement. Perioperative transfu- 
sion of homologous blood carries docu- 
mented risk of infectious and immune 
changes. Recent availability of alternatives to 
autologous blood transfusion should be care- 
fully evaluated. 

Coagulation Monitoring 

As in the operating room, functional evalua- 
tion of coagulation includes platelet number 
and function, activity of coagulation factors, 
and clot breakdown. 



ch04.qxd 4/16/04 3:22PM Page 108 



108 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



Procoagulant activity is evaluated by quan- 
tifying the prothrombin time (PT) and the 
activated partial thromboplastin time (APTT) . 
In the operating room, the time required to 
perform these test may limit their usefulness, 
but postoperatively this should not be a 
problem. Platelet function is evaluated by the 
bleeding time and can be used at the bedside 
to indicate efficacy of coagulation therapy. 
Thromboelastography is a measure of whole 
blood coagulation, and it seems to correlate 
well with other tests of platelet function. It has 
been shown to be useful in the operating room 
to make the diagnosis of factor deficiency, DIC, 
platelet dysfunction, and others, although its 
use is not widespread. 

In the ICU setting, serial monitoring of PT, 
PTT, fibrinogen, fibrin degradation products, 
and platelet count should be done in the 
severely injured patient who has received 
significant amounts of blood products intra- 
operatively or in patients in whom temporary 
hemostatic measures (e.g., packing) were used 
because of diffuse bleeding associated with 
hypothermia, acidosis, and intraoperative 
coagulopathy. Transfusion of clotting factors 
and platelets, treatment of underlying shock 
and hypothermia, and adequate oxygenation 
constitute the basis of therapy in patients with 
post-traumatic coagulopathy. This must be 
done by a constant effort, with both nurses 
and physicians collaborating until the goal is 
accomplished. Less than a full effortwill often 
be met with failure. 



Complications following 
Massive Blood Transfusion 



Citrate intoxication may induce refractory 
hypotension, particularly following massive 
transfusion or continuous infusion at high 
rates. Acute lung injury (ALI) is rare but may 
be eventually seen in the postoperative period 
as a result of complement activation induced 
by the presence donor antibodies interacting 
with recipient granulocytes. Coagulopathy 
following massive transfusion is usually due 
to dilution of platelets and consumption of 
coagulation factors. Microvascular bleeding 
in the setting of massive transfusion and major 
blood loss occurs when platelet counts drop 
to less than 50,000 cells/mm", and fibrinogen 
level is less than 100 mg%. DIC may develop 
postoperatively, secondary to prolonged 
shock, acidosis, and hypoxia. Treatment of 
DIC should focus on the underlying cause and 
replacement of coagulation factors and 
platelets. 

After massive transfusion of citrated blood, 
hypocalcemia may develop. Hypocalcemia 
may lead to cardiac dysfunction and hypoten- 
sion; however, coagulopathy rarely occurs, 
unless serum calcium levels are less than 0.2 
mg/dL. Mobilization of Ca 2+ is usually ade- 
quate after infusion of large amounts of citrate. 
Calcium replacement should be based 
on measured levels and should not given 
prophylactically. 

Patients with normal liver function should 
not receive empirical calcium supplementa- 
tion. Iatrogenic hypercalcemia leads to 
arrhythmias and hypotension. The only 
patients who should receive supplemental 
calcium following massive transfusion are 
those with severe liver disease. 



Box 4-2 lists the most common complications 
after multiple blood transfusions. The inci- 
dence of these complications varies with the 
amount of blood units transfused. Metabolic 
abnormalities are common following trans- 
fusion. Hyperkalemia may be due to potas- 
sium being released from destroyed red blood 
cells. Acidosis may occur as a result of the accu- 
mulation of lactic and pyruvic acids in stored 
blood; however, metabolic alkalosis occurs 
more commonly, because of the conversion 
of citrate to bicarbonate in the liver. 



Mechanical Ventilation 

The routine management of ventilation has 
changed significantly over the last decade. For 
the uncomplicated patient who undergoes 
surgery with no anticipated postoperative 
problems, weaning and extubation should 
follow a standard protocol relying on the rapid 
shallow breathing index as an indicator for 
extubation success. 

A considerable number of severely injured 
patients will develop ALI and acute respira- 



ch04.qxd 4/16/04 3:22PM Page 109 



4 • INITIAL CARE, OPERATIVE CARE AND POSTOPERATIVE CARE 



109 




TRANSFUSION TRANSMITTED DISEASES 

O Hepatitis 

O Human immunodeficiency virus 

O Bacterial infections 

O Viral infections 

HYPOTHERMIA 

COAGULATION DYSFUNCTION 

O Factor dilution 

O Disseminated intravascular coagulation 

O Thrombocytopenia 

ACID-BASE IMBALANCE 

ELECTROLYTE IMBALANCE 

HEMOLYTIC REACTIONS 

ALLERGIC (NONHEMOLYTIC) REACTIONS 

TRANSFUSION-RELATED ACUTE LUNG INJURY 

CITRATE INTOXICATION 



tory distress syndrome (ARDS) . Early 
ALI/ARDS usually follows massive fluid resus- 
citation and its occurrence depends at least 
on the injury severity and hyper-inflammation 
in the post-traumatic period. Late ARDS is 
usually caused or accompanied by sepsis. 

New mechanical ventilation strategies have 
recently been developed to provide adequate 
oxygenation and to decrease the risk of baro- 
trauma and ventilator-induced lung injury. A 
protective strategy can be defined as low tidal 
volumes and the elimination of inspiratory 
plateau pressure while maintaining positive 
end-expiratory pressure (PEEP) above the 
lower inflection point of the pressure-volume 
compliance curve. This can be done with a 
volume ventilator or a pressure ventilator, and 
the recent use of pressure-control ventilation 
has gained popularity because of the relative 
ease in achieving this protective strategy. 



The use of permissive hypercapnia often 
becomes a necessary by-product of this pro- 
tective strategy and has become acceptable 
practice. Multiple studies have suggested this 
strategy is associated with lower mortality. 
Recently, several studies evaluated protective 
strategies and compared them to traditional 
strategies in the treatment of ARDS. Taken 
together, a lung protective strategy including 
lower tidal volumes, permissive hypercapnia, 
and the use of PEEP above the inferior 
inflection point while limiting inspiratory 
plateau pressure seems preferable and prob- 
ably is associated with improved survival. 
Because an actual increase in oxygenation 
does not explain the difference in outcome, 
the reduction in sheer stress and inflamma- 
tion accompanies a lung protective strategy 
could conceivably account for the observed 
effect. Those who follow an evidence-based 



ch04.qxd 4/16/04 3:22PM Page 110 



110 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



strategy in caring for their patients should con- 
sider the routine use of a protective strategy. 
In general, most patients will do well if they 
are placed on initial tidal volume of 4 to 8 
mL/kg, with plateau pressures not to exceed 
35mLH,0. 

Another technique used to improve 
oxygenation is called prone ventilation. The 
rationale for prone ventilation is to decrease 
the volume loss that accompanies patients 
lying on their back and thereby correct 
ventilation/perfusion mismatch. Most believe 
that recruitment of previously atelectatic 
areas induced by altered gravitational forces 
accounts for redistribution of blood flow and 
improvement in ventilation/perfusion and 
oxygenation. Although the evidence is still 
being assessed, this remains an important 
adjunct to patients who are difficult to venti- 
late. Several techniques that allow this to be 
done safely in most patients have emerged, 
including turning devices and protective 
padding. 

Once a patient's ALI or ARDS is resolved, 
all patients must go through a weaning 
process; recent studies suggest that using a 
strategy of a once-daily trial of spontaneous 
breathing is associated with more rapid extu- 
bation than intermittent mechanical ventila- 
tion (IMV) or pressure support weaning. Most 
importantly, a consistent protocol, if used by 
physicians, nurses, and respiratory therapists 
together, seems to be critical to rapid suc- 
cessful weaning. 



Antibiotics 

The use of antibiotics should be guided by 
the general principles of the use of antibiotics 
in trauma patients. In general, these should 
be limited to a preoperative dose and 24 hours 
of postoperative antibiotics. No good data exist 
about whether prolonged antibiotics in 
patients in whom a vascular graft is placed 
reduces the incidence of postoperative infec- 
tion; however, many practitioners will extend 
antibiotic coverage for several days. When 
there is gross contamination and a vascular 
graft needs to be placed (a colon injury and 
iliac artery injury), trying to cover the graft 
or route the graft through uncontaminated 



tissue is best, thereby trying to avoid the 
problem altogether. The use of antibiotics in 
this situation will be user dependent, and even 
here, prolonged antibiotics have some atten- 
dant risks. 

Antibiotics should be limited to second- 
generation cephalosporins and the use of 
multiple antibiotics and in particular amino 
glycosides should be avoided. Recently sur- 
veillance data have documented dramatic 
increases in the incidence in infections caused 
by Staphylococcus aureus, coagulase-negative S. 
aureus, Streptococcus pneumoniae, and Entero- 
coccus. These organisms are associated with a 
rapid increase in resistance to many available 
antimicrobial agents, and prolonged use of 
antibiotics in the initial phase of treatment 
will select out resistant organisms and subse- 
quently cause resistant infection, which may 
be ultimately untreatable. 

Equally important to antibiotics is the pro- 
vision of good graft and anastomosis cover- 
age with local tissue. The use of adjacent 
muscle, omentum, or even the rotation of a 
nonadjacent muscle to get adequate coverage 
and sealing of a graft is essential for avoiding 
infection. 

Once in the postinjury period, antibiotics 
should be targeted to a specific diagnosis, and 
if started for empirical therapy, they should 
be stopped as soon as cultures direct specific 
therapy or indicate that therapy is not needed. 
The length of treatment should be restricted 
to a defined period and the antibiotics 
stopped. Patients should be re-cultured if they 
develop new symptoms. Recent strategies to 
overcome antibiotic resistance include the use 
of rotation of various antibiotics for empiri- 
cal therapy. This avoids the "antibiotic pres- 
sure" that allows resistant organisms to 
emerge. This may be a useful strategy, but it 
will require further study. 

Assessment and Determination 
of Take Back 

Ongoing assessment of vascular injuries 
involves the evaluation of bleeding, assessment 
of peripheral pulses, and the development of 
compartment syndromes of the abdomen and 
extremity. 



ch04.qxd 4/16/04 3:22PM Page 111 



4 • INITIAL CARE, OPERATIVE CARE AND POSTOPERATIVE CARE 



111 



BLEEDING 

Following massive bleeding and coagulopa- 
thy, the assessment of bleeding involves criti- 
cal judgment. On the one hand, until the 
coagulopathy has been reversed and the 
patient warmed, reoperation for bleeding may 
be unsuccessful. Similarly, in the presence of 
profound shock, primary hemostasis may 
have prevented small vessels from bleeding, 
thereby avoiding surgical hemostasis only to 
subsequently vasodilate and bleed in the ICU. 
This dilemma is solved only by careful bedside 
surveillance, concerted correction with coag- 
ulation factors, and continuous monitoring 
of output (e.g., chest tube and drains) , abdom- 
inal distention, hematocrit, and coagulation 
indicators. 

Once a reasonable attempt and success with 
rewarming and factors and platelet repletion 
has occurred, one has to decide whether the 
possibility of unchecked bleeding exists. If 
there is concern and the rate of drain output 
or hematocrit is falling, or if ongoing blood 
replacement does not seem better or contin- 
ually gets worse, then returning to the oper- 
ating room and re-exploration is the most 
appropriate course of action. Reapplication 
of damage control and temporary closure may 
also be appropriate after re-exploration. 



VASCULAR PATENCY 

After vascular repair or reconstruction, par- 
ticularly if accompanied by shock, assessment 
of peripheral pulses may be difficult in the 
cold vasoconstricted patient. The initial pres- 
ence of adequate perfusion can be reassur- 
ing and the presence of symmetrical pulses 
detectable by Doppler flow studies will provide 
initial evidence of patency. With warming, 
distal perfusion should progressively improve 
with brisk capillary refill and good venous 
filling. As resuscitation improves, pulses 
should return or suspicion should be raised 
that there is a problem. Use of segmental 
Doppler flow studies may help, but if there is 
any question about thrombosis, re-explo- 
ration or angiography should be immediately 
pursued. 



ABDOMINAL COMPARTMENT 
SYNDROME 

The abdominal compartment syndrome 
usually occurs in patients undergoing damage- 
control operations, intra-abdominal packing, 
massive fluid resuscitation, and visceral 
swelling. It is characterized by the presence 
of a distended tense abdomen, hypoxia, 
carbon dioxide retention, oliguria, hypoten- 
sion, and high peak inspiratory pressures. The 
diagnosis is suspected on the basis of physical 
findings and is confirmed by measurement of 
intra-abdominal pressure indirectly as bladder 
pressure. Patients with a bladder pressure 
higher than 25 to 30 cm H 2 should return 
to the operating room for decompression and 
the abdomen should be left open. 



EXTREMITY COMPARTMENT 
SYNDROME FOLLOWING 
PROLONGED ISCHEMIA 

Extremity compartment syndrome is the 
result of trauma or reperfusion following 
severe prolonged ischemia, leading to 
increased swelling within a closed fascial com- 
partment. It may also occur after massive fluid 
resuscitation, and continuous surveillance is 
required to avoid delays in diagnosis. This con- 
tained swelling results in an elevation in tissue 
pressure up to the point that blood flow is 
compromised and no longer provides enough 
oxygen to the cells. If left untreated or undi- 
agnosed, itwill resultin myonecrosis and limb 
dysfunction or limb loss. 

The most commonly involved areas are the 
anterior compartment in the lower leg and 
the volar compartment in the forearm. 
Because nerve tissue is more susceptible to 
ischemia than other tissues in the extremity 
(e.g., muscle, bone, and tendons) , initial symp- 
toms are paresthesia and pain. On palpation, 
the muscles are tense, and if the patient is 
awake and able to cooperate with physical 
examination, pain may be severe and even- 
tually increased with passive movement of the 
extremity and contraction of the involved 
muscles. Pulses are usually palpable, even in 
advanced stages, and its presence does not rule 
out this diagnosis. The diagnosis is based on 



ch04.qxd 4/16/04 3:22 PM Page 112 



112 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



physical findings, although a high index of 
suspicion is necessary in the subgroup of 
patients with associated injuries in other body 
areas with competing pain or those sedated 
on mechanical ventilation. Once the diagno- 
sis of compartment syndrome is suspected, 
compartment pressure should be measured. 
Fasciotomy is generally indicated when com- 
partmental pressure is greater than 30 mm Hg. 



SUMMARY 

The management of vascular injuries can be 
one of the most challenging injuries in the 
severely injured patient. They will often be 
accompanied by airway obstruction or trou- 
bled breathing caused by penetrating adjacent 
wounds and will often present in hypov- 
olemic shock. As such, decision making and 
prioritization, decision making in the oper- 
ating room, and limiting operative surgery ini- 
tially and staging it subsequently are complex 
decisions that when made correctly will save 
lives. 

Because of the nature of these injuries, these 
patients are at the highest risk for postoper- 
ative/post-traumatic complications includ- 
ing aspiration, ARDS, renal failure, and 
coagulopathy. The trauma surgeon must be 
equipped to anticipate each of these problems 
and stay ahead of their subsequent deterio- 
ration by aggressive management. 



REFERENCES 

Battistella FD: Ventilation in the trauma and sur- 
gical patient. Crit Care Clin 1998;14:731-742. 

Cosgriff N, Moore EE, Sauaia A, et al: Predicting 
life-threatening coagulopathy in the massively 
transfused trauma patient: Hypothermia and aci- 
dosis revisited. J Trauma 1997;42:857-862. 

Davis JW, Parks SN, Kaups KL, et al: Admission base 
deficit predicts transfusion requirements and risk 
of complications. J Trauma 1996;41:769-774. 

Gentilello LM, Pierson DJ: Trauma critical care. 
Am J Respir Crit Care Med 2001;163:604-607. 

Ham AA, Coveler LA: Anesthetic considerations 
in damage control surgery. Surg Clin North Am 
1997;77:909-919. 

Hirshberg A, Mattox KL: Planned reoperation for 
severe trauma. Ann Surg 1995;222:3-8. 

Ivatury RR, Diebel L, Porter JM, et al: Intraab- 
dominal hypertension and the abdominal com- 
partment syndrome. Surg Clin North Am 
1997;77:783-800. 

Jurkovich GJ, Greiser WB, Luterman A, et al: 
Hypothermia in trauma victims: An ominous pre- 
dictor of survival. J Trauma 1987;27:1019-1024. 

McFarland JG: Perioperative blood transfusions: 
Indications and options. Chest 1 999;1 15:11 3-121 . 

Price JA, Rizk NW: Postoperative ventilatory man- 
agement. Chest 1999;115:130-142. 

Rotondo MF, Zonies DH: The damage control 
sequence and underlying logic. Surg Clin North 
Am 1997;77:761-777. 

Shackford SR, Rich NH: Peripheral vascular injury. 
In Mattox KL, Feliciano DV, Moore EE (eds): 
Trauma. New York, McGraw Hill, 2000. 



ch05.qxd 4/16/04 3:25 PM Page 113 




Diagnosis of 
Vascular Trauma 



JOHN T. ANDERSON 
F. WILLIAM BLAISDELL 



PATHOPHYSIOLOGY 

Classification 

Mechanism 

Ischemia 

Reperfusion Injury 

Compartment Syndrome 

DIAGNOSIS 

History 

Physical Examination 

Hard and Soft Signs of Vascular Injury 

Ancillary Tests 

SUMMARY 



Diagnosis and management of vascular 
injury has evolved dramatically over 
the past century. Early experience 
during combat demonstrated that prompt 
identification and repair of injured arteries 



resulted in improved functional outcome 
and decreased rates of amputation. Military 
experience supported routine operative 
exploration of gunshot wounds of the extrem- 
ities because of a high incidence of vascular 

113 



ch05.qxd 4/16/04 3:25 PM Page 114 



114 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



injury following high-velocity gunshot wounds 
in proximity to major vessels. Application of 
these principles to civilian trauma that typi- 
cally involves low-velocity gunshot wounds, 
shotgun wounds, or stab wounds resulted in 
unacceptably high rates of negative explo- 
rations. Arteriography was promulgated as 
an alternative to mandatory exploration in 
patients without obvious vascular injury (i.e., 
no findings of pulselessness, arterial bleeding, 
or expanding and/or pulsatile hematoma) . 
However, the yield of routine application of 
arteriography, especially for proximity alone, 
is also low. Further, not all arterial injuries iden- 
tified by arteriography require surgical treat- 
ment. Recently, the goal has shifted toward 
the identification of those injuries that require 
operative intervention. To this end, algorithms 
varying from use of physical examination 
alone or in combination with duplex ultra- 
sonography and/or selective arteriography 
have been promoted. The ideal diagnostic 
approach that will maximize accurate detec- 
tion of vascular injury while minimizing mor- 
bidity and cost is still a matter of debate and 
active research. 



PATHOPHYSIOLOGY 
Classification 

Although a wide variety of individual injury 
types may result from trauma (see Fig. 4-1), 
they essentially fall into three basic categories. 
The arterial wall can be completely transected, 
partially transected, or injured without tran- 
section. The patient with a completely tran- 
sected artery will frequently have a history of 
initial active bleeding but present without 
overt hemorrhage. The media of the normal 
artery is capable of significant vasoconstric- 
tion that promotes clot formation and hemo- 
stasis. If the involved artery is a conduit vessel, 
distal pulses will be absent. In certain cir- 
cumstances, hemostasis may not be achieved. 
Iliac and intercostal arteries may continue 
to bleed as tethering of the vessels by sur- 
rounding structures prevents retraction. Also, 
in older patients and those with diseased 



vessels due to atherosclerosis, the artery may 
be incapable of adequate vasoconstriction and 
bleeding may continue unabated. 

Partial transection of an artery limits vaso- 
constriction and the injured area tends to gape 
open. Active external bleeding will continue 
if not contained by surrounding tissues. A 
pseudoaneurysm will form if the tissues 
prevent active external bleeding. Acutely, this 
may be manifest as a pulsatile hematoma at 
the site of injury. Pulses may continue to be 
palpable distal to the site of injury. At times, 
the initial arterial injury is not apparent 
and an expanding pseudoaneurysm may later 
present with pain, a pulsatile mass, or symp- 
toms of nerve impingement. Veins run in prox- 
imity to arteries and are frequently injured 
along with the artery. An arteriovenous (AV) 
fistula may result as blood decompresses from 
a partially transected artery into an adjacent 
injured vein. The AV fistula often is not ap- 
parent on initial presentation. Typically, the 
fistula enlarges over time and may ultimately 
result in high-output cardiac failure or chronic 
arterial or venous insufficiency. 

Finally, the arterial wall can be injured 
without full-thickness transection. The in tima 
of the artery is relatively inelastic in compar- 
ison to the media and the adventitia. Stretch 
or compression of an artery may disrupt the 
in tima and tunica interna of the media while 
leaving the tunica externa of the media and 
the adventitia of the artery intact. Throm- 
bosis of the artery may result from clot 
formation following exposure of the highly 
thrombogenic media or from a mechanical 
obstruction as a result of an intimal flap. Alter- 
natively, small clot fragments may form and 
embolize distally. More severe degrees of 
stretch or compression may weaken or disrupt 
an additional layer of the arterial wall so that 
a pseudoaneurysm may form; extreme 
degrees of stretch will result in complete dis- 
ruption. The use of the term spasm is men- 
tioned only to be discarded. True arterial 
spasm, defined as constriction of the media 
in an otherwise uninjured vessel, is rarely 
present. Spasm identified on arteriography 
invariably represents an intimal injury or 
embolic clot from a more proximal arterial 
injury. 



ch05.qxd 4/16/04 3:25 PM Page 115 



5 • DIAGNOSIS OF VASCULAR TRAUMA 



115 



Mechanism 

Penetrating mechanisms are responsible for 
most vascular injuries, even in rural centers 
that generally care for a predominately blunt 
trauma population. Civilian penetrating 
trauma is almost exclusively from low- 
velocity mechanisms such as handgun, knife, 
or shotgun injuries. Less common injuries 
can occur from penetration by sharp objects 
such as glass, metal, or wood splinters. These 
mechanisms typically cause partial or com- 
plete transection of the artery as a result of 
direct trauma. Occasionally, the vessel is indi- 
rectly injured as a result of an associated frac- 
ture. High-velocity (>2500 feet per second) 
gunshot wounds can directly and indirectly 
injure arteries. Even a trajectory in proximity 
to a major artery may cause arterial damage 
as the kinetic energy of the high-velocity pro- 
jectile is transferred to the tissues. Extensive 



Disrupted 
media 




Lateral geniculate 
collateral flow 

■ FIGURE 5-1 

Mechanism of popliteal artery injury following 
posterior knee dislocation. (Redrawn from 
American College of Surgeons: ACS Surgery: 
Principles and Practice. New York: WebMD, 
2003.) ■ 



soft tissue and skeletal damage and collateral 
circulation disruption also occur. 

Blunt trauma generally causes vascular 
injury as a result of stretch or compression. 
Usually, arterial thrombosis results. The arter- 
ies are particularly susceptible to injury at sites 
of arterial fixation and around joints, for 
example, the popliteal artery, which may be 
injured following knee dislocation (Fig. 5-1). 
At times, bony fragments can puncture the 
vessel directly, as with a supracondylar femur 
fracture (Fig. 5-2) . 

Associated injuries, many of which are life 
threatening and require immediate inter- 




■ FIGURE 5-2 

Injury to the distal superficial femoral artery 
associated with a supracondylar femur 
fracture. ■ 



ch05.qxd 4/16/04 3:25 PM Page 116 



116 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



vention, are frequently present in combina- 
tion with penetrating and blunt vascular 
injuries. Damage due to penetrating trauma 
is generally confined to the area of the tra- 
jectory of the penetrating object, although 
multiple injury sites are common. Blunt 
trauma results in a wider distribution of 
affected structures. Overall, mortality and 
amputation are more common following 
blunt trauma. 



Ischemia 

As a general rule, re-establishing perfusion 
within a "golden period" of 6 hours from the 
time of injury is a desirable goal to ensure 
optimal functional outcome. Warm ischemia 
less than 4 hours generally will not lead to 
muscle necrosis, whereas delays beyond 6 
hours may be associated with significant 
muscle damage. Clinical decisions should not 
be made with strict adherence to these time 
limit guidelines because some degree of 
collateral circulation is often present and 
reperfusion even beyond 6 hours following 
injury may result in successful functional 
outcome. In blunt trauma, associated tissue 
trauma may interrupt collaterals to a greater 
extent then penetrating trauma and likely 
accounts in part for the increased severity of 
ischemia and the higher rate of amputation 
following blunt trauma. An exception to this 
generalization may be seen with high-velocity 
bullet wounds in which associated soft tissue 
injury and collateral disruption may be 
significant. 

The peripheral nerves are especially sus- 
ceptible to ischemia. This is the consequence 
of a high basal metabolic rate and a general 
lack of significant glycogen stores. Dysfunc- 
tion of the nerves due to ischemia results in 
a "stocking-glove" distribution sensory deficit. 
This finding portends progression to gan- 
grene if perfusion is not re-established 
promptly. This should be distinguished from 
direct peripheral nerve injury that will present 
with a neurologic deficit in the distribution 
of the nerve. Paralysis associated with an anes- 
thetic limb carries a bad prognosis. Restora- 
tion of blood flow in such a limb, even within 



the golden period cited earlier, may result in 
limb loss. 



Reperfusion Injury 

"Reperfusion injury" is the damage caused 
locally (i.e., to skeletal muscle and peripheral 
nerves) and systemically following re- 
establishment of blood flow to an ischemic 
body region. Ischemia sets into process a 
number of biochemical alterations that cumu- 
late in cellular damage following reperfusion. 
The severity of the reperfusion injury is cor- 
related with the volume of ischemic tissue (i.e., 
lower limb vs. upper limb) and duration of 
ischemia. A variety of substances are released 
including superoxide anion, a highly reactive 
free radical. Reperfusion results in microvas- 
cular endothelial membrane damage, neu- 
trophil activation, platelet aggregation, and 
decreased nitric oxide production. Ultimately, 
microvascular perfusion is compromised 
resulting in progression of the original 
ischemic injury. Release of metabolic prod- 
ucts into the systemic circulation may cause 
hyperkalemia, acidosis, and myoglobulinemia. 
Additionally, the inflammatory and coagula- 
tion systems are activated. Cardiac arrhyth- 
mias, acute respiratory distress syndrome, 
renal failure, multiorgan failure, and death 
may follow if not identified and aggressively 
treated. Reperfusion of the entire lower limb 
may be life threatening. Reperfusion of 
the lower leg is less morbid but can still have 
life-threatening consequences in the older 
patient. 



Compartment Syndrome 

Compartment syndrome results from swelling 
of soft tissues enclosed within a relatively rigid 
fascial space. As pressure increases within 
the compartment, microvascular perfusion is 
limited, and ultimately, tissue necrosis results. 
Most commonly, compartment syndrome 
occurs in the lower leg or forearm, however, 
additional locations can be involved. Swelling 
may result from hemorrhage into the soft 
tissues, from tissue edema as a result of 
venous occlusion, from ischemic or dying 



ch05.qxd 4/16/04 3:25 PM Page 117 



5 • DIAGNOSIS OF VASCULAR TRAUMA 



117 



muscle, or from external causes such as tight 
casts or circumferential burn eschars. 

The diagnosis of compartment syndrome 
is clinical and based on clinical findings of the 
four Ps: pressure, pain, paresthesia, and intact 
pulses. Increased pressure is manifested as a 
tense compartment to palpation and can be 
confirmed by direct pressure measurement. 
Pain is out of proportion to that expected from 
the extremity injury. Also, passive stretching 
of the ischemic muscle aggravates the pain. 
Paresthesia, which may progress to complete 
anesthesia and paralysis, is a late finding in 
compartment syndrome. Distal pulses are 
often intact, a finding that when present serves 
to distinguish compartment syndrome from 
arterial insufficiency. 

Compartment pressures are readily mea- 
sured with either handheld devices (e.g., 
Stryker pressure monitor) or with a side-port 
catheter attached to an arterial pressure trans- 
ducer. Blood flow to muscle is cut off when 
compartment pressures exceed venous pres- 
sure. Criteria based on an absolute compart- 
ment pressure value are of limited utility 
in hypotensive patients. Several investigators 
have advocated calculation of a gradient 
between the measured compartment pressure 
and either the mean arterial pressure or the 
arterial diastolic pressure. A gradient of less 
than 10 to 30mmHg below the diastolic or 
less than 30 to 40mmHg below the mean 
arterial pressure has improved specificity in 
the diagnosis of extremity compartment syn- 
drome. Of note, patients with venous injury 
or obstruction are particularly susceptible to 
subsequent compartment syndrome and 
should be closely monitored. 

An effort should be made to determine the 
cause of the compartment syndrome. The 
ultimate functional outcome of fasciotomy 
depends on the etiology of the compartment 
syndrome and the extent of muscle necrosis. 
Increased compartment pressures due to 
either hemorrhage into the compartment or 
venous obstruction, especially with underly- 
ing viable muscle, are clear-cut indications of 
the need for fasciotomy. Controversy regard- 
ing the utility of fasciotomy arises in patients 
who have compartment syndrome on the basis 
of ischemia alone (e.g., compartment syn- 
drome of the calf following prolonged femoral 



artery occlusion). Release of the fascial enve- 
lope will not result in recovery of necrotic 
muscle. Subsequent infection, nonhealing, 
and need for amputation generally result. 
These patients are best served without fas- 
ciotomy. The muscle will become fibrotic; 
however, the patient may be left with a func- 
tional limb. 



DIAGNOSIS 



Identification and management of life- 
threatening injuries and treatment of shock 
should be the first priority. Advanced Trauma 
Life Support (ATLS) guidelines should be fol- 
lowed. Initial treatment and evaluation should 
proceed simultaneously. It is important to rec- 
ognize and control external hemorrhage. 
Generally, direct pressure is effective. Patients 
should be promptly resuscitated as the pres- 
ence of shock itself may lead to diminished 
pulses in the extremities and confusion about 
the presence of vascular injury. Associated frac- 
tures and dislocations may compromise vas- 
cular patency and should be reduced. 
Frequently, there are associated injuries to the 
abdomen, chest, or head that require imme- 
diate intervention. Prompt resuscitation and 
identification and management of vascular 
injuries should be the goals to limit warm 
ischemia and ensure optimal functional 
outcome. 

History 

The patient and prehospital personnel should 
be questioned about the mechanism of injury. 
With penetrating trauma, information such 
as the length of the knife, the number and 
direction of bullets fired, and the body posi- 
tion at the time of injury should be sought. 
With blunt trauma, the severity of the injury 
mechanism (e.g., distance of fall, vehicle 
speed, and damage) and evidence of fracture, 
dislocation, or altered perfusion of extremi- 
ties should be elucidated. Further, certain 
mechanisms such as "car bumper" injuries or 
posterior knee dislocations may be associated 
with vascular injury and their occurrence 
should be sought. The amount and charac- 



ch05.qxd 4/16/04 3:25 PM Page 118 



118 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



ter of blood loss should be ascertained. Bright 
red pulsatile bleeding is suggestive of an 
arterial injury, whereas dark blood suggests 
a venous origin. Evidence of shock must be 
soughtfrom the prehospital personnel, aswell 
as the volume of fluid administered. The use 
of a tourniquet and duration of its applica- 
tion should be determined. Information 
about neurologic symptoms including sensory 
and motor deficits should be obtained. Also, 
the patient should be questioned regarding 
a history of peripheral vascular disease, dia- 
betes, or other conditions such as coronary 
artery disease that carry a high incidence of 
associated vascular disease. 



Physical Examination 

The patient should be adequately exposed and 
thoroughly examined. Deformity due to frac- 
ture or dislocation should be identified. 
Careful attention should be directed to skin- 
folds in the axilla or perineum and buttocks 
that may hide wounds due to penetrating 
trauma. In the case of penetrating trauma, the 
trajectory of the wounding object should be 
estimated, particularly with reference to major 
arteries. Wounds should be inspected for 
evidence of active bleeding or hematoma 
formation. The character of the bleeding, 
pulsatile bright red blood, or a steady ooze 
of dark blood should be noted. A tense or 
expanding hematoma indicates the presence 
of an arterial injury with bleeding contained 
by surrounding soft tissues. Finally, the oppo- 
site uninjured extremity should be inspected 
for evidence of chronic peripheral vascular 
disease. Absent pulses in the non-injured leg 
markedly decreases the likelihood of vascu- 
lar injury in the traumatized extremity. 

The pulse examination should include pal- 
pation of pulses proximal and distal to the 
injury. Skin temperature and capillary refill 
distal to the injury should also be assessed as 
indexes of perfusion. A difference in the char- 
acter of the pulse or skin perfusion should 
prompt additional workup. Of note, pulses 
may be palpable and normal in up to one third 
of patients with avascular injury. Comparison 
of skin perfusion and pulses of the injured 
extremity to that of the non-injured extrem- 



ity is very helpful. Hypoperfusion and dimin- 
ished peripheral pulses due solely to shock 
will be similar on both sides. Further, dimin- 
ished or absent pulses as a result of periph- 
eral vascular disease are generally symmetrical 
between the extremities. Occasionally, patients 
may have a congenital absence of the dorsalis 
pedis pulse. 

AV fistulas may be identified by ausculta- 
tion of a bruit over the involved arterial 
segment; a thrill, palpable evidence of an AV 
fistula, is rarely present in acute injury. Aglove 
should be placed over the bell of the stetho- 
scope to keep the stethoscope free of blood 
when there is an open injury. Thrills and bruits 
may not be obvious, particularly early after 
injury. AVfistulas generally progress over time 
and bruits that were not initially present may 
appear the next day. 

Complete preoperative evaluation and doc- 
umentation of neurologic function is impor- 
tant, as ultimate functional outcome largely 
depends on intact sensory and motor func- 
tion. As mentioned, a "stocking-glove" distri- 
bution sensory deficit indicates neurologic 
dysfunction due to ischemia. Development 
of gangrene will ensue if flow is not promptly 
re-established. 

Hard and Soft Signs of 
Vascular Injury 

Findings identified on history and physical 
examination may be divided into two cate- 
gories, hard signs and soft signs, each with a 
varying degree of association with arterial 
injury (Table 5-1 ) . Hard signs are strong pre- 
dictors of the presence of an arterial injury 
and the need for operative intervention. 
Obvious examples are pulsatile bleeding 
or an expanding hematoma. Evidence of 
ischemia manifested with the six Ps: Pulse- 
lessness, pallor, pain, paralysis, paresthesia, 
and poikilothermia are further strong evi- 
dence of arterial injury. A thrill, palpable evi- 
dence of an AV fistula, is not as commonly 
noted as is a bruit. An arterial pressure index 
(API) of less than 0.90 is included as a hard 
sign. The API is determined by dividing the 
systolic pressure of the injured limb by the 
systolic pressure of the non-injured limb. 



ch05.qxd 4/16/04 3:25 PM Page 119 



5 • DIAGNOSIS OF VASCULAR TRAUMA 



119 



TABLE 5-1 

HARD VERSUS SOFT SIGNS OF 
VASCULAR INJURY 



Hard Signs 

Active arterial bleeding 

Pulselessness/evidence 

of ischemia 
Expanding pulsatile 

hematoma 
Bruit or thrill 
Arterial pressure index 

< 0.90 pulse deficit 



Soft Signs 

Neurologic injury in 
proximity to vessel 

Small to moderate-sized 
hematoma 

Unexplained hypotension 

Large blood loss at scene 

Injury (due to penetrating 

mechanism, fracture, or 

dislocation) in proximity 

to major vessel 



trauma to look for foreign bodies or evidence 
of fracture or dislocation. Radiopaque 
markers should be placed on all penetrating 
wounds for identification on subsequent radi- 
ographs. The number of bullets visualized 
should be correlated with the number of 
wounds. The sum of the number of bullets 
and the number of wounds should equal an 
even number. If the sum results in an odd 
number, the possibility of a missile embolism 
should be entertained. The bullet may travel 
within the vascular system to the heart and 
pulmonary system if the bullet gains access to 
the venous system (Figs. 5-3 and 5-4) or the 
distal artery if the bullet gains access to the 
arterial system. Additional x-ray films, includ- 
ing fluoroscopy, should be obtained and the 



Johansen and colleagues (1991) found that 
an API of less than 0.90 had 95% sensitivity 
and 97% specificity for identification of occult 
arterial injury. Further, an API of more than 
0.90 had a 99% negative predictive value for 
an arterial injury. However, an API may be 
normal in patients who have injuries to non- 
conduit vessels such as the femoris profunda. 
Also, API can be unreliable in the evaluation 
of penetrating injuries in the region of the 
groin. Nonetheless, the API is readily obtained 
at bedside and is a useful extension of the phys- 
ical examination, particularly when the pulse 
strength is questionably diminished. 

Soft signs are suggestive of an arterial 
injury, though with a much decreased sensi- 
tivity and specificity than hard signs (see Table 
5-1). The incidence of arterial injury varies 
with the specific finding. When proximity is 
the only indicator of possible vascular injury, 
evaluation with arteriography generally finds 
identifiable injuries in fewer than 10% of 
patients, and many do not require additional 
specific treatment other than observation 
alone. Much of the controversy of vascular 
trauma evaluation centers on the workup of 
patients with soft signs. 



Ancillary Tests 

Plain x-rays of the injured extremity should 
be obtained in both penetrating and blunt 




■ FIGURE 5-3 

Bullet embolism to pulmonary artery from iliac 
vein injury. (From What's Your Diagnosis: 
Photographic Case Studies in General Surgery. 
Greenwich, Conn: Cliggott Publishing, 1994.) ■ 




■ FIGURE 5-4 

Extraction of bullet embolism to right pulmonary 
artery. (From What's Your Diagnosis: 
Photographic Case Studies in General Surgery. 
Greenwich, Conn: Cliggott Publishing, 1994.) ■ 



ch05.qxd 4/16/04 3:25 PM Page 120 



120 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



patient re-examined until the discrepancy is 
resolved. Occasionally, folds in the axilla and 
perineum may hide additional wounds. 
Finally, the foreign body image should be scru- 
tinized. Blurring of the edges of the bullet sug- 
gests movement that could be the result of an 
intimate relationship with a pulsatile artery. 

Application of routine operative explo- 
ration of penetrating extremity trauma to civil- 
ian trauma proved to result in a large number 
of negative explorations. In a landmark paper, 
Snyder and colleagues (1978) validated the 
use of arteriography to accurately detect vas- 
cular lesions. They evaluated 177 patients with 
183 penetrating extremity injuries using arte- 
riography and subsequent operative explo- 
ration. They identified 1 false-negative and 14 
false-positive arteriograms. Arteriography is 
generally not required in patients with obvious 
arterial injury (e.g., active arterial bleeding, 
pulselessness, and/or expanding hematoma) . 
However, arteriography is invaluable in 
patients in whom the diagnosis is less clear or 
the extent or location of vascular injury is not 
readily apparent (Table 5-2) . Arteriography 
is generally well tolerated. Complications can 
occur in 2% to 4% of patients. Usually, these 
are minor, most frequently limited groin 
hematomas. Major complications such as 
iatrogenic pseudoaneurysms, AV fistulas, or 
embolic occlusion are uncommon. In part, 
this may reflect the younger population typical 
for trauma. 

Patients who require urgent operation 
either for an obvious vascular injury or for 
life-threatening associated injuries should 
have any necessary arteriograms performed 
in the operating room to minimize warm 



TABLE 5-2 

INDICATIONS FOR ARTERIOGRAPHY: 
EXTREMITY TRAUMA 

Unclear location or extent of vascular injury 

Extensive soft tissue injury 

Fracture or dislocation 

Trajectory parallel to an artery 

Multiple wounds 

Shotgun injuries 

Peripheral vascular disease 



ischemia time. Arteriograms are obtained with 
percutaneous cannulization of the artery 
proximal to the site of suspected injury. Con- 
trast is then injected into the artery, and plain 
films are obtained. Fluoroscopy can be used 
as an alternative to plain radiographs to min- 
imize the amount of contrast required and to 
aid with timing of the contrast injection. If 
visualization of the axillary artery is necessary, 
outflow occlusion with a cuff can be per- 
formed to allow filling of the axillary artery 
proximal to the site of contrast injection as 
described by O'Gorman and colleagues 
(1984). Of note, O'Gorman and colleagues 
(1984) described the use of surgeon-per- 
formed angiography in the emergency room 
to exclude significant vascular injury. They 
were subsequently able to discharge some 
patients from the emergency room. 

Duplex ultrasonographic scanning has been 
shown by several investigators to have a sen- 
sitivity and specificity approaching 100% for 
the investigation of penetrating extremity 
trauma. The modality combines real-time 
two-dimensional imaging with guided Dop- 
pler insonation. Flow to or from the point of 
Doppler investigation can be represented on 
a color scale. Duplex ultrasonography is more 
sensitive than an API evaluation. Further, non- 
conduit arterial injuries, which do not alter 
the API, can be evaluated. 

Evaluation of the patient with penetrating 
extremity trauma who presents with only soft 
signs of arterial injury continues to be a subject 
of debate. Routine use of arteriography on 
patients with proximity injuries will identify 
abnormalities in up to 10% of cases. Several 
series report the need for vascular repair in 
between 0.6% and 4.4% of patients with prox- 
imity penetrating injuries (Dennis, 1998) . 
Dennis, Frykberg, and colleagues (1998) has 
championed physical examination alone in 
this group, arguing that patients those patients 
who need operative intervention will ulti- 
mately develop identifiable hard signs. A 
number of investigators have promulgated 
duplex ultrasonography as a noninvasive alter- 
native between routine arteriography and 
physical examination alone. The lesions 
missed on initial physical examination that 
ultimately require operative intervention are 



ch05.qxd 4/16/04 3:25 PM Page 121 



5 • DIAGNOSIS OF VASCULAR TRAUMA 



121 



injuries to the profunda femoris artery, 
pseudoaneurysms, and AV fistulas. Both of 
these lesions progress with time and delayed 
operative repair is technically more chal- 
lenging. Further, many investigators have 
documented the poor follow-up possible in 
the trauma population. Duplex ultrasonog- 
raphy is capable of detecting these lesions at 
the time of initial presentation. The major lim- 
itations are that the modality is technician 
dependent and often not readily available 
during off-hours when most patients are 
injured. However, even proponents of physi- 
cal examination alone advocate admission and 
observation for 24 hours. Moreover, more than 
43% of patients with proximity penetrating 
extremity trauma at University of California- 
Davis required physical therapy, and 46% 
required complex wound care. Thus, admis- 
sion during which duplex ultrasonography 
can be obtained is easilyjustified. Ultimately, 
the choice is a balance between the need to 
promptly diagnose all arterial injuries requir- 
ing treatment and prevent unnecessary 
morbidity on the one hand and the cost, 
availability, and morbidity of diagnostic modal- 
ities on the other hand. 

Patients with suspected vascular injury fall 
into three basic priorities: (1) Patients with 
evidence of pulselessness/ischemia, active 
bleeding, or a pulsatile hematoma, (2) patients 
with hard signs and a palpable pulse, and (3) 
patients with soft signs or an injury known to 
be associated with vascular injury. In all 
patients, life-threatening injuries take prior- 
ity and should be addressed immediately. The 
patient should be resuscitated and shock 
managed appropriately. Fractures and dislo- 
cations should be reduced and the pulse exam- 
ination and skin perfusion evaluated. Hard 
and soft signs should be specifically elicited 
(Fig. 5-5). 

Patients who are pulseless or show evidence 
of ischemia or those who have active bleed- 
ing manifested with either external bleeding 
or an expanding pulsatile hematoma should 
be taken promptly to the operating room. 
Often, the location of the injury can be 
identified from the history and physical 
examination and operative intervention can 
proceed directly. However, in certain cir- 



cumstances, the exact location or extent of 
injury may be unclear (see Table 5-2). In these 
patients, on-table arteriography, performed 
in the operating room, is used to assess vas- 
cular injury and minimize warm ischemia 
time. 

Patients who have hard signs but have a pal- 
pable pulse and no evidence of ischemia can 
generally undergo a more deliberate workup. 
Usually information required to guide inter- 
vention is obtained from formal arteriogra- 
phy in the radiology suite. In general, the 
quality of formal arteriograms is better than 
that of those obtained in the operating room. 
Additionally, therapeutic endovascular pro- 
cedures such as embolization of bleeding 
muscular branches, pseudoaneurysms, or AV 
fistulas can be performed in the radiology 
suite. Occasionally urgent operation is 
required for associated injuries, such as intra- 
abdominal injury. In these patients, on-table 
arteriography provides a means to promptly 
identify arterial injuries to guide subsequent 
operative or nonoperative management. 

Patients who have soft signs or who have a 
mechanism suggestive of an arterial injury, 
such as a posterior knee dislocation, can be 
evaluated in various ways. Viable options, each 
with their ardent advocates and literature 
support, include routine arteriography or 
duplex ultrasonography and/or serial physical 
examination. Vascular abnormalities identi- 
fied by duplex ultrasonography are subse- 
quently evaluated with arteriography as a 
diagnostic and possibly therapeutic inter- 
vention. Development of hard signs in patients 
followed by physical examination is generally 
evaluated by formal arteriography. 

Patients with injury in the region of the 
groin, thoracic outlet, or neck should undergo 
arteriography. Duplex ultrasonography of 
the subclavian, axillary, and iliac vessels is 
limited. Further, consequences of missed 
injuries in these areas, such as exsanguination 
from intrapleural hemorrhage, may be cata- 
strophic. At times, the vascular lesion may 
be amenable to treatment with endovascular 
techniques. 

Once arterial injuries are identified and 
delineated, management proceeds as 
appropriate. 



ch05.qxd 4/16/04 3:25 PM Page 122 



122 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 




































"D 










c 










D 








o 

z 


o 

CO 
CO 










±1 
















D 










X 



































Q. 








•o 












o 


" o- 












z 


s- § 


































- ° V, 




CO 
















o - co 
CO c o 




£ 










So -5 




6 










iEc 




c 

< 














v 




























£ 








1 >- 










CO 






w 






CO 








c 













O) 












o 








>- 


q 






















CO 




o 













o 




^J 






t 








o 




c 






CO 






.*: w 

















85 




CO ~.. 
£ Q. 




5 


o 


"c0 

£ 




£ 


-C co 
en 

O 

o 

"O 



>- 


Q. O 

CO 
c > 

"D 

CO 

X 




O 
CO 
CO 

> c- 

1.1 

£ 8. 

TO o 


Z 


o 




■2 

1 

a. 

to 

IB 

C 
0) 




^- 


o 

z 




















































3 

"D 






£ 

CO 




E 
2> 








o- 

CO 

o- lo- 
co S ? 




C0 








z 




>- 


q 





IB 
C 

I 
















CO CO .<= 
















Pulselessne 

:panding hem 

Active bleed 






CO 















> 










to 


>■ 




CO 





o. 
o 














>- 


CO 
















x 










c 








LU 




o 

CO 




































o 












c 












D 
























C 



























X 












CO 


<D 




























>- 


c 
o 

o 
o 

_i 






CO 

o 
a. 

X 













* 


' 








o 


> 














z 




Q. 




























O 









V 




(1) 




TI 




(1) 






in 


u 


1 


o 


in 


Cfl 










fa 




oi 


u 


U 


o 





m 


h+ 


zs 


fa 


CO 



ch05.qxd 4/16/04 3:25 PM Page 123 



5 • DIAGNOSIS OF VASCULAR TRAUMA 



123 



SUMMARY 



Diagnostic modalities and algorithms have 
evolved as treatment paradigms have pro- 
gressed over the past century from expectant 
management, to repair of all arterial injuries, 
to the repair of selected arterial injuries prac- 
ticed today. Continued refinements are to be 
expected with ongoing clinical research and 
patient follow-up. 



REFERENCES 

Dennis JW, Frykberg ER, Veldenz HC, et al: 
Validation of nonoperative management of 
occult vascular injurie s and accuracy of physical 
examination alone in penetrating extremity 
trauma: 5- to 10-year follow-up. J Trauma 1998; 
44(2):242-252. 

The authors present long-term outcome data to 
validate safety and efficacy of physical examina- 
tion alone to determine the treatment of pene- 
trating extremity trauma. Two groups of patients 
are presented, the first during a period of liberal 
use of arteriography, and the second with the 
use of physical examination alone. Group 1 had 
43 patients with 44 clinically occult injuries sub- 
sequently demonstrated on angiography. Four 
(9%) had deterioration within a month and 
required operative repair. Follow-up, with a mean 
of 9.1 years, was possible in 58% of the remain- 
ingpatients; all were asymptomatic. Group 2 had 
287 patients with 309 asymptomatic proximity 
injurie s evaluated by physical examination alone . 
Four (1.3%) deteriorated and required surgery. 
Follow-up, with a mean of 5.4 years, was possi- 
ble in 29%; no patient reported vascular 
symptoms. 

Fry WR, Smith RS, Sayers DV, et al: The success of 
duplex ultrasonographic scanning in diagnosis 
of extremity vascular proximity trauma [see Com- 
ments] . Arch Surg 1993;128(12):1368-1372. 

Study of the use of duplex ultrasonographic scan- 
ning in the evaluation of penetrating extremity 
vascular trauma. Two-hundred patients with 225 
penetrating extremity injuries were evaluated 
with duplex ultrasonography for either vascular 
proximity injury or diminished pulse strength. 
Arteriograms were obtained in the first 50 
patients. The sensitivity and specificity were both 
100% in this initial cohort. Duplex ultrasonog- 
raphy was used in the remaining 175 injuries. 



Eighteen injuries were identified, seventeen of 
which were confirmed by either arteriography 
or operative exploration. The remaining patient 
had spasm of the superficial femoral artery on 
arteriography, which did not require treat- 
ment. Seven unsuspected venous injuries were 
identified. 

Johansen K, Lynch K, Paun M, Copass M: Non- 
invasive vascular tests reliably exclude occult 
arterial trauma in injured extremities. J Trauma 
1991;31(4):515-522. 

Follow-up study to validate the use of arterial pres- 
sure index (API) to exclude significant arterial 
damage in patients with extremity trauma. 
Overall, a value of 0.90 was found to have a sen- 
sitivity and specificity of 95% and 97%, respec- 
tively, for the presence of significant arterial 
injury. A value of more than 0.90 had a negative 
predictive value of 99% . Arteriography was advo- 
cated for those limbs with an API of less than 
0.90. The authors argue the API is safe, accu- 
rate, and cost-effective in the evaluation of 
extremity vascular trauma. 

O'Gorman RB, Feliciano DV, Bitondo CG, et al: 
Emergency center arteriography in the evalua- 
tion of suspected peripheral vascular injuries. 
Arch Surg 1984;119(5):568-573. 

Description of a surgeon-performed arteriog- 
raphy in a group of 488 patients with suspected 
vascular injuries. The majority of arteriograms 
were obtained for proximity (353/488) ; 76 arte- 
riograms were performed for a diminished 
pulse. Overall, 20% of the patients were found 
to have a vascular injury requiring subsequent 
operative intervention. Only one false-normal 
and four false-abnormal arteriograms were 
reported. The authors conclude that the method 
is simple, sensitive, and cost-effective in patients 
with potential peripheral vascular injuries. 

Richardson JD, Vitale GC, Flint LM Jr: Penetrat- 
ing arterial trauma. Analysis of missed vascular 
injuries. Arch Surg 1987;122(6):678-683. 

Classic article describing an experience of 677 
patients with penetrating wounds to the upper 
and lower extremity and neck with suspected 
vascular injury. Patients were evaluated with a 
combination of surgical exploration and/or 
arteriography. Long-term follow-up for an 
average of 5.1 years was obtained in 33% of the 
patients. Missed vascular injurieswere identified 
in patients undergoing either surgical explo- 
ration alone or arteriography alone. No missed 
injuries were identified in patients who 
underwent both arteriography and surgical 
exploration. 



ch05.qxd 4/16/04 3:25 PM Page 124 



124 I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 

Snyder WH III, Thai ER, Bridges RA, et al: The raphy and subsequent operative exploration, 

validity of normal arteriography in penetrating Compared to operative exploration, arteriogra- 

trauma. Arch Surg 1978;1 13(4) :424-426. phy had 36 true positives, 132 true negatives, 14 

Landmark paper comparing arteriography to false positives, and 1 false negative. The authors 

operative vessel exploration. One hundred conclude that arteriography is sensitive enough 

seventy-seven patients with 183 penetrating to exclude arterial injury in patients with 

extremity wounds were evaluated with arteriog- equivocal clinical signs of vascular injury. 



ch06.qxd 4/16/04 3:28 PM Page 125 




Vascular Diagnostic Options in 
Extremity and Cervical Trauma 



KAJ JOHANSEN 



O INTRODUCTION 

O PENETRATING AND BLUNT TRAUMA TO THE EXTREMITIES 

A Historical Perspective 

Noninvasive Physiologic Vascular Tests 

Limb Swelling and Pain following Extremity Revascularization 

O PENETRATING OR BLUNT INJURIES TO THE BRACHIOCEPHALIC 
VESSELS 

Physical Examination 

O SUMMARY 



INTRODUCTION 



Sometimes the diagnosis of extremity arter- 
ial trauma is straightforward: Torrential hem- 
orrhage, acute limb ischemia, a pulsatile 
hematoma, or other such urgent problems 
generally require little attention to dia- 
gnostic maneuvers other than immediate 
operation. However, a large proportion of 
peripheral vascular injuries may not be imme- 
diately apparent, presenting in subtle, con- 
fusing, or obscure fashion. Alternatively, 
vascular injuries may be entirely silent, 



discovered only during a general diagnostic 
survey of the trauma patient. This chapter elu- 
cidates currently accepted "best practices" 
for the diagnosis of extremity and cervical 
vascular trauma. 



PENETRATING AND BLUNT 
TRAUMA TO THE EXTREMITIES 

Injury to the major arteries of the extremities 
may result in severe bleeding or in immedi- 
ate or delayed ischemia — in either case, a 

125 



ch06.qxd 4/16/04 3:28 PM Page 126 



126 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



"SEVEN Ps" OF ACUTE ARTERIAL INSUFFICIENCY 

Pain 

Paresthesia 

Pulselessness 

Pallor 

Poikilothermia (or Polar) 

Paralysis 

(Past midnight!) 



threat to limb viability. Further, as is noted later, 
technically successful revascularization of the 
traumatized extremity may threaten extrem- 
ity viability anew by producing an ischemia- 
reperfusion phenomenon, manifested 
clinically by compartment syndrome. 

Patients with substantial ongoing external 
bleeding, rapidly expanding hematoma, 
evidence for acute arterial insufficiency — the 
"Seven Ps" (Box 6-1) — or other less common 
objective signs of major arterial injury (e.g., 
the presence of a large arteriovenous [AV] 
fistula) almost always warrant immediate 
operation as the first diagnostic test. Other 
diagnostic tests such as arteriography are dila- 
tory and rarely add useful decision-making 
information (occasional patients, such as 
those with shotgun injuries or with extremity 
fractures at multiple levels, may require 
imaging studies preoperatively to define the 
precise anatomic site of arterial disruption) . 
The need for immediate operation is obvious 
in patients with exsanguinating hemorrhage, 
and equivalent urgency is present in patients 
with acute arterial insufficiency, in whom 
experimental and clinical data as well as long- 
established clinical observation document at 
most a 6-hour "grace period" to restore per- 
fusion before a substantially increased likeli- 
hood of postoperative tissue infarction and 
limb loss can be anticipated. This acceptable 
"golden period" may be even shorter if shock, 
crush injury, or other comorbidities compli- 
cate the clinical picture. 

Unfortunately, most patients with extrem- 
ity trauma do not have "hard" signs of 



vascular injury but only "soft" indications that 
arterial or venous injury has occurred. Alter- 
natively, they may have no evidence to support 
the possibility that a vascular injury is present, 
but only clinical suspicion based on the mecha- 
nism of injury or on proximity of the injury 
tract to important vessels. 



A Historical Perspective 

The "gold standard" for making the diag- 
nosis of occult extremity injury has changed 
substantially during the professional lifetime 
of many still-active clinicians. Wound explo- 
ration was the norm in the 1950s and 1960s 
and had even been mandated in the battle- 
field setting. However, the morbidity of this 
approach (including the extremely low yield 
of mandatory wound exploration) became 
clear and resulted in a switch to the use of 
contrast arteriography (usually by a trans- 
femoral arterial approach) in the 1970s and 
1980s. This approach was fueled by the 
general belief (promoted by experienced 
trauma surgeons at several major trauma 
centers) that physical examination is inade- 
quately accurate for the assessment of vascu- 
lar status in injured extremities (Perry, Thai, 
and Shires, 1971). Contrast arteriography 
proved relatively rapid and extremely sensi- 
tive and specific for the identification of arte- 
rial disruption in trauma victims; several 
studies documented false-positive and false- 
negative rates of less than 2% in contrast 
arteriography performed to rule out arterial 



ch06.qxd 4/16/04 3:28 PM Page 127 



6 • VASCULAR DIAGNOSTIC OPTIONS IN EXTREMITY AND CERVICAL TRAUMA 



127 



injury in the extremities (Snyder and col- 
leagues, 1978; Rose and Moore, 1988). 

However, contrast arteriography generally 
requires transfer of the patient out of the emer- 
gency department (ED) to a site in which 
ongoing surveillance, volume resuscitation, 
and management of secondary injuries are 
difficult to conduct. Contrast arteriography 
is invasive and expensive and has a small but 
definite risk of contrast dye reactions or arte- 
rial puncture-site complications. Most impor- 
tantly, it became clear that the real clinical 
yield of contrast arteriography, when per- 
formed as the screening technique of choice 
in injured extremities, is extremely low. In 
several series, fewer than 5% of patients actu- 
ally required operative intervention for arte- 
rial injuries discovered by means of contrast 
arteriography (Frykberg and colleagues, 1989; 
Anderson and colleagues, 1990). 

Noninvasive Physiologic 
Vascular Tests 

In the 1980s, some clinicians began to use 
several noninvasive diagnostic techniques in 
the acute setting, which had previously been 
found to be of major diagnostic value in the 
assessment of chronic arterial occlusive 
disease. These included measurement of 
Doppler-derived arterial pressure indexes 
(APIs) and the use of duplex sonography. 
These two techniques have proven extremely 
useful as initial screening tests for patients 
thought potentially to harbor an occult 
extremity arterial injury. 

Lynch andjohansen (1991) initially demon- 
strated that among 100 injured limbs in 93 
patients, in whom both Doppler APIs and con- 
trast arteriography were carried out, a Doppler 
API of 0.90 had a sensitivity of 87% and a speci- 
ficity of 95% for arterial injury. Because two 
(2%) of the contrast arteriograms were actu- 
ally falsely positive in circumstances in which 
the ultimate outcome had been accurately pre- 
dicted by the Doppler API, the sensitivity and 
specificity of the Doppler API technique was 
actually even higher — 95% and 97% — when 
clinical outcome was used as the comparison 
standard. The negative predictive value for an 
API of more than 0.90 was 99%. 



In a subsequent study, Johansen and 
colleagues (1991) evaluated 100 consecutive 
limbs in 96 vascular trauma victims by screen- 
ing Doppler API; arteriography was reserved 
for patients in whom Doppler API was less than 
0.90. In this series, 83 limbs had a normal API 
(>0.90) and 17 limbs had an abnormal API 
and underwent contrast arteriography. The 
patients with a normal Doppler API were fol- 
lowed up clinically and by duplex sonography; 
none required further vascular intervention, 
and all (except for two patients who under- 
went normal arteriograms as a protocol 
violation) had been spared contrast 
arteriography. Among the 17 limbs undergo- 
ing contrast arteriography, 16 (94%) arterial 
abnormalities were found and seven under- 
went operative intervention. 

These studies demonstrated that use of 
Doppler API could substantially reduce the 
number of "exclusion" contrast arteriograms 
performed in our trauma center (an 80% 
reduction compared with the 12-month 
period before the trial, P<.01), markedly 
increase the diagnostic "yield" when contrast 
arteriography was required, facilitate the 
overall management of most patients not 
requiring contrast arteriogram, and save a sub- 
stantial sum in hospital charges (Lynch and 
Johansen, 1991; Johansen and colleagues, 
1991). 

Many of the trauma victims in the studies 
in which Doppler API was validated were the 
victims of penetrating trauma. For this 
reason, some trauma specialists — especially 
orthopedic surgeons — were reluctant to 
accept Doppler API as a screening test for 
proximal extremity injuries in blunt trauma — 
fractures, dislocations, and crush injuries. 
Accordingly, Cole and colleagues (1999) 
recently conducted a study of 70 trauma 
victims (75 limbs) who had suffered fractures 
and dislocations around the knee. Among 
these patients, Doppler API was normal in 57 
limbs and abnormal in 18. By clinical outcome 
(including duplex scan in about one third of 
the patients) , no late abnormalitieswere iden- 
tified in the individuals who had an initially 
normal Doppler API. In those with a Doppler 
API of less than 0.90, contrast arteriography 
was performed in 16 (88%) and was positive 
in 14 (87% of arteriograms) ; operative repair 



ch06.qxd 4/16/04 3:28 PM Page 128 



128 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



was performed in 6 (33%). The negative pre- 
dictive value of a normal Doppler API in these 
bluntly traumatized lower extremities was 
100% (Cole and colleagues, in press), sug- 
gesting that Doppler API is as accurate and 
useful a screening tool in blunt extremity 
trauma as it is for penetrating injury. 

Studies from other trauma centers regard- 
ing the utility of Doppler API in screening 
through the extremities for a potential arte- 
rial injury came to similar conclusions 
(Schwartz and colleagues, 1993; Frykberg, 
1995). Schwartz and colleagues (1993) use a 
higher threshold Doppler API of 1.0, thereby 
slightly increasing sensitivity at the expense 
of a substantially higher number of negative 
contrast arteriograms. 

The limitations of Doppler API as a screen- 
ing tool for occult arterial injury in the 
extremities must also be made clear. The tech- 
nique does not accurately diagnose damage 
in branch arteries (e.g., the profunda femoris 
or profunda brachii arteries) , cannot accu- 
rately detect small intimal flaps, AV fistulas or 
pseudoaneurysms, and will not, of course, 
discover significant venous injuries. This diag- 
nostic technique is clearly much less accurate 
in the interrogation of arteries proximal to 
the inguinal or axillary crease, for example, 
the iliac or subclavian/axillary arteries; such 
vessels are best evaluated by contrast arteri- 
ography. The Doppler API technique can be 
"fooled" (as can contrast arteriography) by 
arterial spasm and may not detect arterial 
lacerations. 

Accordingly, Doppler API has been vali- 
dated as an accurate, rapid, inexpensive, and 
noninvasive bedside screening examination 
for the purposes of initial screening of a bluntly 
or sharply injured extremity for occult arter- 
ial injury. 

The excellent diagnostic capabilities of 
duplex sonography — pulsed wave Doppler 
ultrasound — became clear in the mid 1980s, 
again in the evaluation of carotid (and other 
types of) atherosclerosis. This technology has 
been thoroughly assessed as being portable, 
rapid, and noninvasive in the evaluation of 
trauma victims. 

Panetta and colleagues (1992) demon- 
strated, in a carefully performed compara- 
tive study of duplex sonography and 



arteriography among different types of exper- 
imental arterial injuries, that when evaluated 
by blinded observers, duplex sonography was 
overall more accurate than contrast arteriog- 
raphy in diagnosing arterial disruption 
(P< .02), especially for arterial lacerations 
CP<.001). 

Among 89 patients with 93 sites of extrem- 
ity or cervical trauma, Meissner, Paun, and 
Johansen (1991) demonstrated that duplex 
sonography resulted in only four false posi- 
tives and no false negatives for significant 
arterial injury. A similar study in 198 trauma 
patients by Bynoe and colleagues (1991) 
from the University of South Carolina found 
only two false-positive and one false-negative 
study. In this study, sensitivity was 95% and 
specificity was 99% for arterial injury. 

These studies might appropriately be crit- 
icized because arteriography control was not 
consistently carried out. Fry and colleagues 
(1993) conducted a trial in patients with 
extremity trauma in which the first 50 sub- 
jects who were studied by Doppler ultra- 
sound also underwent contrast arteriography 
(or operative exploration). When perfect 
agreement was discovered between ultra- 
sonographic and arteriographic diagnostic 
modalities in these patients, a subsequent 1 75 
patients were studied by duplex scan alone, 
with arteriography reserved for patients with 
an abnormal ultrasonographic study. This trial 
of duplex scan showed 100% sensitivity and 
97% specificity for major arterial trauma; only 
one false-positive study resulted. In addition, 
the investigators reported discovery of seven 
major venous injuries by means of duplex 
sonography, which they asserted would not 
have been identified had only contrast arte- 
riography been performed as a diagnostic tool 
(Fry and colleagues, 1993). 

The advantages of duplex scanning as a 
screening examination for arterial injury in 
a patient with extremity trauma are obvious; 
it can be brought to the bedside in the ED 
and is noninvasive, rapid, easily repeated, and 
inexpensive. In addition, certain injuries not 
readily identified by other means — for 
example, major venous disruptions — may 
potentially be identified by this technique. 

The limitations of duplex sonography must 
also be emphasized. These include potentially 



ch06.qxd 4/16/04 3:28 PM Page 129 



6 • VASCULAR DIAGNOSTIC OPTIONS IN EXTREMITY AND CERVICAL TRAUMA 



129 



reduced access because of open wounds, dress- 
ings, splints, or casts; relatively lesser accuracy 
in identifying truncal vascular injury; and a 
substantial "learning curve "for technologists 
and interpreting physicians. In addition, at a 
time of continued economic retrenchment by 
urban trauma hospitals, it has become increas- 
ingly difficult to sustain night/weekend 
vascular laboratory coverage, obligatory for 
evaluation of trauma victims in the ED. 

Do all arterial injuries need to be repaired? 
Although some continue to adhere to the 
traditional tenet that any arterial disruption 
warrants operative exploration (Stain and 
colleagues, 1993), more contemporary 
studies, based on the excellent natural history 
data which can be accumulated from repeated 
duplex ultrasonograms of various arterial 
injuries, have suggested that many "minor" 
arterial injuries — small intimal flaps, pseu- 
doaneurysms, and AV fistulas — resolve on 
their own without intervention. The obser- 
vation that most intimal injuries that result 
from catheter arteriography go on to "heal" 
without operation certainly predicts such a 
conclusion. Stain and colleagues (1993) 
observed 80 "minimal" pseudoaneurysms, 
intimal flaps or dissections, and AVfistulas with 
serial duplex ultrasound examinations; at the 
end of 12 months, only 4 (5%) of the lesions 
had required operative repair. 

Thus, the noninvasive vascular physiologic 
examinations of Doppler API measurement 
and duplex sonography can be employed with 
accuracy and cost-effectiveness in the evolu- 
tion of extremity arterial injuries. We have 
developed an algorithm (Fig. 6-1) that incor- 
porates these modalities. 

Limb Swelling and Pain 
following Extremity 
Revascularization 

The unwary clinician may ignore the fact 
(or fail to recall) that prolonged or severe limb 
ischemia followed by successful revasculari- 
zation can result in the ischemia-reperfusion 
phenomenon, manifested clinically as com- 
partment syndrome. This may be seen par- 
ticularly after crush injuries, combined arterial 
and venous trauma, closed fractures of the 



Significant bleeding or ischemia, or shotgun wound? 



Yes 




Doppler pressure 
measurement 



APK0.90 API > 0.90 



Arteriogram 
(or operation) 



Observation 

(serial exams, ? 

noninvasive imaging?) 



■ FIGURE 6-1 

Algorithm for diagnostic management of 
patients potentially harboring an extremity 
vascular injury. ■ 



extremity, and ischemia complicated by sys- 
temic hypotension or shock. Compartment 
syndrome, if not recognized and treated in a 
timely fashion, is associated with a substantial 
risk of myonecrosis and limb loss. 

In the otherwise uncomplicated trauma 
victim, the diagnosis of compartment syn- 
drome is usually straightforward. Such 
patients have pain out of proportion to what 
would be expected, as well as inexorably wors- 
ening neurologic dysfunction of the extrem- 
ity as characterized by both numbness and 
extensor weakness. Calf or forearm muscles 
(for practical purposes, the only two sites 
where compartment syndrome normally pre- 
sents) will be unnaturally tight, swollen, and 
tender. Thus, the diagnosis of compartment 
syndrome is not difficult, given the appro- 
priate clinical scenario and the symptoms and 
signs noted earlier. 

However, relevant symptoms and signs may 
be obscured by one or more of a constella- 
tion of comorbid conditions. These may 
include intoxication with alcohol or other 
drugs, closed head injury, general or neuraxial 
anesthesia, spinal cord injury resulting in para- 
plegia or quadriplegia, or obscuration of the 
examination by casts, splints, or dressings. In 
such patients, the diagnosis of compartment 
syndrome may be obscured or ignored until 
muscle necrosis has already occurred. 

One approach, espoused by many, is to 
adopt a liberal posture toward the 



ch06.qxd 4/16/04 3:28 PM Page 130 



130 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



performance of prophylactic fasciotomy when 
even the possibility of compartment syn- 
drome is contemplated. I am a proponent of 
such a view. However, in certain clinical set- 
tings, such an approach might be imprudent 
or unwarranted, and alternative dependable 
means of making the diagnosis of compart- 
ment tissue hypertension are required. 

The time-honored technique is that of tissue 
manometry, using various commercially avail- 
able devices to transduce tissue pressure after 
insertion of a needle into a given compart- 
ment of the calf or forearm. This technique, 
in use since the mid 1970s (Whiteside and col- 
leagues, 1975; Matsen, 1978) is both sensitive 
and specific for compartmental hypertension; 
its only drawbacks are its invasive nature (thus 
making it difficult to do repeat studies) and 
the fact that devices for measuring may occa- 
sionally be unavailable, out of commission, or 
unfamiliar to the clinician. Tissue pressures 
higher than 40mmHg or higher than 
30 mm Hg for longer than 3 hours have been 
considered diagnostic of compartmental 
hypertension and are an indication for imme- 
diate fasciotomy (Matsen, 1978). A more 
contemporary understanding of the patho- 
physiology of compartmental hypertension 
compares the measured compartment pres- 
sure to either the diastolic or the calculated 
mean arterial pressure, with compartmental 
hypertension being characterized by a pres- 
sure differential of less than 30 mm Hg 
(Matava and colleagues, 1994). 

Other techniques, such as the measurement 
of somatosensory evoked potentials (Present 
and colleagues, 1993), the use of near-infrared 
spectrophotometry (Giannotti and col- 
leagues, 2000), and the objective measure- 
ment of tissue hardness (Steinberg and 
Gelberman, 1994) have been assessed. 

A clever conceptual leap by Jones, Perry, 
and Bush (1989), experimentally validated by 
Ombrellaro and colleagues (1996), permits 
the assessment of effects of tissue pressure on 
calf or forearm venous hemodynamics by use 
of venous duplex scanning. Arterial pressures 
and flows should clearly be only minimally (if 
at all) impacted by changes in ambient tissue 
pressure. However, venous hemodynamics 
should be exquisitely sensitive to local tissue 
pressure: Normal venous pressures in the calf 



and forearm are virtually identical to tissue 
pressures (5 to 10 mmHg). Whereas the sen- 
sitivity of duplex scanning of the calf or 
forearm veins for compartment syndrome may 
not be particularly high (i.e., abnormal venous 
hemodynamics might be due to crush injury, 
hematoma, splints or dressings, or compart- 
mental hypertension) , the specificity of exam- 
ination should be quite close to 100% (i.e., 
normal tibial venous respiratory variation and 
phasi city in a particular calf compartment indi- 
cate that compartmental hypertension cannot 
be present). Duplex scanning of tibial or 
forearm veins is the screening test of choice 
at my medical center in patients in whom 
the diagnosis of compartment syndrome is 
entertained. 



PENETRATING OR BLUNT 
INJURIES TO THE 
BRACHIOCEPHALIC VESSELS 



Among many other complications of 
trauma to the head, neck, and upper chest is 
the possibility of injury to the large arteries 
and veins of the head and upper extremities. 
Not only is early or delayed exsanguination a 
risk, but the late implications of arterial 
thrombosis or embolization secondary to 
dissection, intimal flap, or pseudoaneurysm 
include the risk of disabling or even lethal 
stroke. An ongoing controversy attends the 
question of the proper diagnostic pathway to 
be followed in patients with penetrating cer- 
vical trauma — mandatory exploration or selec- 
tive operation based on the results of a panel 
of diagnostic tests (arteriography, triple 
endoscopy, barium swallow). This dispute 
remains undecided despite careful ongoing 
evaluation over the last 4 decades. 

As for other anatomic sites in the body, signs 
of significant hemorrhage (pulsatile external 
or oropharyngeal bleeding, expanding/ 
pulsatile hematoma) mandate immediate 
operation. In addition, it has become increas- 
ingly clear that cervical vascular injury asso- 
ciated with any degree of neurologic deficit 
warrants emergency extracranial carotid (or 
vertebral) arterial reconstruction as well 
(Richardson and colleagues, 1992). Although 



ch06.qxd 4/16/04 3:28 PM Page 131 



6 • VASCULAR DIAGNOSTIC OPTIONS IN EXTREMITY AND CERVICAL TRAUMA 



131 



the results of such surgical repair in patients 
with carotid artery injury associated with coma 
are generally dismal, case reports suggest that 
even in this desperate setting, occasional cere- 
bral salvage can occur by timely carotid arte- 
rial repair (Robbs and colleagues, 1983) . 

As elsewhere, much more major controversy 
attends the management of patients with "soft" 
signs of cervical carotid (or vertebral) arter- 
ial injury or in whom there is concern about 
an occult arterial injury based on the sound- 
ing mechanism or the location of the injury 
tract. Because such wounds potentially also 
involve the airway or (as seriously) the esoph- 
agus or the oropharynx, either routine opera- 
tive exploration as the first diagnostic test or 
a series of radiographic, endoscopic, and arte- 
riographic studies is obligatory in this setting. 
As noted already, no clear consensus has been 
achieved in defining the superiority of one 
diagnostic approach over the other. 

Detection of occult cervical arterial injuries 
is assisted by now well-established means of 
categorizing such potential injuries by 
anatomic site. It is generally agreed that 
injuries below the sternal notch (zone 1) will 
likely require median sternotomy or thora- 
cotomy for management; accordingly, pre- 
operative contrast arteriography is generally 
considered necessary. Injuries above the angle 
of the jaw (zone 3), because they are surgi- 
cally remote, may require craniotomy or 
(potentially) even more complex vascular 
exposure; contrast arteriography is thus indi- 
cated in this setting as well. Arteriography for 
lesions in zone 3 also occasionally identifies 
lesions surgically inaccessible enough (e.g., 
in the carotid siphon) so that treatment by 
catheter-directed means (e.g., coil emboliza- 
tion of carotid artery-cavernous sinus fistula) 
is the preferred therapeutic choice. 

In zone 2 penetrating injuries to the neck, 
duplex sonography has been found to play a 
highly dependable diagnostic role. Fry and 
colleagues (1994), using control arteriogra- 
phy "run-in"for the first 15 patients in a series 
of 100 patients, demonstrated equivalent 
100% sensitivities and specificities for duplex 
sonography in examining cervical carotid 
injuries. Demetriades and colleagues (1995) 
compared contrast arteriography, duplex 
sonography, and simple serial physical 



examination in 82 patients with penetrating 
neck trauma. As previously demonstrated with 
peripheral arterial injuries in the extremities, 
contrast arteriography was "hypersensitive "for 
arterial injuries; 11 arterial disruptions were 
discovered, but only 2 required operation. 
Physical examination was accurate for all clin- 
ically significant injuries but missed six minor 
vascular injuries. Duplex sonography found 
10 of the 1 1 injuries detected by contrast arte- 
riography, and sensitivity and specificity for 
clinically relevant injuries were 91 % and 99%, 
respectively. 

Blunt cervical arterial injuries are even more 
complicated to diagnose, because unlike pen- 
etrating trauma (which usually involves the 
common carotid artery), blunt trauma more 
commonly involves the internal carotid artery. 
Closed head injury, basilar skull fractures, 
various forms of deceleration motor-vehicle 
accidents resulting in injuries to the head and 
neck, and the increased use of shoulder-lap 
restraints have resulted in a sharply increased 
recent incidence of blunt carotid injury 
(Kerwin and colleagues, 2001 ) . Because these 
patients frequently present without initial 
symptoms only later developing neurologic 
deficits related to thrombosis or dissection of 
the internal carotid artery, a screening tech- 
nique that is accurate and rapid would clearly 
be of use in this clinical scenario. 

Duplex sonography may play a substantive 
role as a screening tool in this setting. As 
demonstrated in the landmark animal studies 
by Panetta and colleagues (1992) , duplex scan- 
ning is as accurate as contrast arteriography 
in the diagnosis of dissections, intimal flaps, 
and arterial lacerations. In a large series of 
patients with blunt cervical trauma, Fabian 
and colleagues (1996) demonstrated the diag- 
nostic value of duplex sonography in detect- 
ing occult arterial injuries. Because logistic 
regression analysis of the data in this study 
demonstrated independent survival benefit 
associated with heparinization in those 
patients not requiring operation, the value of 
early diagnosis of such initially silent injuries 
(either by sonography or by contrast arteri- 
ography) is abundantly clear. 

Contrast-enhanced cervical computed 
tomographic (CT) scanning with fine cuts at 
the cervical level was demonstrated by Zeman 



ch06.qxd 4/16/04 3:28 PM Page 132 



132 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



and colleagues (1995) at the University of 
Vermont to be more accurate even than ultra- 
sonography or arteriography in determining 
whether a bluntly traumatized or dissected 
carotid artery is patent or not. Indeed, because 
many such patients undergo head CT scans 
to rule out concurrent cerebral injuries, an 
effective strategy might potentially be to con- 
tinue the scan down to the level of the carotid 
bifurcation, thereby demonstrating in a few 
extra minutes whether the internal carotid 
arteries are intact and obviate the need to 
perform either duplex sonography or four- 
vessel cerebral arteriography. 

Transcranial Doppler (TCD) studies have 
been used only rarely in the trauma setting. 
However, conceptually this technology may 
play a useful role in selected patients with 
actual or potential cerebrovascular trauma, 
on either a blunt or a penetrating basis 
(Rae-Grant and colleagues, 1996). For 
example, TCD studies can demonstrate the 
adequacy of intracranial arterial flow and can 
contribute to a decision about whether 
extracranial carotid (or vertebral) revascular- 
ization needs to be performed. In addition, 
information regarding whether temporary 
carotid shunting during arterial reconstruc- 
tion should be used can be derived from TCD 
studies. Finally, in patients with devastating 
head injuries (or in whom irreversible brain 
injury is suspected) , TCD is an acknowledged 
means of demonstrating (based on intracra- 
nial arterial flow arrest) that the patient is brain 
dead (Wejdiecks ejection fraction, 2001). 

Physical Examination 

The simplest diagnostic tool is a careful phys- 
ical examination. As previously intimated, the 
adequacy of physical examination in the diag- 
nosis of vascular trauma has been heavily 
debated over the past 3 to 4 decades. Experi- 
enced trauma surgeons at Parkland Hospital 
in Dallas, in a series of reports in the 1970s, 
suggested the relative inaccuracy (or at least 
inadequacy) of physical examination alone in 
patients potentially harboring an arterial 
injury and were in the vanguard of those pro- 
moting the use of routine "exclusion" arteri- 
ography in patients with extremity trauma 



(Perry, Thai, and Shires, 1971; Snyder and 
colleagues, 1978). 

However, it has subsequently become clear 
that physical examination has an important 
role to play, at least as a screening tool for 
various forms of penetrating and blunt extrem- 
ity and cervical trauma. I consider measure- 
ment of Doppler arterial pressure in injured 
extremities and calculation of Doppler API 
to be a simple extension of palpation and other 
aspects of the physical examination; Doppler 
API has been found, in studies in which I as 
well as others have participated, to be highly 
sensitive and specific in the diagnosis of 
important forms of flow-limiting lesions of 
extremity arteries (Lynch an djohansen, 1991; 
Johansen and colleagues, 1991; Cole and 
colleagues, in press; Schwartz and colleagues, 
1993; Frykberg, 1995). 

Physical examination has also been found 
to be accurate for important extracranial 
carotid artery injuries in a controlled trial 
comparing serial physical examination with 
duplex sonography and contrast arteriogra- 
phy (Demetriades and colleagues, 1995). 
Coming full circle, Francis and colleagues 
( 199 1 ) from Parkland Hospital confirmed the 
validity of serial physical examination by an 
experienced surgeon in ruling in or out 
extremity vascular injury. 



SUMMARY 

The morbidity and mortality associated with 
major central or peripheral vascular injury 
mandate diagnostic measures that are rapid 
(or at least timely) and accurate. Because of 
the invasiveness and morbidity of contrast 
arteriography and operative exploration, such 
diagnostic maneuvers are optimally rapid, 
portable, noninvasive, repeatable, and inex- 
pensive. 

Contrast arteriography continues to be the 
"gold standard" for establishing the presence 
and anatomic location of arterial injuries 
within the chest or abdomen and in zones 1 
and 3 for penetrating neck trauma. However, 
widespread use of contrast arteriography in 
the diagnosis of vascular injury is hampered 
by its invasiveness, expense, the time taken to 



ch06.qxd 4/16/04 3:28 PM Page 133 



6 • VASCULAR DIAGNOSTIC OPTIONS IN EXTREMITY AND CERVICAL TRAUMA 



133 



perform such studies, and the necessity in most 
cases to carry such studies out in a site remote 
from the ED or the operating room. 

Prospective studies in trauma victims have 
demonstrated the validity of Doppler arterial 
pressure measurement as a sensitive and 
specific screening tool for both penetrating 
and blunt arterial injuries that are axial and 
flow limiting. Duplex sonography subse- 
quently has been found to be as accurate as 
contrast arteriography in the diagnosis of 
extremity arterial injury, and the technique 
has the additional benefit of being able to diag- 
nose major venous injuries of the extremities. 
Duplex ultrasonography has been demon- 
strated to have diagnostic accuracy equivalent 
to that of contrast arteriography in penetrat- 
ing zone 2 injuries of the neck and has equiv- 
alently excellent diagnostic accuracy in blunt 
trauma to the extracranial carotid and verte- 
bral arteries. Detection of patency of the inter- 
nal carotid artery following blunt trauma 
and subsequent dissection is accurately made 
with contrast-enhanced cervical CT scanning, 
a more accurate means of assessing flow 
than either contrast arteriography or duplex 
sonography. 

Although compartment syndrome may be 
best averted by adoption of a liberal policy of 
prophylactic fasciotomy in trauma victims at 
significant risk of developing this compli- 
cation, alternative diagnostic methods may 
include a series of minimally invasive or 
noninvasive techniques measuring tissue pres- 
sure or hardness, neurologic function, or com- 
partment venous hemodynamics. 

Finally, after being long discounted as a valid 
means of diagnosing occult arterial injury, 
serial physical examination has been resur- 
rected as an accurate diagnostic technique by 
well-performed prospective clinical trials. 

REFERENCES 

Abou-Sayed H, Berger DL: Blunt lower-extremity 
trauma and popliteal artery injuries: Revisiting 
the case for selective arteriography. Arch Surg 
2002;137(5):585-589. 

Anderson RJ, Hobson RW, Padberg FT, et al: 
Reduced dependency on arteriography for pen- 
etrating extremity trauma (PET): Influence of 
wound location and non-invasive vascular studies. 
J Trauma 1990;30:1059-1065. 



Barnes CJ, Pietrobon R, Higgins LD: Does the pulse 
examination in patients with traumatic knee dis- 
location predict a surgical arterial injury? A meta- 
analysis. J Trauma 2002;53(6):1109-1114. 

Bynoe RP, Miles WS, Bell RM, et al: Noninvasive 
diagnosis of vascular trauma by duplex ultra- 
sonography. J Vase Surg 1991;14:346-350. 

Cole P, Campbell R, Swiontkowski M, Johansen K: 
Doppler arterial pressures reliably exclude occult 
arterial injury in blunt lower extremity trauma. 
J Orthop Trauma (in press). 

Demetriades D, Theodorou D, Cornwell E, et al: 
Penetrating injuries of the neck in patients in 
stable condition: Physical examination, angiog- 
raphy or color flow Doppler imaging. Arch Surg 
1995;130:971-975. 

Fabian TC, Patton JH Jr, Croce MA, et al: Blunt 
carotid injury: Importance of early diagnosis and 
anticoagulant therapy. Ann Surg 1996;223:513- 
522. 

Francis H III, Thai ER, Weigelt JA, Rodman HC: 
Vascular proximity: Is it a valid indication for arte- 
riography in asymptomatic patients? J Trauma 
1991;31:512-514. 

FryWR, DortJA, Smith RS, etal: Duplex scanning 
replaces arteriography and operative explo- 
ration in the diagnosis of potential cervical 
vascular injury. Am J Surg 1994;168:693-695. 

Fry WR, Smith RS, Sayers DV, et al: The success of 
duplex ultrasonographic scanning in the diag- 
nosis of extremity vascular proximity trauma. 
Arch Surg 1993;128:1368-1372. 

Frykberg EP: Advances in the diagnosis and treat- 
ment of extremity vascular trauma. Surg Clin 
North Am 1995;75:207-215. 

Frykberg ER, Crump JM, Vines FS, et al: A reassess- 
ment of the role of arteriography in assessing 
acute vascular injuries. J Trauma 1989;29:1041- 
1052. 

Giannotti G, Cohn SM, Brown M, et al: Utility of 
near-infrared spectroscopy in the diagnosis of 
lower extremity compartment syndrome. 
J Trauma 2000;48:397-399. 

Johansen K, Lynch K, Paun M, Copass MK: 
Non-invasive vascular tests reliably exclude occult 
arterial trauma in injured extremities. J Trauma 
1991;31:515-522. 

Jones WG II, Perry MO, Bush HL Jr: Changes in 
tibial blood flow in the evolving compartment 
syndrome. Arch Surg 1989;124:801-804. 

Kerwin AJ, Bynoe RP, Murray J, et al: Liberalized 
screening for blunt carotid and vertebral artery 
injuries is justified. J Trauma 2001;51:308-314. 



ch06.qxd 4/16/04 3:28 PM Page 134 



134 



I • GENERAL PRINCIPLES OF VASCULAR TRAUMA 



Lynch K,Johansen KH: Can Doppler pressure mea- 
surements replace "exclusion" arteriography in 
extremity trauma? Ann Surg 1991;214:737- 
741. 

Matava MJ, Whiteside TE Jr, Seiler JG III, et al: 
Determination of the compartment pressure 
threshold of muscle ischemia in a canine model. 
J Trauma 1994;37:50-58. 

Matsen FA III: Compartmental syndrome : a unified 
concept. Clin Orthop 1978;113:8-13. 

Meissner M, Paun M, Johansen K: Duplex scanning 
for arterial trauma. AmJ Surg 1991 ;161 :552-555. 

Ombrellaro MP, Stevens SL, Freeman ML, et al: 
Ultrasound characteristics of lower extremity 
venous flow for the early diagnosis of compart- 
ment syndrome: An experimental study. J Vase 
Technol 1996;20:71-75. 

Panetta TF, Hunt JP, Buechter KJ, et al: Duplex 
ultrasonography versus arteriography in the 
diagnosis of arterial injury: An experimental 
study. J Trauma 1992;33:627-635. 

Perry MO: Complications of missed arterial injuries. 
J Vase Surg 1993;17:399-403. 

Perry MO, Thai ER, Shires GT: Management of 
arterial injuries. Ann Surg 1971;173:403-408. 

Present DA, Nainzedeh NK, Ben-Yishay A, Mazzara 
JT: The evaluation of compartmental syndromes 
using somatosensory evoked potentials in 
monkeys. Clin Orthop 1993;287:376-285. 

Rae-Grant AD, Eckert N, Barbourt PJ, et al: 
Outcome of severe brain injury: A multimodal- 
ity neurophysiologic study. J Trauma 1996;40:401- 
407. 



Richardson R, Obeid FN, Richardson JD, et al: Cere- 
brovascular injury: Neurologic consequences. 
J Trauma 1992;32:755-760. 

Robbs JV, Human RR, Rajaruthnam P, et al: Neu- 
rological deficit and injuries involving the neck 
arteries. Br J Surg 1983;70:220-222. 

Rose SC, Moore EE: Trauma angiography: The use 
of clinical findings to improve patient selection 
and care preparation. JTrauma 1988;28:240-245. 

Schwartz MR, Weaver FA, Yellin AE, et al: Refining 
the indications for arteriography in penetrating 
extremity trauma: A prospective analysis. J Vase 
Surg 1993;17:166-170. 

Snyder WH III, Thai ER, Bridges RA, et al: The 
validity of normal arteriography in penetrating 
trauma. Arch Surg 1978;113:424-426. 

Stain SC, Yellin AE,WeaverFA,etal: Selective man- 
agement of nonocclusive arterial injuries. Arch 
Surg 1989;124:1136-1140. 

Steinberg BD, Gelberman RH: Evaluation of limb 
compartments with suspected increased inter- 
stitial pressure: A non-invasive method for deter- 
mining quantitative hardness. Clin Orthop 
1994;300:248-253. 

Wejdiecks EF: The diagnosis of brain death. N Engl 
J Med 2001;344:1215-1221. 

Whiteside TE Jr, Haney TC, Morimoto K, Harada 
H: Tissue pressure measurements as a determi- 
nant for the need of fasciotomy. Clin Orthop 
1975;113:43-51. 

Zeman RK, Silverman PM, Vieco PT, Costello P: 
CT angiography. AJR Am J Roentgenol 1995; 
165:1079-1088. 



ch07.qxd 4/16/04 3:29 PM Page 137 





Access, Control and Repair 



Techniques 

KENNETH L. MATTOX 
ASHER HIRSHBERG 



O POSITIONING 

O INITIAL HEMORRHAGE CONTROL 

General Principles of Vascular Control 
Adjuncts to Hemorrhage Control 
O EXPOSURE AND CONTROL OF SPECIFIC INJURIES 
Neck 

Thoracic Outlet 
Chest 

Upper Extremity 
Axillary Artery 
Brachial Artery 
Abdomen and Pelvis 
Lower Extremity 
Groin 

Distal Superficial Femoral Artery 
Popliteal Artery and Branches 
O REPAIR PRINCIPLES AND TECHNIQUES 
O SUMMARY 



137 



ch07.qxd 4/16/04 3:29 PM Page 138 



138 



II • PRINCIPLES OF OPERATIVE CARE 



The repair of vascular injuries is one of 
the most challenging aspects of trauma 
for the surgeon. In the massively bleed- 
ing patient with a major vascular injury, rapid 
and effective exposure and control of the 
bleeding vessel often mark the difference 
between a spectacular save and on-table death 
from exsanguination. Despite recent advances 
in trauma systems, prehospital care, operative 
techniques, and critical care, many critically 
injured patients still die in the operating room 
(OR) from uncontrolled hemorrhage from 
major vessels. Vascular trauma is especially 
demanding also because there is a very narrow 
margin for technical and judgment errors. 
Compared with gastrointestinal injuries, 
for example, vascular repairs are much less 
forgiving and less tolerant of technical 
imperfection. 

The operative sequence in vascular trauma 
consists of access, exposure, control, and 
repair. While the specific techniques used in 
addressing individual injuries are described 
in other chapters in this book, this chapter 
addresses the general principles underlying 
the operative approach to injuries to blood 
vessels. Our purpose is therefore to provide, 
in one location, a single comprehensive 



reference to operative principles in vascular 
trauma, with special emphasis on universal 
considerations that form the foundation for 
the control and repair of vascular injuries. 



POSITIONING 



Correct positioning of the injured patient on 
the operating table and an accurate definition 
of the operative field are the keys to a smooth 
operative procedure. Incorrect positioning 
can turn a straightforward operation into a 
technical nightmare, severely limits the 
surgeon's options, and reflects lack of under- 
standing of the ramifications and potential sce- 
narios into which the procedure may evolve. 
The "generic" position of the trauma patient 
in the OR is in the supine position, with both 
armsfully extended (Fig. 7-1). One of the car- 
dinal principles in trauma surgery is that the 
surgeon must be prepared to rapidly shift his 
or her attention to another visceral cavity, so 
the potential operative field for truncal trauma 
extends from the chin to below the knees and 
as far laterally as the posterior axillary lines 
on both sides, even if the initial procedure is 



■ FIGURE 7-1 

Drawing depicting supine 
and right decubitus (left chest 
up) positions. ■ 




ch07.qxd 4/16/04 3:29 PM Page 139 



7 • ACCESS, CONTROL AND REPAIR TECHNIQUES 



139 



focused on the abdomen or the chest. When 
the operative procedure is limited to an 
extremity, the surgeon must still be prepared 
for an unexpected deterioration that may 
require access to the chest (for chest tube 
placement) or the groin (for insertion of a 
line). Thus, for example, it would be a bad 
mistake to attempt to use the posterior 
approach to the popliteal artery in the trauma 
patient, because with the patient in the prone 
position, the surgeon's options for interven- 
tions in other anatomic locations are severely 
limited. 

A left posterolateral thoracotomy, which is 
performed with the patient in a right lateral 
decubitus position, is a notable exception. 
Because gaining access to the posterior medi- 
astinal structures (such as the descending 
thoracic aorta or the esophagus) through an 
anterolateral thoracotomy is difficult, it may 
occasionally be necessary to place the injured 
patient in the right lateral decubitus position, 
thus limiting access not only to the con- 
tralateral chest but also to the abdomen or 
the extremities. Choosing the right lateral 
decubitus position in the trauma patient is 
therefore a calculated risk that is typically 
undertaken only after injuries to other visceral 
compartments have been ruled out. 

When positioning the patient for a periph- 
eral vascular repair, the surgeon must keep in 
mind several important principles: The poten- 
tial operative field extends at least one joint 
above and below the injured segment. An 
uninjured lower limb must be included in the 
field to enable rapid harvesting of the saphe- 
nous vein. For injuries in proximity to the 
groin or axilla, considerations of proximal 
control dictate that the abdomen or chest, 
respectively, be included in the operative field. 
Lastly, full mobility of the injured extremity 
within the operative field is mandatory to 
enable the surgeon to adjust the position of 
the relevant vascular segments as the opera- 
tion unfolds. 

In summary, when positioning the patient 
with vascular trauma, it is a good general 
principle to always consider the "worst-case 
scenario." This means not only optimizing 
exposure of the relevant anatomic area but 
also being fully prepared either for a large 
extension of the incision or for an urgent 



intervention in another visceral compart- 
ment (Box 7-1). 



INITIAL HEMORRHAGE 
CONTROL 

Initial control of external hemorrhage, 
whether in the field, emergency department, 
or OR, is one of the first priorities addressed 
during the primary survey of the injured 
patient according to Advanced Trauma Life 
Support principles. Control of external hem- 
orrhage (typically from an injured extremity) 
is usually achieved by simple digital or manual 
compression, which will almost invariably 
control bleeding without damaging adjacent 
elements of the neurovascular bundle. In 
unusual circumstances, such as combat trauma 
care or a mass casualty scenario, an arterial 
tourniquet may be lifesaving, albeit at the price 
of compromising both the collateral circula- 
tion and venous drainage from the injured 
extremity. 

The classic error in temporary control of 
external hemorrhage is an attempt to use sur- 
gical instruments (such ashemostats) instead 
of digital pressure to obtain control in the 
field or in the emergency department. Blind 
groping withhemostats in the face of ongoing 
torrential hemorrhage is not only ineffective 
but also likely to result in iatrogenic damage 
to the adjacent structures of the neurovascu- 
lar bundle and convert a simple partial injury 
into a complete transection with a crushed 
arterial wall. 

Manual compression of the bleeding site 
(usually by a member of the trauma team other 
than the surgeon) should be continuously 
maintained into the OR, until proper proxi- 
mal and distal control is obtained. The com- 
pressing hand should then be prepared in the 
operative field. While the surgeon makes the 
incision to obtain proximal and distal control 
and expose the injured vessel, the first assis- 
tant should maintain external manual pres- 
sure. Selective clamping of the vessel should 
thus be performed under optimal conditions 
in the OR, away from the site of injury and 
using appropriate vascular instruments and 
technique. 



ch07.qxd 4/16/04 3:29 PM Page 140 



140 



II • PRINCIPLES OF OPERATIVE CARE 



INCISIONS USED FOR CONTROL, EXPOSURE, AND REPAIR OF 
VASCULAR INJURY 



Anterior neck, anterior to the sternocleidomastoid muscle 

Supraclavicular 

Infraclavicular 

Combined supraclavicular/infraclavicular 

Axillary 

Inner arm 

Antecubital fossa 

Forearm 

Anterior left second interspace 

Left fourth or fifth posterolateral thoracotomy 

Median sternotomy with anterior cervical or supraclavicular extension 

Midline laparotomy 

Low transverse lateral abdominal (kidney transplant incision) 

Groin incision 

Medial distal thigh 

Medial proximal calf 

Fasciotomy incisions of the extremity 



Balloon catheter tamponade is a very useful 
adjunct to initial control of external hemor- 
rhage, especially for penetrating injuries to 
the groin, clavicular fossa, and axilla, where 
manual pressure is not as effective and a 
tourniquet cannot be applied. A Foley balloon 
catheter is rapidly inserted into the actively 
bleeding tract of a bullet or a stab wound and 
then is inflated. This simple maneuver creates 
local extraluminal compression of the injured 
vessel, which temporarily controls hemor- 
rhage and frees the compressing hand of the 
assistant (Fig. 7-2). 

General Principles of 
Vascular Control 

Definitive control of a major vascular injury 
is the accurate placement of vascular clamps 
on both the inflow and the outflow tract of 
the injured vessel. This cardinal principle of 
obtaining proximal and distal control before 
approaching the injured segment is one of 



the fundamentals of surgery for vascular 
trauma, and its importance cannot be 
overstated. 

Most vascular injuries exhibit some degree 
of tamponade, be it from a hemostatic plug, 
surrounding tissues, local pressure, spasm of 
the injured vessel, or a combination thereof. 
Entering the hematoma without first obtain- 
ing proximal and distal control away from the 
site of injury is the worst mistake a surgeon 
can commit, a mistake that often leads to 
unnecessary blood loss, a disorganized 
attempt to regain control, and sometimes 
exsanguination and death. 

Proximal control is obtained outside the 
hematoma surrounding the injured segment. 
This frequently requires extension of the sur- 
gical incision and dissection through virgin 
tissue planes. An important principle in 
obtaining proximal control is to try and go 
beyond an anatomic structure that serves as 
a natural barrier to the expansion of the 
hematoma. For example, dissection in the 
groin to gain control of an injured common 



ch07.qxd 4/16/04 3:29 PM Page 141 



7 • ACCESS, CONTROL AND REPAIR TECHNIQUES 



141 




Kenneth L. Matlcs, M.D. 



■ FIGURE 7-2 

Drawing depicting the insertion 
of a balloon catheter through 
the site of a lower neck 
penetrating wound to control 
hemorrhage. ■ 



femoral artery is difficult and fraught with 
danger. If the surgeon extends the incision 
cranially and dissects above the inguinal lig- 
ament, he or she discovers that the ligament 
serves as a natural barrier to the expansion 
of the hematoma, and the tissue planes above 
it are much easier to identify. Similarly, the 
pericardium is a barrier to the extension of a 
mediastinal hematoma from an innominate 
artery injury, and the parietal pleura is a 
barrier to the extension of an axillary 
hematoma. 

Occasionally, precise definition of the 
injured vessel is impossible and vigorous 
bleeding presents an immediate and grave 
danger to the patient's life. Under these cir- 
cumstances, an alternative "last-resort" tech- 
nique is application of a large noncrushing 
vascular clamp to the total gross area of active 
hemorrhage, including adjacent structures. 
Once "global" proximal and distal vascular 
control has been achieved, the clamp can 
either be removed or be gradually advanced 



toward the site of the specific injury as dis- 
section proceeds in a relatively bloodless 
field. 

Another important adjunct is the use of 
intraluminal Fogarty balloon catheters. When 
the proximal or distal segment of the injured 
vessel is inaccessible to direct clamping (e.g., 
deep in the pelvis), a useful alternative is the 
insertion of a Fogarty balloon catheter con- 
nected to a three-way stopcock into the orifice 
of the bleeding vessel. Inflation of the balloon 
inside the vessel lumen achieves direct intra- 
luminal hemostasis and obviates the need for 
time-consuming and difficult dissection to 
define the vessel from the outside. 



ADJUNCTS TO 
HEMORRHAGE CONTROL 

An important aspect of hemorrhage control 
is fluid resuscitation. The trauma team 
must direct the resuscitative efforts toward 



ch07.qxd 4/16/04 3:29 PM Page 142 



142 



II • PRINCIPLES OF OPERATIVE CARE 



controlled rather than overaggressive fluid 
administration. A crucial part of initial hem- 
orrhage control is creation of a hemostatic 
plug, a soft clot that is formed at the site 
of injury by the hemostatic mechanisms of 
the body. Dislodgment of this clot is now 
presumed to occur at a lower systemic blood 
pressure than previously appreciated. The 
resuscitating team should therefore avoid 
attempts to achieve a blood pressure at 
"normal" preinjury levels and remember that 
a systolic pressure in the range of 80 mm Hg 
is actually in the bleeding patient's best inter- 
est. Aggressive resuscitation that leads to 
rebleeding and the need for additional fluids 
(the so-called "cyclic hyper-resuscitation") 
creates a dilutional coagulopathy, activates 
inflammatory mediators, and promotes 
further bleeding. Avoiding hypothermia and 
dilution, both of which directly contribute 
to a coagulopathy state, is also very impor- 
tant in enabling effective hemostasis by the 
coagulation cascade. 

The surgeon may elect to use an intravas- 
cular temporary shunt (see Chapter 8) 
as a hemorrhage control technique. When 
inserted into an injured vessel and held in 
place proximally and distally, the shunt effec- 
tively controls hemorrhage while preserving 
distal flow. Topical hemostasis has been an aid 
to hemorrhage throughout history using a 
wide variety of hemostatic agents ranging from 
cellulose, thrombin-like products, and fibrin 
glue. The common denominator of all these 
topical measures is reliance on the body's phys- 
iologic hemostatic mechanisms. The benefit 
of the topical device is not always clear, and 
in the context of vascular trauma, it is never 
a substitute for a carefully placed vascular 
suture. It may however serve as a hemostatic 
adjunct near avascular repair, to help control 
oozing from the suture line or from adjacent 
raw surfaces. During the last decade, topically 
applied "fibrin glue" has been used in liver, 
spleen, and raw surface bleeding. A dry fibrin 
dressing technology, based on thrombin 
powder, is under development, which has 
shown promise in the laboratory as being 
able to stop bleeding even from medium-size 
arteries and veins. 

Recent research focuses on enhancing the 
body's physiologic clotting mechanism in 



areas of endothelial disruption. Recombinant 
activated factor Vila, a hemostatic product 
used in the treatment of hemophilia, recently 
has been successful in treating coagulopathic 
bleeding in critically injured patients. Several 
case reports have led to laboratory studies 
in animal models. If proven effective in con- 
trolled clinical trials, this agent may represent 
a paradigm shift from external control to 
initiating focused clotting "from within." 



EXPOSURE AND CONTROL 
OF SPECIFIC INJURIES 

Gaining access and exposure to perform 
precise reconstruction is a surgical art form 
made possible by a detailed knowledge of 
surgical anatomy, experience, andjudgment, 
as well as an intuitive ability to rapidly access 
difficult areas without adding iatrogenic 
injury. In vascular trauma, there are times 
when aggressive blunt dissection is necessary 
to achieve access and control, while other 
situations demand the most delicate touch 
in dissecting, exposing, and repairing a 
complex injury. Nowhere else in surgery is 
this dichotomy of the surgical craft more 
obvious than in a patient with a vascular 
injury. Some injuries pose special problems 
of access and hemorrhage control, primarily 
because the injury is not in a body area com- 
monly operated on and familiar to the surgeon 
(Box 7-2) . 

Incisions for vascular trauma are selected 
with the goal of control, exposure, and repair. 
Some incisions are those routinely used for 
elective vascular reconstruction, and others 
have been specifically adapted for vascular 
trauma. Over the years, some incisions have 
declined in use or become obsolete. For 
example, the "trapdoor" incision (whereby a 
left supraclavicular incision is connected to 
an anterolateral thoracotomy incision by 
means of a partial median sternotomy) was 
found to add very little to the exposure of a 
vascular injury in the thoracic outlet while 
leading to significant chronic causalgia-like 
pain. The anterior thoracoabdominal incision 
was found to be too time consuming in the 
trauma setting, did not allow for adequate 



ch07.qxd 4/16/04 3:29 PM Page 143 



7 • ACCESS, CONTROL AND REPAIR TECHNIQUES 



143 



LIST OF INJURIES THAT POSE SPECIAL DIFFICULT ACCESS AND 
HEMORRHAGE CONTROL CHALLANGES 



High carotid injury 
Vertebral artery hemorrhage 
Thoracic outlet vascular injury 
Azygous vein 
Axillary artery injury 
Proximal abdominal aorta 
Intrathoracic inferior vena cava 
Suprarenal inferior vena cava 
Deep pelvic iliac vascular injury 
Complex groin vascular injury 
Distal popliteal/tibial vascular injury 



exposure of the thoracic aorta or other 
thoracic vasculature, and had significant long- 
term healing complications (Box 7-3). 



Neck 

Access to virtually all vascular injuries in the 
neck is accomplished via an incision along the 
anterior border of the sternocleidomastoid 
muscle. The patient is positioned with the 
head rotated as much as possible away from 



the operated side and extended, with the 
shoulders supported. The operative field 
always includes the anterior chest and an 
uninjured lower extremity. 

A typical incision for neck exploration for 
trauma extends from the suprasternal notch 
upward toward the ear lobe. Slightly curving 
the upper part of the incision away from the 
jaw will prevent inadvertent damage to the 
marginal mandibular branch of the facial 
nerve. During an exploration for penetrating 
trauma, and particularly in the presence of 



INCISIONS THAT ARE INFREQUENTLY OR NO LONGER USED FOR 
VASCULAR TRAUMA 

Collar neck incision 

Across the clavicle incision 

"Trapdoor" or "book" thoracotomy 

Anterior thoracoabdominal 

Posterior thoracoabdominal 

Combined midline abdominal extending across the groin 

Posterior popliteal 

Transverse abdominal 

Abdominal paramedian 



ch07.qxd 4/16/04 3:29 PM Page 144 



144 



II • PRINCIPLES OF OPERATIVE CARE 



active hemorrhage, many anatomic land- 
marks in the neck are obscured or distorted, 
and the surgeon has to rely on rapid 
identification of three key landmarks: the ante- 
rior border of the sternocleidomastoid, the 
internal jugular vein immediately behind it, 
and the facial vein. 

After division of the platysma the incision 
"opens up," allowing identification of the 
anterior border of the sternocleidomastoid. 
Lateral retraction of the muscle and further 
dissection in the middle cervical fascia exposes 
the jugular vein (Fig. 7-3). Dissection along 
the anterior border of the jugular vein allows 
the surgeon to identify, isolate, and divide the 
common facial vein between ligatures. This 
vein, a major branch of the internal jugular, 
is the "gateway to the neck" because free access 
and exposure of the common carotid artery 
and its bifurcation hinges upon division of the 
facial vein. Furthermore, the facial vein is fre- 
quently at the level of the carotid bifurcation. 

The sequence of dissection in the neck 
depends on the operative findings. In most 
cases, the focus of interest is the content of 
the carotid sheath, in which case dissection 
proceeds medial to the internal jugular vein, 
with special care being taken to identify and 




protect the vagus nerve. However, when arte- 
rial bleeding emanates lateral to the carotid 
sheath, the entire neurovascular bundle 
(including the internal jugular vein, the 
carotid artery, and the vagus) should be 
retracted medially, and dissection lateral to 
this anatomic compartment will allow access 
to the transverse processes of the cervical 
vertebrae and hence to the vertebral artery, 
an uncommon but life-threatening source of 
hemorrhage. 

The cardinal principle of obtaining proxi- 
mal and distal control before entering the 
injured segment applies in the neck. Occa- 
sionally, this will entail extending the neck 
incision into a median sternotomy to gain 
proximal control at the thoracic outlet for 
injuries to the proximal common carotid 
artery (zone 1 ). On rare occasions with simul- 
taneous bilateral injury, especially with a 
bullet across the neck ("transcervical" trajec- 
tory) , a U- or //-shaped incision coming across 
the trachea caudally to the thyroid cartilage 
facilitates bilateral neck exploration or expo- 
sure of both carotid sheaths and the anterior 
airway. 



GKerawth [.. Maltov, M.D. 



■ FIGURE 7-3 

Drawing depicting a "standard" neck incision, 
anterior to the sternocleidomastoid muscle with 

division of the anterior facial vein. ■ 



THORACIC OUTLET 

The thoracic outlet is a transitional anatomic 
area between the neck and the chest, where 
vascular injuries are difficult to access. Most 
surgeons infrequently operate in this area; 
therefore, deciding on the correct incision can 
be problematic. 

A patient with a thoracic outlet injury may 
be hemodynamically stable, thus allowing a 
precise angiographic localization of the injury. 
Under these circumstances, the location of 
the injury dictates the incision and the oper- 
ative approach. However, when the patient is 
actively bleeding from a vascular injury in the 
thoracic outlet, the decision has to rely on the 
clinical presentation. A penetrating injury 
around the distal clavicle that is bleeding exter- 
nally can initially be controlled with a Foley 
balloon tamponade inserted into the missile 
tract (Fig. 7-2) . An incision above and paral- 
lel to the clavicle will expose the injured vessel 
(see later discussion). On the other hand, a 



ch07.qxd 4/16/04 3:29 PM Page 145 



7 • ACCESS, CONTROL AND REPAIR TECHNIQUES 



145 



hemothorax is usually associated with a more 
medial injury, which may require proximal 
control of the subclavian artery through a high 
anterolateral thoracotomy on the left, or a 
median sternotomy on the right. For a patient 
with a thoracic outlet hematoma evident on 
the chest radiograph and without supra- 
clavicular or neck hematoma, a median 
sternotomy is the incision of choice. 

Access to the innominate artery, proximal 
left carotid, intrathoracic superior vena cava, 
and innominate vein is via a median ster- 
notomy. The key landmarks to performing 
a safe median sternotomy is to identify the 
sternal notch, the xiphoid, and the sternal 
midline, extending the incision a few cen- 
timeters below the xiphoid and opening the 
linea alba. A probing finger bluntly develops 
the space under the sternum both from below 
and from above behind the manubrium. One 
should be careful with the use of electro- 
cautery at the manubrial notch when divid- 
ing the retrosternal ligament attachment to 
the anterior neck fascia, because iatrogenic 
injury to the innominate artery and carotid 
may occur. This blunt dissection just beneath 
the sternum is relatively easy and proceeds in 
a bloodless plane. The sternal saw divides the 
sternum from the xiphoid to the manubrium, 
taking care to always stay in the midline. A 
partial sternotomy usually does not yield 
appropriate exposure and should therefore 
be avoided. Bleeding from the edge of the 
sternum can be impeded by the use of elec- 
trocautery. Only in the coagulopathic patient 
will bone wax be required for hemostasis from 
the marrow of the sternum. The sternal 
retractor is gradually opened as dissection con- 
tinues. If one immediately opens the sternal 
retractor to its full extent, an area of vascular 
injury can be "opened" more widely during 
forceful retraction of the sternum. In addi- 
tion, excessive and over-retraction of the 
sternum can create rib fractures, sternal frac- 
ture, and stretch injury to the brachial plexus. 
The pericardium is sharply entered in the 
midline and the dissection is carried cepha- 
lad to the area of an injury or to the innomi- 
nate vein that crosses in front of the aortic 
arch. This opening of the pericardium allows 
for the sternal retractor to be opened more 
widely for more exposure. 



Access to the extrathoracic left subclavian 
artery may be difficult, especially if the arm 
is extended. It is best achieved with the arm 
prepared free and initially placed at the 
patient's side. Exposure of the extrathoracic 
portion of the subclavian artery requires an 
incision about one fingerbreadth above and 
parallel to the clavicle from the sternal notch 
extending laterally. After division of the 
platysma muscle, the clavicular portion of the 
sternocleidomastoid is either retracted medi- 
ally or more conveniently divided. The scalene 
fat pad is encountered and removed, expos- 
ing the clavicular head of the sternocleido- 
mastoid muscle and the anterior scalene 
muscle. The phrenic nerve crosses in front of 
the anterior scalene muscle and must be iso- 
lated and carefully preserved. The more pos- 
teriorly located brachial plexus should also 
be protected while dividing the anterior 
scalene muscle. It is at this point that the sub- 
clavian artery first comes into view. It can now 
be carefully dissected and exposed. Care must 
be taken, because this is one of the most fyrigile 
arteries in the body. Only on rare occasions 
where the injury is proximal to the insertion 
of the scalene anticus muscle will the head 
of the clavicle need to be removed. 

In the presence of an expanding hematoma 
in the supraclavicular area, often it is difficult 
to define the anatomic landmarks. Under 
these circumstances, the artery can be rapidly 
exposed through the bed of the clavicle. The 
incision is made along the clavicle itself, 
and the bone is rapidly exposed. A periosteal 
elevator is used to peel off the periosteum 
around the clavicle and thus separate the bone 
from the adjacent muscles. The bone is 
rapidly divided as laterally as possible, lifted 
from its bed by grasping it with a towel clip, 
and the head is separated from the stern- 
oclavicular joint and removed. The subclav- 
ius muscle is sharply divided along the bed of 
the clavicle, providing access to the anterior 
scalene muscle and the phrenic nerve. 

For an extrathoracic left subclavian artery 
injury, a short anterior thoracotomy above 
the nipple (in the left second or third inter- 
space) may facilitate looping of the intratho- 
racic portion of the left subclavian artery 
proximal to the injury (Fig. 7-4) . A clamp or 
a snare tourniquet is then applied, which can 



ch07.qxd 4/16/04 3:29 PM Page 146 



146 



II • PRINCIPLES OF OPERATIVE CARE 



be tightened as the subclavian injury is 
exposed through a separate clavicular incision. 
Because of rich collateral circulation, this 
mode of proximal control does not stop bleed- 
ing completely but does help to make the 
situation more amenable to repair. 

An innominate artery injury may present 
intraoperatively as a large superior mediasti- 
nal hematoma. Blind dissection in such a 
hematoma is fraught with danger, because vas- 
cular structures (e.g., the innominate vein) 
are difficult to identify. Under these circum- 
stances, it is often prudent to deliberately enter 
the pericardium, which serves as a natural 
barrier to the extension of the hematoma. The 
vessels can be identified and controlled prox- 
imally, and a bypass graft can be inserted 
before dissection within the hematoma itself 
is undertaken (Fig. 7-5). 

Chest 

The chest is composed of three separate vis- 
ceral compartments, each accessible through 
different incisions. In the presence of massive 




■ FIGURE 7-4 

Drawing depicting an extrathoracic hematoma 
around the left subclavian artery and a left 
second interspace anterolateral thoracotomy, 
picking up the proximal left subclavian artery, 
encircling it with a ligature tourniquet (snare), to 
be used if necessary for vascular control when 
the hematoma is entered from a supraclavicular 
approach. ■ 



hemothorax, the utility incision is an antero- 
lateral thoracotomy, because it is and easy to 
perform and it does not require special posi- 
tioning. An anterolateral thoracotomy is per- 
formed in the fourth or fifth intercostal space 
on the side of the presumed injury, that is, 
below the nipple in the male patient or the 
manually retracted breast in the female 
patient. The incision extends from just lateral 
to the sternum to the midaxillary line. In the 
female patient, it is made in the inframam- 
mary crease. The pectoralis muscle is divided 
by dissection and the ribs are exposed. The 
intercostal muscles are divided along the 
upper aspect of the rib to avoid the neu- 
rovascular bundle, and the pleura is entered. 
A rib spreader is inserted with the handle 
pointing toward the axilla. Once inside the 
pleural cavity, the surgeon's first act is to mobi- 
lize the lung by dividing the pulmonary liga- 
ment up to the level of the inferior pulmonary 
vein. The lung is retracted anteriorly and the 
posterior mediastinum is thus exposed. In the 
left hemithorax, this incision provides good 
exposure of the descending thoracic aorta, 
left subclavian artery, left pulmonary artery, 
and pulmonary veins. In the right side of the 
chest, the intrathoracic inferior vena cava 
(IVC), right pulmonary artery, azygous vein, 
and superior vena cava can be seen. Atransster- 
nal bilateral anterolateral incision (clamshell) 
is accomplished by joining left and right 
anterolateral thoracostomies across the 
sternum using a Gigli saw, large-bone cutters, 
or an electric saw. Care must be taken to make 
the transsternal incision in the midportion 
of the body of the sternum, and not at the 
xiphoid, so a firm osseous closure will be pos- 
sible at the end of the operation. Care must 
also be taken to identify and ligate the divided 
internal mammary arteries. This clamshell 
incision is the only one that provides access 
to all three thoracic cavities, albeit at the cost 
of slight increased morbidity. 

A patient with precordial penetration who 
is not in extremis and whose bullet trajectory 
appears to be through the mid upper medi- 
astinum is best approached via a median ster- 
notomy. With some difficulty, this incision 
affords access also to the azygous vein, right 
main pulmonary artery, and left proximal sub- 
clavian artery. 



ch07.qxd 4/16/04 3:29 PM Page 147 



7 • ACCESS, CONTROL AND REPAIR TECHNIQUES 



147 




& Konnoth ].. VI alto*, M.D. 



■ FIGURE 7-5 

Drawing of a hematoma 
around the innominate artery 
and proximal left carotid artery 
with a median sternotomy and 
ligation of the innominate vein 
covering this area. Also shown 
is a "bypass" technique from 
the ascending aorta (end to 
side) to the distal innominate 
artery (end to end), with 
oversewing of the stump of the 
innominate artery at the aortic 
arch. ■ 



Although a resuscitative thoracotomy is 
usually performed via an anterolateral tho- 
racotomy, when a descending thoracic aortic 
repair is planned, a posterolateral thoraco- 
tomy is preferable. In either case, the poste- 
rior mediastinum is exposed by rotating the 
lung anteriorly (Fig. 7-6) . Division of the pari- 
etal pleura over the posterior mediastinum 
and mobilization of the hilum of the lung will 
assist in this exposure. The esophagus is 
locatedjust anterior to the aorta and the recur- 
rent laryngeal nerve recurs around the liga- 
mentum arteriosum. It is important not to 
injure these structures during access, expo- 
sure, or reconstruction. 

If the surgeon performs an anterolateral 
thoracotomy and discovers a posterior injury 
(e.g., to the esophagus or the descending 
aorta) , he or she is better advised to close the 
anterior incision and put the patient into a 
right lateral decubitus position. A left pos- 



terolateral fourth interspace incision is per- 
formed and the chest entered. 

Approaching a thoracic aortic injury in its 
usual location just distal to the ligamentum 
arteriosum, the lung is retracted anteriorly. 
The distal thoracic aorta, well away from the 
hematoma is encircled first, followed by the 
subclavian artery as it exits the chest. At this 
point, the transverse arch is dissected free from 
the pulmonary artery and the aortic arch 
between the left carotid artery and the left 
subclavian artery is encircled. This "control" 
is achieved well away from the undisturbed 
hematoma. If the surgeon elects to use car- 
diopulmonary bypass or active shunting, the 
cannula for this adjunct can be inserted at this 
point. 

An injury to the azygous vein is usually 
serendipitously discovered at emergency 
thoracotomy. Anterior incisions make 
exposure and ligation of a bleeding azygous 



ch07.qxd 4/16/04 3:29 PM Page 148 



148 



II • PRINCIPLES OF OPERATIVE CARE 




Kvnnulhl.. Mattox,M.D. 



■ FIGURE 7-6 

Exposure of the proximal descending thoracic 
aorta, by rotating the lung anteriorly and 
demonstrating that the esophagus is anterior to 
the aorta. Vascular clamp on the descending 
thoracic aorta as a temporary attempt to 
impede the rate of distal hemorrhage. ■ 



vein very difficult, contributing to the high 
mortality of this injury. Using an extra long 
needle holder and approaching the injury 
from the left side of the operating table pro- 
vides the surgeon with the greatest chance for 
achieving suture ligation of a bleeding azygous 
vein. 

The thoracic IVC is entirely within the 
pericardium and very difficult to expose. The 
surgeon may encounter an injury to this 
structure, digitally control the bleeding deep 
within the right lower pericardium, and then 
have great difficulty exposing the area for 
repair. Should bleeding be controlled, the 
trauma surgeon should call for a thoracic 
surgeon for assistance before attempting 
to proceed, because repair is exceedingly 



difficult and may require cardiopulmonary 
bypass, with double caval cannulation and 
repair of the caval injury from within the 
opened right atrium. 



Upper Extremity 

AXILLARY ARTERY 

The first and second parts of the axillary artery 
are approached through an infraclavicular 
incision that extends from the mid-clavicle 
to the deltopectoral groove. The fibers of 
the pectoralis major are separated bluntly, 
revealing the clavipectoral fascia medial to 
the pectoralis minor muscle. Opening the 
clavipectoral fascia and dissection in the axil- 
lary fat reveals first the axillary vein and then 
deep and superior to it, the axillary artery with 
the adjacent elements of the brachial plexus. 
The second part of the axillary artery is 
exposed by hooking up and then taking down 
the insertion of the pectoralis minor muscle 
as close as possible to the coracoid process 
using the electrocautery (Fig. 7-7) . In the pres- 
ence of an axillary hematoma, itmay be advis- 
able to first obtain proximal control on the 
subclavian artery through a supraclavicular 
incision and then perform a separate axillary 
incision or alternatively transect the clavicle 
to join the incisions. 

Other axillary exposures that are sometimes 
used in elective situations are rarely if ever 
used in the trauma situation. These include 
a lateral approach to the distal artery through 
a vertical incision along the lateral border of 
the pectoralis major or the deltopectoral 
groove approach. Lastly, endovascular control 
is possible when active extravasation is noted 
from the subclavian-axillary complex at 
angiography. A pair of occluding balloons, 
proximal and distal to the site of injury, will 
provide temporary control and minimize 
blood loss. 



BRACHIAL ARTERY 

The proximal brachial artery is usually 
approached via a medial upper arm incision 
placed in the groove between the biceps and 



ch07.qxd 4/16/04 3:29 PM Page 149 



7 • ACCESS, CONTROL AND REPAIR TECHNIQUES 



149 




3**' O Kenneth L. Mattos, M.D. 



■ FIGURE 7-7 

Drawing of exposure of the 
axillary artery by division of the 
muscle fibers between the 
clavicular and pectoral 
portions of the pectoralis major 
muscle and detachment of the 
insertion of the pectoralis minor 
on the coracoid process of the 
scapula. ■ 



triceps muscles. Care should be taken in an 
arm with a large hematoma, as the neurovas- 
cular bundle is closer to the surface than one 
might expect and iatrogenic injury is to be 
avoided. Care must also be taken to identify 
any concomitant injury to the brachial vein 
and adjacent nerves. Dissection in the groove 
between the triceps and biceps muscles reveals 
the neurovascular bundle, and the first struc- 
ture that is encountered is the median nerve, 
which should be carefully preserved. 

A distal brachial artery injury often requires 
exposure at the antebrachial fossa, through 
a sigmoid incision that avoids crossing in the 
antecubital skin crease. The artery is located 
immediately below the biceps tendon, which 
can be divided with impunity. The sigmoid 
incision may be carried upward along the 
medial part of the upper arm or distally to 
expose the brachial artery bifurcation. 



Access to vascular injuries in the extremi- 
ties is based on the Henry principle of exten- 
sile exposure. Every incision can be extended 
proximally or distally or joined with an inci- 
sion exposing a more proximal or distal 
vessel. Thus, the subclavian and axillary expo- 
sures can be joined by dividing the clavicle. 
The axillary and brachial incisions can be 
joined by extending the former in the del- 
topectoral groove across the shoulder. 



Abdomen and Pelvis 

Abdominal vascular injuries account for 
the majority of truncal vascular trauma 
seen in a civilian practice. These injuries are 
approached via a midline laparotomy incision, 
one of the most commonly used incisions 
in trauma. After incision of the skin and 



ch07.qxd 4/16/04 3:29 PM Page 150 



150 



II • PRINCIPLES OF OPERATIVE CARE 



subcutaneous tissue using the xiphoid process 
and umbilicus as markers, the linea alba is 
gained by identifying the midline decussation 
of the fibers of the anterior rectus sheaths on 
both sides. In a trauma laparotomy, time is 
typically not wasted on superficial hemostasis 
and the entire incision is rapidly performed 
with a scalpel. The peritoneum is typically 
entered immediately above the umbilicus, 
where rapid atraumatic penetration of the 
peritoneum is usually possible. Very rarely will 
the surgeon choose a different incision. In a 
patient who is in shock and has had multiple 
previous operations through a midline inci- 
sion, it may be wise to avoid the dense and 
time-consuming adhesions by rapidly per- 
forming a subcostal ("chevron" or "rooftop") 
incision instead. 

When considering definitive control and 
repair of intraabdominal vascular injury, the 
surgeon has several distinct patterns of 
retroperitoneal hematoma to guide him or 
her to specific vascular injuries. An upper 
abdomen (supramesocolic) midline retrope- 
ritoneal hematoma is associated with injury 
to the suprarenal aorta, celiac axis, and the 
superior mesenteric artery. The midabdomi- 
nal midline retroperitoneal (inframesocolic) 
hematoma is associated with proximal renal 
artery and infrarenal aortic or vena cava injury. 
A perinephric hematoma may be associated 
with renal or renal vascular injury. A pelvic 



midline hematoma is most often associated 
with a pelvic fracture or bladder injury, and 
a large or expanding lateral pelvic hematoma 
is associated with iliac vascular injury. A right 
lateral retroperitoneal hematoma suggests an 
IVC injury that may be infrarenal or retro- 
hepatic. Finally, a hematoma presenting in the 
porta hepatis indicates an injury to the portal 
venous system. 

Currently, initial vascular control of 
intraabdominal hemorrhage is achieved in the 
abdomen alone, initially by using laparotomy 
pads or manual/digital pressure. 

Rapid evisceration of the small bowel will 
allow the surgeon to define the area of major 
hemorrhage. In the presence of profuse 
bleeding from a midline retroperitoneal 
hematoma, the first assistant digitally occludes 
the aorta at the esophageal hiatus. Use of 
various aortic occluding instruments is much 
less effective than simple digital occlusion. 
Virtually all abdominal venous bleeding can 
initially be controlled by pressure packs. 

Temporary control of gross bleeding in the 
area of the celiac trunk often presents a 
difficult technical situation to the surgeon 
because visibility in this area is very limited 
without elaborate dissection. Hemostasis 
can sometimes be achieved with a large gross 
ligature, using rather large suture material, 
such as 1 or suture on a large needle 
(Fig. 7-8) . Although intended as a temporary 



■ FIGURE 7-8 

Drawing depicting a gross 
ligature of an area of gross 
bleeding such as in the area of 
the celiac axis. This suture 
should be rather large suture 
material, such as 1 or suture 
on a large needle. This is a 
temporary tactic and might be 
removed later after other 
control techniques. ■ 




9KcEinclhLMatlc<, U.D. 



ch07.qxd 4/16/04 3:29 PM Page 151 



7 • ACCESS, CONTROL AND REPAIR TECHNIQUES 



151 



hemostatic maneuver, if the maneuver is 
effective and the surgeon is satisfied that 
mesenteric and hepatic injuries have not 
resulted from this "blind" suturing, this 
temporary suture may be left in place 
permanently. 

Cross clamping of the aorta has tradition- 
ally been viewed as an important maneuver 
in trauma surgery. Such clamping is per- 
formed both to control exsanguinating hem- 
orrhage in the abdomen or pelvis and as a 
resuscitative maneuver. Historically, this has 
been done by cross clamping the descending 
thoracic aorta in the chest, away from the site 
of intraabdominal bleeding (Fig. 7-6). Unfor- 
tunately, this addition of a thoracic incision 
in a patient with a complex abdominal injury 
who is already coagulopathy, acidotic, and 
hypothermic serves only to aggravate this often 
fatal triad and should therefore be avoided. 
The aortic cross-clamp maneuver is per- 
formed for several reasons, the most common 
one being to preserve the residual blood 
volume for vital perfusion of the heart, brain, 
and lungs during resuscitative emergency 
department thoracotomy. However, when 
performed for proximal control of an injury 
to the abdominal aorta or its visceral branches, 
the inexperienced surgeon rapidly discovers 
that because of very rich collateral circulation, 
this does not dry up the operative field. 
However, digital occlusion of the abdominal 
aorta at the esophageal hiatus will markedly 
reduce bleeding (Fig. 7-9). Numerous com- 
plications have occurred after thoracic or 
abdominal aortic cross clamping, especially 
when the procedure is performed by an 
inexperienced surgeon. If clamping of the 
supraceliac aorta is required, the safest tech- 
nique is either to take down the left triangu- 
lar ligament of the liver or to bluntly enter 
the lesser omentum and then perform a blunt 
digital separation of the fibers of the diaphrag- 
matic crus immediately above and behind the 
origin of the abdominal aorta, as described 
by Veith, Gupta, and Daly (1980). The 
surgeon's index finger is then insinuated 
through the diaphragmatic crus on each side 
of the aorta, to create just enough space for 
the clamp on both sides of what is in fact the 
lowermost part of the thoracic aorta. Using 
this technique, the surgeon avoids the 




S Kenneth L. Mdttox. VI. D. 



■ FIGURE 7-9 

Drawing depicting the right hand of the first 
assistant (left-hand side of the patient's body), 
compressing the abdominal aorta, with a 
hematoma around the aorta at the level of the 
mesenteric vessels and achieving vascular 
control. ■ 



hazardous and frustrating dissection in the 
thick periaortic tissue that surrounds the first 
part of the visceral aortic segment. 

Access to the injured suprarenal aorta is 
one of the greatest operative challenges in 
abdominal trauma. An anterior approach 
would require the stomach and pancreas to 
be either retracted or transected, and the 
dense periaortic nerve and fibrous tissue make 
dissection in this area difficult. A medial rota- 
tion of all the intraabdominal viscera to the 
patient's right from a dissection plane lateral 
to the left colon and going behind the spleen, 
kidney, and tail of the pancreas allows a lateral 
and relatively easy approach to the aorta. This 
intraoperative maneuver has been called the 
"Mattox maneuver" for the past 2 decades 
(Fig. 7-10). This dissection plane is on top of 
the psoas muscle and can be rapidly achieved 
by rapid blunt dissection that begins with the 
peritoneal reflection lateral to the distal 
descending colon and is carried upward 



ch07.qxd 4/16/04 3:29 PM Page 152 



152 



II • PRINCIPLES OF OPERATIVE CARE 



■ FIGURE 7-10 

Rightward visceral rotation 
from the left side from the 
diaphragm to the iliac arteries 
("Mattox maneuver"). ■ 




4*r 



D Konni'lli L. U.illos, Ml) 



lateral to the spleen and up toward the 
diaphragmatic hiatus, rotating all left-sided 
abdominal viscera medially and exposing the 
abdominal aorta from the esophageal hiatus 
to where the external iliac arteries exit the 
abdomen to the groin. In most situations 
where this maneuver is needed, the retroperi- 
toneal hematoma itself already achieves a 
significant separation of the relevant dissec- 
tion planes, thus greatly facilitating the 
maneuver. The left lateral diaphragmatic 
crux can be divided and a lateral incision in 
the diaphragm can further expose the distal 
thoracic aorta for proximal control even as 
high as the T6 vertebra without having to open 
the chest. A thick fascial layer separates the 
aorta from the dissection plane and must be 
carefully incised to cleanly define the aortic 
wall in preparation for clamping. In the pres- 
ence of proximal vascular control, the aorta 
loses its pulse and is a flaccid tube that is not 
always easy to identify in a large hematoma. 
As soon as the aorta is exposed, vascular clamps 
are then precisely placed on the injured vessel 
for control and reconstruction. 

The infrarenal aorta is exposed by eviscer- 
ating the small bowel to the patient's right and 
upward, mobilizing the ligament of Treitz, and 
then longitudinally dividing the posterior 
peritoneum between the duodenum and the 
inferior mesenteric vein. The pitfall in this 
exposure is failure to identify the left renal 



vein in the presence of a large infrarenal 
hematoma, which may lead to an iatrogenic 
injury and additional blood loss. 

The most complete and extensive access to 
the mid and lower retroperitoneal structures 
is by performing the Cattell-Braasch maneu- 
ver, an extensive mobilization of the peritoneal 
structures off the aorta, vena cava, and their 
major branches (including the renal and iliac 
vessels) (Fig. 7-11). This maneuver involves 
division of the peritoneal attachments of the 
duodenum, right colon, and mesentery of the 
small bowel from the posterior abdominal 
wall. The line of incision is a triangle that 
begins at the lateral edge of the hepatoduo- 
denal ligament (adjacent to the common bile 
duct) , is carried downward to the cecum, and 
then upward along the insertion of the small 
bowel mesentery to the ligament of Treitz. This 
allows full rotation of the midgut from the 
duodenum to the transverse colon with its 
accompanying mesentery, up onto the ante- 
rior chest. The extended Kocher maneuver 
is mobilization of the duodenum and right 
colon only, a limited version of the full Cattell- 
Braasch maneuver that is often sufficient to 
expose vena cava injuries. 

The renal vessels can be controlled either 
proximally ("midline looping") or by mobi- 
lizing the kidney itself and clamping across 
the renal hilum. Midline looping conforms 
to the principle of proximal control and 



ch07.qxd 4/16/04 3:29 PM Page 153 



7 • ACCESS, CONTROL AND REPAIR TECHNIQUES 



153 




Kciuuth L. Matto*, M.D. 



■ FIGURE 7-1 1 

Drawing depicting the 
combined Kocher and Cattell- 
Braasch maneuvers showing 
that the movement of the 
duodenum, small bowel, and 
right colon to the left exposes 
the entire inferior vena cava, 
right renal vessels, and the 
right iliac vessels. ■ 



involves identification and isolation of the left 
renal vein as it crosses the aorta, and then 
identification and looping of the right or left 
renal artery as it comes off the aorta. In the 
presence of an actively bleeding perinephric 
hematoma, it is sometimes quicker to simply 
mobilize the entire kidney from its bed in a 
manner akin to a splenectomy and place a vas- 
cular noncrushing clamp en masse across the 
entire hilum. 

Control of the portal vein between the top 
of the pancreas and the liver is aided by a 
Pringle maneuver, which consists of placing 
a vascular clamp across all of the structures 
of the porta hepatis. Under rare conditions, 
a "double Pringle" maneuver is performed by 
placing vascular clamps on either side of an 
injury in the hepatoduodenal ligament. 

The control of portal venous injuries behind 
the pancreas is often challenging and can be 
aided by deliberate division of the neck of the 
pancreas (Fig. 7-12. Control by direct pres- 
sure or application of large vascular clamps 
is accomplished preparatory to this division. 
Dissection in the hepatoduodenal ligament 
and identification and division of the gastro- 
duodenal artery is the most time-consuming 
part of this elaborate (but sometimes lifesav- 
ing) maneuver. This procedure is the only 
practical way to gain access to the confluence 
of the splenic and superior mesenteric veins 
to form the portal vein. 



The suprarenal retrohepatic IVC is another 
area of difficult exposure. A contained 
hematoma in this location is best left undis- 
turbed because there are no major retro- 
hepatic arterial structures, so the injury is 
invariably venous and can often be controlled 
with local pressure. If free bleeding from this 




© Kenneth 1_. MattOX, M.D. 



■ FIGURE 7-1 2 

Drawing depicting exposure of the portal vein 
and proximal superior mesenteric vein, by 
dividing the mesenteric root and deliberately 
dividing the neck of the pancreas, showing the 
confluence of the superior mesenteric vein, and 
splenic vein to form the portal vein. ■ 



ch07.qxd 4/16/04 3:30PM Page 154 



154 



II • PRINCIPLES OF OPERATIVE CARE 



area is encountered, containment with laparo- 
tomy packs is the goal, with atrial caval shunts 
reserved for uncontrolled hemorrhage from 
the retrohepatic IVC. 

Injury to the iliac vasculature, either at the 
confluence of the iliac veins to the IVC or at 
the inguinal ligament, poses several exposure 
and control challenges (Fig. 7-13). After 
opening the abdomen and discovering a 
hematoma in the pelvic retroperitoneum, 
hemorrhage control might require initially 
local pressure using folded sponges on a 
"sponge stick." This is mainly because "blind" 
clamping of the iliac arteries in a hematoma 
is likely to result in injury to the immediately 
underlying iliac veins or the overlying ureters. 
On rare occasions, large vascular occluding 
clamps are required to gain control en masse 
of both the iliac artery and the iliac vein. 

A careful technique for gaining control of 
an iliac vascular injury in a gradual fashion 
has been described by Burch and colleagues 
(1990). Initially, the distal aorta and vena 
cava are clamped away from the pelvic 
hematoma, and the distal external iliac artery 
is controlled by "towing in"with a large Deaver 
retractor over the lower edge of the abdomi- 
nal incision, thus globally compressing the 
external iliac vessels against the pelvic brim. 
As dissection proceeds into the pelvis, the 




KluhiUi I.. MattOX. Mil. 

■ FIGURE 7-13 

Drawing depicting a lateral lower abdominal 
hematoma suggesting an injury to the iliac 
vasculature. ■ 



upper clamps are gradually advanced distally 
to selectively control the injured arterial or 
venous segment of the iliac vasculature, a 
technique called "walking the clamps." On 
very rare occasions, the groin must be opened 
to gain control of backflow by occluding 
the common femoral artery and the deep 
femoral vein. 



Lower Extremity 

Three areas in the lower extremity are the 
focus of the surgeon considering a vascular 
injury: the groin, distal thigh, and proximal 
calf. At the groin, access is achieved via a 
routine groin incision, exposing the common 
femoral artery and its branches. The distal 
superficial femoral and proximal popliteal 
arteries are approached via a distal medial 
thigh incision. 



GROIN 

In the emergency department, active bleed- 
ing from the groin is often controlled by 
direct pressure. This challenging bleeding 
can also be controlled by balloon tamponade. 
The dilemma is whether to go into the 
abdomen for proximal control or to approach 
the injury directly in the groin. If a pene- 
trating wound such as a gunshot injury also 
obviously enters the abdomen, then rapid 
proximal control in the abdomen is indicated 
because there is an independent indication 
for laparotomy. However, if the injury appears 
to be limited to the groin, options are either 
to do a vertical utility incision over thefemoral 
triangle with an extension unto the inguinal 
ligament and then divide the ligament and 
gain proximal control or to first gain proxi- 
mal control through an oblique incision 
above and parallel to the inguinal ligament 
and expose the external iliac vessels in this 
extraperitoneal location. In our experience, 
it is almost always possible to gain proximal 
control of femoral injuries in the groin. A 
useful trick is to identify the inguinal ligament 
and instead of incising it to just separate the 
fibers of the ligament approximately 1 to 
2 cm above its shelving edge, and thus bluntly 



ch07.qxd 4/16/04 3:30PM Page 155 



7 • ACCESS, CONTROL AND REPAIR TECHNIQUES 



155 



"penetrate" through the ligament into the 
retroperitoneum. This small opening is usually 
enough to insert a narrow and deep retrac- 
tor, to feel the external iliac pulse, and to clamp 
the artery safely. 

The technical key to a safe dissection in the 
groin in the presence of a vascular injury is 
to identify the common, superficial, and deep 
femoral arteries and their accompanying 
veins before clamping, to avoid troublesome 
back bleeding and to preserve the deep 
femoral artery. The lateral circumflex iliac vein 
crosses the deep femoral artery very close to 
its origin and is easily injured when looping 
of the profunda femoris is attempted in the 
presence of a large hematoma and hostile 
groin anatomy. The specific location of the 
injury to the femoral vessels has profound 
implications on the repair options. Often the 
surgeon must extend the initial incision to 
create a broader exposure than was initial 
thought to be sufficient. 



DISTAL SUPERFICIAL 
FEMORAL ARTERY 

A medial distal thigh incision is the "utility" 
approach to the distal superficial femoral 
artery, deep femoral vein, and proximal 
popliteal artery. The sartorius muscle is 
reflected upward or downward and Hunter's 
canal is opened. Proximal control can be 
achieved by a proximal tourniquet, proximally 
occluding the distal femoral artery, or control 
from a groin incision with exposure of the 
proximal superficial femoral artery. Care 
should be taken not to iatrogenically injure 
the saphenous vein or its accompanying nerve 
that in the thigh travels in proximity to the 
superficial femoral artery. 



POPLITEAL ARTERY AND BRANCHES 

A difficult area for vascular access is the 
popliteal artery. Injuries at the popliteal bifur- 
cation are best exposed by a liberal incision 
below the knee, which in the presence of a 
large hematoma, fractures, or soft tissue 
destruction may begin proximal to the knee 
in an uninjured area and extend distally 



according to the principle of extensile 
exposure. Often adj acent nerves and veins are 
also injured. Although it is acceptable to cut 
across the pes anserinus at the medial aspect 
of the knee, it is less morbid to make two inci- 
sions (one above the knee and the other below 
the knee) if the popliteal artery injury is in 
the proximal or mid segment of this artery. A 
pneumatic tourniquet in place at the groin 
may assist access to the proximal tibial 
arteries during dissection and identification 
of the injured vessels. 

Care must be taken to wo£injure the greater 
saphenous vein at the time of the skin inci- 
sion so that it can serve as a collateral venous 
outflow tract should the popliteal vein require 
ligation. The contralateral leg is prepared and 
draped so that this uninjured site can be used 
for a substitute conduit if necessary. 

As a useful general guideline, the major 
neurovascular bundles of the lower extrem- 
ity are always located immediately behind the 
bone. This is especially important to remem- 
ber in the presence of hematoma and gross 
anatomic distortion. Thus the distal superficial 
femoral artery and the popliteal artery will be 
found immediately behind the femur, and the 
popliteal bifurcation and tibioperoneal trunk 
will be found immediately behind the tibia. 
One often encounters the accompanying 
vein before the artery during the dissection. 

The incision to expose the tibioperoneal 
trunk and its branches is made just posterior 
to the medial edge of the tibia. It is carried 
down to the medial head of the gastrocnemius 
muscle that is sharply divided off the tibia (Fig. 
7-14) . Often the attachments of the soleus 
muscle must also be detached to expose the 
posterior tibial and peroneal vessels. In the 
presence of a hematoma, it is often advisable 
to begin the dissection far proximally, 
identify and isolate the uninjured proximal 
popliteal segment, and then gradually advance 
distally toward the injury. 

The medial approach affords only proxi- 
mal control of the origin of the anterior tibial 
artery. The vessel itself is best exposed through 
an anterolateral incision placed approxi- 
mately two fingerbreadths lateral to the ante- 
rior edge of the tibia and carried past the fascia 
and between the tibialis anterior and the 
extensor hallucis longus muscles. 



ch07.qxd 4/16/04 3:30PM Page 156 



156 



II • PRINCIPLES OF OPERATIVE CARE 




■ FIGURE 7-14 

Drawing depicting a long below the knee 
medial incision, division of the medial head of 
the gastrocnemius muscle to expose the distal 
popliteal and tibial vessels. ■ 



Assess the extent of the injury or injuries. 

Decide whether "vascular damage control" is 
required. 

Determine the amount of debridement 
required. 

Determine the type of repair required. 

Set up the vessels for evaluation and repair by 
placement of "stay sutures." 

Pass Fogarty catheters to ensure the proximal 
and distal clot has been removed. 

Instill local heparin into the open vessels. 

Apply local vascular occluding devices. 

Perform the suture line. 

Flush proximally and distally before comple- 
tion of the suture line. 

Complete the suture line. 

Remove the clamps. 

Assess the distal circulation. 

Consider the need for fasciotomy. 

Determine special post-repair requirements. 



REPAIR PRINCIPLES 
AND TECHNIQUES 



For both vascular trauma and elective vascu- 
lar reconstruction, a number of basic princi- 
ples apply to all areas of vascular surgery. These 
principles especially apply to the injured 
vessel that may be surrounded by a hematoma, 
may be actively bleeding, or may be in an area 
of disrupted or "hostile" anatomy. Before any 
vascular procedure, the surgeon should ascer- 
tain the availability of any special instruments, 
equipment, assistants, imaging devices, or 
other devices that might be required during 
the procedure. After making the decision to 
explore an area, gaining access and control, 
and then discovering a vascular injury, the pro- 
gression through repair is as follows: 



Ensure proximal and distal control. 

Explore the injury. 

Carefully enter the hematoma. 



Each of these steps are not covered in detail, 
but the major points of interest to the trauma 
surgeon are covered. 

1. Ensure proximal and distal control. 

To enter a hematoma without the ability 
to have proximal and distal control will 
cause the trauma surgeon to have con- 
siderable difficulty with the vascular injury 
and add to the potential for additional 
iatrogenic injury. 

2. Explore the injury. 

The area of the hematoma or the trajec- 
tory of a wounding agent is evaluated 
and explored. Associated injuries are 
tabulated and the surgeon determines 
priorities. Because of the potential for 
exsanguinating hemorrhage and distal 
ischemia, vascular injury usually takes the 
highest priority. 

3. Carefully enter the hematoma. 

Anatomy is often distorted following 
trauma. The surgeon enters the area of 



ch07.qxd 4/16/04 3:30PM Page 157 



7 • ACCESS, CONTROL AND REPAIR TECHNIQUES 



157 



specific vascular injury and assesses the 
extent of the vascular injury. Although 
proximal and distal control has been 
obtained, local control is often required, 
by direct digital pressure, movement of 
vascular clamps closer to the vascular 
wound, application of a partially occlud- 
ing vascular clamp, and/or the use of 
intraluminal balloons. 

4. Decide whether "vascular damage 
control" is required. 

The patient with a vascular injury presents 
in a hemodynamically unstable condi- 
tion, often acidotic, coagulopathic, and 
hypothermic, thus differing considerably 
from the patient requiring elective vas- 
cular surgery. It is at this point that the 
surgeon should consider options for 
"vascular damage control." This might 
include ligation of this specific vascular 
injury for local hemorrhage control and 
performing an extra-anatomic bypass for 
distal perfusion. Vascular damage control 
might also entail the insertion of a tem- 
porary intraluminal shunt. Finally, a 
damage control tactic of ligation might 
become the final procedure if hemor- 
rhage is controlled and distal circulation 
is intact. 

5. Determine the amount of debridement 
required. 

The extent of an "adequate" debridement 
of an injured vessel is a matter of judg- 
ment. With simple lacerations and 
penetrations from low-velocity missiles, 
debridement should be to the extent to 
demonstrate a normal-appearing intima. 
With extensive arterial destruction, such 
as with high-velocity gunshot wounds, 
blast injury, and crush injury, more 
extensive debridement is necessary. 
Detection of a normal-appearing arter- 
ial wall, including an intact intima, is 
usually satisfactory evidence of suffi- 
cient debridement. The surgeon should 
closely observe the quality of both inflow 
and outflow because this gives some 
indication of problems proximal to the 
repair and on the adequacy of collateral 
circulation. 



6. Determine the type of repair required. 

It is at this point that the surgeon deter- 
mines whether the injury can be repaired 
by simple lateral repair, apply a patch 
angioplasty, perform an end-to-end anas- 
tomosis, or insert a substitute conduit. 
This decision might require another 
member of the operative team to obtain 
a saphenous vein from the previously 
prepared leg donor site. 

7 . Set up the vessels for evaluation and repair 
by placement of "stay sutures." 

The vessel to be repaired is "set up" for 
the reconstruction. This often entails the 
application of lateral stay sutures and 
establishment of a new clean operative 
field. 

8. Pass Fogarty catheters to ensure the prox- 
imal and distal clot has been removed. 

A surgeon who uses Fogarty catheters only 
occasionally should be reminded of a 
number of caveats (Box 7-4) . Over- 
inflation of the balloon will cause it to 
rupture and potentially cause intimal 
injury at the site of rupture. In addition, 
there is a risk of remote perforation of 
the vessel and an increased risk to the 
intima with each repeated pass of the 
catheter. Much of the art form of using 
Fogarty balloon catheters is in the feel and 
touch of pressure or resistance felt by 
the surgeon at the time of advancing the 
catheter, inflation of the balloon, and 
extraction of the catheter. At least two 
"clean" passes should be made before 
declaring that an artery is free from distal 
clots. The operating surgeon should 
control three items simultaneously: the 
pressure on the syringe connected to the 
Fogarty balloon catheter, the pull of 
the catheter extracting any clots, and the 
orifice of the vessel. Many surgeons will 
infuse a small amount of heparinized 
saline after the final pass of the catheter 
as the clamp is reapplied to the vessel. 

9. Instill local heparin into the open 
vessels. 

Systemic anticoagulation is rarely used in 
patients with acute trauma, especially with 



ch07.qxd 4/16/04 3:30PM Page 158 



158 



II • PRINCIPLES OF OPERATIVE CARE 



CAVEATS IN THE USE OF FOGARTY CATHETERS 
IN VASCULAR TRAUMA 



Choose the smallest balloon that will accomplish the task required. 

Estimate the size of the most distal site to which the balloon is to be inserted 
in determining the size of the balloon to be used. 

Always read the volume of liquid required to inflate the balloon. 

Put only the volume of liquid required to inflate the balloon in the syringe 
used for inflation. 

Test the balloon inflation before insertion into the artery to ensure that the 
balloon is functional and to see the diameter of the inflated balloon. 

Always advance the ballooned catheter with the balloon deflated. 

Always advance the balloon with the surgeon's fingers, not an instrument. 

Should resistance be met, further advancement should not be attempted. 

Remember that the Fogarty catheter can perforate an artery if forced. 

The surgeon who advances the catheter should inflate the balloon, and if 
resistance is met, the balloon should not be inflated further. 

With the balloon inflated, the catheter should be withdrawn. During the 
withdrawal if the surgeon meets resistance, the balloon should be allowed 
to deflate slightly before beginning the withdrawal anew. 

Remember that an overinflated Fogarty balloon can tear the intima of an 
artery and forcibly extract long segments of intima, denuding the inte- 
rior of the artery. 

Should clot be removed, a second pass should be accomplished. 

Passes to the distal artery should continue until no clot remains to be 
removed. 



multisystem acute trauma. The trauma 
and/or vascular surgeon is always con- 
cerned about coagulopathies and worried 
that with systemic heparinization, hem- 
orrhage at the sites of injury will com- 
pound those injuries, especially the head 
and orthopedic trauma. Furthermore, 
many trauma patients often are already 
somewhat, if not frankly, coagulopathy 
by the time they get to the OR. Aggres- 
sive crystalloid fluid resuscitation of as 
little as 750 mL of fluid results in a statis- 
tically difference in the clotting studies 
compared with matched patients who 
received little or no resuscitative fluid. The 
first concern of a trauma surgeon is that 
no new factors are introduced that would 
contribute to a coagulopathy. Hypother- 



mia, dilution of clotting factors, and 
addition of drugs that alter clotting all 
contribute to a coagulopathy. 

Except for patients who are placed on 
total cardiopulmonary bypass, systemic 
anticoagulation is avoided acutely in the 
trauma patient. However, under certain 
circumstances, systemic heparinization 
may be in the patient's best interest. When 
the arterial injury is the result of a single 
penetrating injury (such as a stab wound 
to the brachial artery), the risk of bleed- 
ing is minimal, and in a teaching situa- 
tion or when the reconstruction is very 
time consuming, systemic heparinization 
may be considered. Similarly, in the pres- 
ence of severe and prolonged distal 
ischemia (e.g.,whenafirstreconstruction 



ch07.qxd 4/16/04 3:30PM Page 159 



7 • ACCESS, CONTROL AND REPAIR TECHNIQUES 



159 



is unsatisfactory and an immediate redo 
is required) , when there is concern about 
clotting of the microcirculation and irre- 
versible damage, again systemic heparin 
may be considered provided that no 
other injuries are likely to bleed. 

Most patients with vascular injury 
undergo operation in this acute period. 
Many trauma patients with vascular 
trauma have undergone a series of arte- 
riograms in the angiographic suite, where 
repeated aliquots of heparin have been 
administered. Should an early operation 
follow, the surgeon would be well served 
to determine the level of anticoagulation 
by an intraoperative determination of the 
activated clotting time. 

Many surgeons will consider the admin- 
istration of heparin to patients with an 
acute, totally occluded artery, to preserve 
distal function, with the theory being that 
collaterals and venous return are pre- 
venting from thrombosing. Whether 
this is true for trauma patients, who 
are already relatively coagulopathic, is 
unknown. Because many of the coagula- 
tion profiles available in many hospitals 
report a result at least 1 to 2 hours after 
the blood was obtained and the patient's 
coagulation status may have changed 
considerably in that period, tests that 
demonstrate concurrent status are pre- 
ferred. Most trauma centers also have 
either cardiac or vascular surgical capa- 
bility and have equipment to measure 
an activated clotting time available in 
the ORs; this test is preferred. The acti- 
vated clotting time is well recognized by 
cardiac surgeons to be an excellent deter- 
minant of the effect of heparin on clot- 
ting activity. 

Many surgeons elect to inject through 
the open ends of an injured vessel prox- 
imally and distally small aliquots of 
heparinized saline. These solutions 
contain a variable number of units of 
heparin, depending on the local recipe. 
Often these "local" injections result in a 
"systemic" heparinization dose, which 
literally occurs within minutes of the 
infusion. 



10. Apply local vascular occluding devices. 

The locally applied vascular occluding 
device should be noncrushing and gentle 
to the vessel. A vascular clamp should not 
be maximally applied but closed only to 
the extent required to prevent bleeding 
from the open ends of the vessel. In some 
locations, a customized snare tourniquet 
allows for the occlusion to be complete 
but keeps the instruments out of the 
operative field. In other instances, intra- 
luminal balloon catheters serve as the 
occluding device. 

11. Perform the suture line. 

Anastomoses must be tension free and 
carefully constructed to create an everted, 
smoothly coapted layer of intact and 
healthy intima. The surgeon should wear 
magnifying loupes if necessary, espe- 
cially for small vascular reconstructions. 
Several options exist for vascular trauma 
reconstruction. 

a. Simple vascular repair techniques 

Any vascular reconstruction in vascu- 
lar trauma should be tailored to both 
the patient's condition and the par- 
ticular injury encountered. There is 
no single "practice guideline" that is 
applicable to every injury. A large 
number of acceptable standards of 
practice exist. For instance, for an 
unstable patientwith extensive truncal 
injury, an extremity arterial injury may 
be left unreconstructed to focus on a 
critical life-threatening truncal injury. 
In such an omission or delay, an ampu- 
tation might be the ultimate outcome 
in a patient who is now alive because 
the truncal and cerebral injuries were 
addressed. Several simple approaches 
to vascular reconstruction exist. 

A "simple" vascular repair tech- 
nique is a lateral venorrhaphy or 
arteriorrhaphy. Should lateral arteri- 
orrhaphy or venorrhaphy be accom- 
plished without narrowing the vessel 
and without tension on the suture line, 
this technique is the preferred ap- 
proach over more complex vascular 



ch07.qxd 4/16/04 3:30PM Page 160 



160 



II • PRINCIPLES OF OPERATIVE CARE 



repair techniques. Lateral repair is 
sometimes performed in a linear and 
at other times in a horizontal manner. 
In principle, the axis of the suture line 
should be oriented perpendicular to 
the axis of the vessel, to avoid nar- 
rowing. However, in special circum- 
stances when the vessel is large (such 
as the IVC) and a perpendicular repair 
is impossible, a suture line parallel to 
the axis of the vessel is an acceptable 
second-best approach. 

The choice relates to the size of the 
vessel and the surgeon's preference. 
Surgeons choose the smallest suture 
material possible to accomplish the 
closure safely, most often using poly- 
propylene suture material. 

Ligation of a bleeding vessel has 
been a form ofvascular control/repair, 
because the word suture was used in 
the Edwin Smith Surgical Papyrus. The 
ancient and modern literature aptly 
demonstrates the natural history 
following ligation (or thrombosis) of 
major arteries and some major veins, 
that being distal ischemia and loss of 
the distal organ, often expressed as 
an amputation. Ligation is an option 
in almost all venous injuries and in a 
number of arterial injuries when the 
patient's condition and overall trauma 
burden preclude a reconstruction. 
Examples of arteries that can be ligated 
include subclavian, internal iliac, 
superficial femoral, one of the tree 
distal vessels in the lower arm or calf. 
In other instances, ligation (e.g., 
external iliac artery) may be required 
for hemorrhage control in a very 
complex injury, where a secondary 
reconstruction outside the major 
injury is accomplished (a femoral- 
femoral arterial crossover graft) . 
Ligation of one of paired arteries 
(brachial and ulnar arteries, anterior 
and posterior tibial arteries) is toler- 
ated provided distal crossover collat- 
eral circulation exists (as in the case 
of an intact palmar arterial arch). If 
ligation is used as a procedure of 
choice for vascular hemorrhage 



control, the surgeon must early and 
frequently assess the viability and func- 
tion of the circulation distal to the lig- 
ature and make a decision if secondary 
procedures are indicated. 

b. End-to-end repair 

If a lateral arteriorrhaphy or venor- 
rhaphy is not possible, an end-to-end 
repair is preferable if possible. Such 
an anastomosis must be tension free. 
When debridement has occurred, it 
may be very difficult to bring the vessel 
ends together into a tension-free anas- 
tomosis. Mobilization of an artery by 
tying off branches in order to "gain 
length" is time consuming and often 
leads to the need for revision. Our pref- 
erence is, when an artery is completely 
transected and debrided, to consider 
the insertion of a substitute conduit. 
Often a microscope or magnifying 
loupes are used for very small vessels. 
For very small vessel anastomoses, an 
interrupted suture line has greater 
long-term patency. Precision in the 
performance of a vascular anastomo- 
sis is paramount to immediate and 
long-term patency. The smaller the 
vessel, the more unforgiving of lack 
of precision and attention to detail. 
As with the use of Fogarty balloons, a 
number of principles exist relating to 
the performance of a vascular anas- 
tomosis (Box 7-5). With an exercise 
of precision and abiding by these prin- 
ciples, the surgeon should expect a 
high degree of success from the vas- 
cular anastomosis. Reasons for failure 
include lack of distal flow, lack of 
inflow, narrowed anastomosis, pres- 
ence of distal clot, and kinking of the 
conduit, among others. 

For a continuous anastomosis, the 
surgeon should use "triangulation" or 
lateral "stay" suture techniques to 
ensure that the anastomosis is not nar- 
rowed. The first assistant must be vig- 
ilant to ensure that the continuous 
anastomosis is not "purse stringed" by 
the assistant pulling to tightly on the 
suture as he or she follows for the 



ch07.qxd 4/16/04 3:30PM Page 161 



7 • ACCESS, CONTROL AND REPAIR TECHNIQUES 



161 



PRINCIPLES RELATING TO THE PERFORMANCE OF 
A VASCULAR ANASTOMOSIS 



The anastomosis must be tension free. 

Consideration must be given to the various positions of the adjacent and 
distal anatomy after repair. 

Consideration should be made for redundancy to allow for full extension 
of an extremity after reconstruction. 

Consideration must be made regarding coverage of the repair. 

The suture material must be nonabsorbable. 

The finest suture material to accomplish a permanent anastomosis should 
be selected. 

The needle size and shape should be chosen to maximize an ideal 
anastomosis. 

The needle must enter the vessel at a right angle. 

The rotation on the needle should follow the curve of the needle. 

The needle should be grasped with the needle driver somewhere between 
the middle of the curve and the tip. 

The suture material should not be used to "pull" the anastomosis together 
but lie together without tension. 



surgeon. Another consideration is to 
spatulate the anastomosis to make the 
anastomosis actually larger than the 
repaired vessel. Some end-to-end 
anastomoses are accomplished using 
vascular staples. Some appropriately 
chosen techniques should be used to 
determine the adequacy of the anas- 
tomosis at the completion of the 
procedure. Some surgeons choose 
Doppler ultrasound, and others will 
use arteriography, depending on the 
size of the vessel. 

Insertion of a substitute conduit 

A substitute interposition conduit has 
been used extensively in vascular 
trauma. An interposition (end-to-end, 
end-to-side, or side-to-side) conduit is 
used when extensive destruction exists 
and one of the other reconstruction 
options does not exist. Considerable 
discussion, debate, and research have 
focused on the synthetic versus auto- 
genous conduits. This concern basi- 



cally comes down to a consideration 
in only two locations, the superficial 
femoral and the subclavian arteries. In 
the trunk, use of PTFE or Dacron pros- 
theses are an issue of size match and 
durability. Long-term favorable results 
have been extensively reported. In the 
neck, distal extremities, and smaller 
truncal arteries, the size match of 
currently available prostheses is 
unacceptable and use of the scavenged 
saphenous vein is most appropriate. 
Currently, for vessels 5 mm or smaller, 
the use of the saphenous vein is the 
preferred conduit. 

Debate has also occurred regarding 
which graft material to use in the 
presence of potential infection. One 
option would be to use ligation and 
extra-anatomic routing around the 
area of infection when potential or real 
infection occurs. In some instances, 
such as reconstruction of an injured 
abdominal aorta, it is virtually im- 
possible to avoid reconstruction in an 



ch07.qxd 4/16/04 3:30PM Page 162 



162 



II • PRINCIPLES OF OPERATIVE CARE 



area of potential infection. In other 
instances, infection is not a consider- 
ation until a graft is later exposed, or 
a secondary infection or abscess occurs 
in an area of infection. Another argu- 
ment has been whether venous or 
arterial autografts are "living" at the 
time that the conduit is scavenged and 
repositioned elsewhere, devoid of its 
vasovasorium. A case can be made that 
this (foreign body) collagen tube 
becomes "living" after it has become 
re-endothelized at a later date. If this 
is correct, all substitute conduits — 
regardless of being autologous, 
homologous, frozen, xenographic, or 
manufactured — the infectious risk 
should be similar. Despite the use of 
Dacron substitute conduits in the 
injured abdominal aorta for almost all 
reported successfully managed cases 
and a high rate of enteric contamina- 
tion at the time of implantation, no 
infected aortic grafts in these cases 
have been documented in the litera- 
ture. Other synthetic grafts, such as 
those constructed with PTFE, have 
been percutaneously punctured and 
yet have a suggested "resistance to 
infection." More than 30 laboratory 
studies have been reported in which 
purposeful infections have been 
created around grafts, comparing 
synthetic with autogenous material. 
The infectivity is almost identical, but 
the complications are different, both 
in the laboratory and in people. 
With synthetic conduits, perigraft 
infections result in either suture line 
aneurysms, thrombosis of the graft, or 
occasionally sepsis from chronic graft 
infection. With "autogenous" con- 
duits, periconduit infections result 
in dissolution of the collagen tube, 
distal embolization, and often exsan- 
guinating hemorrhage, sometimes 
uncontrollable. 

Although end-to-end anastomoses 
are often used at both ends of the sub- 
stitute conduit, consideration for an 
end-to-side reconstruction at either 
end or both ends of the conduit war- 



rants consideration. In some instances, 
such as injury to the popliteal artery, 
immediately behind the knee, this 
variation offers an additional option 
with long-term favorable results. 

d. Patch angioplasty 

Patch angioplasty using autogenous 
venous material is actually used very 
infrequently in vascular trauma. When 
it is used, it is often as a secondary 
procedure to correct a narrowing at a 
previous reconstruction. With current 
technology, a catheter-based inter- 
ventional dilation and stenting would 
precede a secondary open procedure 
to widen a previously constructed vas- 
cular repair. 

1 2 . Flush proximally and distally before com- 
pletion of the suture line. 

Before the completion of the suture 
line, the proximal and distal clamps are 
temporally removed to ensure that pro- 
grade and retrograde back bleeding 
occurs. If there is no back bleeding from 
the distal suture line, one might 
consider another pass of the Fogarty 
catheter. 

1 3. Complete the suture line/clamp removal. 

When the surgeon is ensured that 
inflow and outflow are adequate, the 
suture line is completed and the clamps 
are removed. 

14. Assess the distal circulation. 

After completion of a vascular recon- 
struction, the surgeon must evaluate the 
adequacy of the anastomosis for any 
stenosis, kinking of the prosthesis, and 
patency of the distal outflow tract. This is 
best accomplished using completion arte- 
riography, which despite the availability 
of Doppler and ultrasound technologies 
still remains the "gold standard." 
However, in the critically injured patient, 
there may not be time for a completion 
study. When dealing with large arteries, 
such as the iliac, subclavian, and femoral 
vessels, a good distal pulse and a normal 
triphasic Doppler signal are often taken 



ch07.qxd 4/16/04 3:30PM Page 163 



7 • ACCESS, CONTROL AND REPAIR TECHNIQUES 



163 



as evidence of a technically satisfactory 
repair. The best time to accomplish any 
needed re-reconstruction is at the time 
of the first operation. 

15. Consider the need for fasciotomy. 

Fasciotomy and compartment syndromes 
are addressed in Chapter 23. In cases of 
an ischemic limb for more than 4 hours, 
the surgeon is well advised to consider per- 
forming a fasciotomy before a vascular 
reconstruction. In cases of ligation of an 
outflow vein, especially the iliac, axillary, 
deep femoral, or popliteal veins, apostre- 
pair fasciotomy is strongly encouraged. If 
there be any concern for the performance 
of a fasciotomy, compartment pressures 
can be measured. Although debate exists 
concerning the exact pressures where a 
fasciotomy must be performed, com- 
partment pressures of more than 30 cm 
H 2 should cause the surgeon to strongly 
consider the procedure. 

16. Ensure appropriate coverage. 

A cardinal principle in vascular trauma 
is that a vascular reconstruction must 
always be covered with viable soft tissue; 
otherwise, failure with catastrophic bleed- 
ing is all but certain. Coverage can be 
achieved using various techniques, but in 
the presence of massive soft tissue destruc- 
tion, covering an arterial graft with viable 
soft tissue can be both challenging and 
time consuming. Most often, coverage is 
with the tissue within the operative field, 
which in the normal closure adequately 
covers the vascular repair. On occasion, 
special flaps will be necessary to bring vas- 
cularized pedicles over the reconstruc- 
tion. In extremely rare situations, use of 
porcine xenograft or homograft mater- 
ial might be neces-sary to temporarily 
cover a vascular reconstruction. 

The trauma surgeon undertaking avas- 
cular reconstruction must keep in mind 
the importance of soft tissue coverage 
because occasionally an unusual or 
unorthodox extra-anatomic route will be 
selected for the graftjust because the con- 
ventional anatomic route is exposed or 
will present a cover problem. 



SUMMARY 



This chapter is intended to communicate 
the fundamentals required by a general 
surgeon approaching a patient with a sus- 
pected or proven vascular injury upon 
arrival in the OR. The principle of initial 
control of external hemorrhage is followed 
by considerations for positioning on the 
OR table and determination of incision 
placement. 

Fundamental in approaching a vascular 
injury is initially obtaining access and control 
away from the area of suspected injury so no 
additional injury ensues as the area of specific 
injury and hemorrhage is dissected. An area- 
by-area review of some general access and 
control suggestions is provided for specific 
injuries. 

Finally, some general repair techniques 
are presented, which are standard for the 
reconstruction of any vascular injury. 



REFERENCES 

Buckman RF, Miraliakbari R, Badellino MM: 
Juxtahepatic venous injuries: A critical review 
of reported management strategies. J Trauma 
2000;48:978-983. 

Burch JM, Richardson RJ, Martin RR, Mattox KL: 
Penetrating iliac vascular injuries: Recent expe- 
rience with 233 consecutive patients. J Trauma 
1990;30:1450-1459. 

Feliciano DV, Burch JM, Mattox KL, et al: Balloon 
catheter tamponade in cardiovascular wounds. 
Am J Surg 1990;160:583-587. 

Henry AK: Extensile Exposure, 2nd ed. Baltimore: 
Williams & Wilkins, 1957. 

Hoyt DB, Coimbra R, Potenza BM, Rappold JF: 
Anatomic exposures for vascular injuries. Surg 
Clin North Am 2001 Dec;81(6):1299-1330. 

Martin RR, Barcia PJ, Johnson EA: Making 
matters worse: Complications of initial evalua- 
tion, treatment and delayed diagnosis. In Mattox 
KL (ed) : Complications of Trauma. New \brk, 
Churchill Livingstone, 1994, pp 139-154. 

Mattox KL: Red River anthology. J Trauma 1997; 
42:353-368. 

Mattox KL, Hirshberg A: Vascular trauma in vas- 
cular surgery. In Haimovici H, Ascer E, Hollier 
LH, et al (eds) : Haimovici's Vascular Surgery — 



ch07.qxd 4/16/04 3:30PM Page 164 



164 



II • PRINCIPLES OF OPERATIVE CARE 



Principles and Techniques, 4th ed. Cambridge, 
Mass, Blackwell Scientific, 1995. 

Mattox KL, McCollum WB, Jordan GL Jr, et al: 
Management of penetrating injuries of the 
suprarenal aorta. J Trauma 1975;15:808-815. 

Rutherford RB: Atlas of Vascular Surgery. Basic 
Techniques and Exposures. Philadelphia, 
Harcourt Brace Jovanovich, 1993. 

Surgical exposure of vessels. In Haimovici H, 
Ascer E, Hollier LH, et al (eds): Haimovici's 



Vascular Surgery — Principles and Techniques, 
4th ed. Cambridge, Mass, Blackwell Scientific, 
1995, pp 351-420. 

Veith FJ, Gupta S, Daly V: Technique for oc- 
cluding the surpaceliac aorta through the 
abdomen. Surg Gynecol Obstet 1980;151(3): 
426-428. 

Wind GG, Valentine RJ: Anatomic Exposures in 
Vascular Surgery. Baltimore, Williams &Wilkins, 
1991. 



ch08.qxd 4/16/04 3:31PM Page 165 




Damage Control for 
Vascular Trauma 



ASHER HIRSHBERG 
BRADFORD G. SCOTT 





o 


INTRODUCTION 




o 


EVOLUTION OF THE DAMAGE-CONTROL CONCEPT 

The Physiologic Envelope 

Practical Application of "Damage Control" 




o 


VASCULAR REPAIR TECHNIQUES 


o 


TEMPORARY SHUNTS 


o 


POSTOPERATIVE LIMB ISCHEMIA 


o 


PLANNED REOPERATION 



INTRODUCTION 



"Damage control" is a surgical strategy for the 
staged management of multivisceral trauma 
that represents a major paradigm shift in 
trauma surgery. With this approach, the tra- 
ditional single definitive operation is replaced 
by a staged repair, whereby a rapid "bailout" 
operation (to control hemorrhage and 
spillage) is followed by a delayed reconstruc- 
tion after the patient's physiology has been 
stabilized. In the last decade, this approach 
has become part of the standard repertoire 



of trauma surgeons when operating on their 
most critically wounded patients. 

Major vascular trauma is often part of the 
injury complex in patients with exsanguinat- 
ing hemorrhage in whom the damage-control 
approach is the patient's only hope. There- 
fore, a detailed acquaintance with this strat- 
egy and its application to the management of 
arterial and venous injuries is mandatory for 
every surgeon involved in trauma care. 

Damage control for vascular injuries is par- 
ticularly challenging because of the inherent 
conflict between the need for a precise and 
time-consuming vascular reconstruction on 

165 



ch08.qxd 4/16/04 3:31PM Page 166 



166 



II • PRINCIPLES OF OPERATIVE CARE 



the one hand and the urgency of an abbrevi- 
ated procedure on the other. However, the 
futility of attempting a complex arterial repair 
in the presence of diffuse coagulopathy should 
be quite obvious even to a surgeon who is unac- 
quainted with the damage-control strategy. 

The concept of staged repair in emergen- 
cies is not new to vascular surgeons. For 
example, the current operative management 
of infected intra-abdominal aortic grafts often 
consists of a two-stage operation whereby an 
extra-anatomic bypass is inserted first, and 
removal of the infected graft is delayed for a 
subsequent procedure. The reason for this is 
not technical, but it is the desire to avoid a 
huge physiologic insult in a compromised 
patient. Similarly, a staged repair of a bleed- 
ing aortoduodenal fistula (the first operation 
consisting of temporary control of bleeding 
and of the duodenal perforation much like 
in a damage-control procedure) has been 
advocated as a more effective approach than 
the traditional one-stage operation because 
the results of the latter carry a prohibitive 
mortality. 

This chapter presents the general philoso- 
phy of damage control and the underlying 
physiologic considerations that form the ratio- 
nale for employing the strategy. This will then 
serve as a background for a discussion of the 
application of damage-control principles to 
the modern management of vascular trauma, 
and a detailed description of specific "bailout" 
techniques for the management of major arte- 
rial and venous injuries. 



EVOLUTION OF THE DAMAGE- 
CONTROL CONCEPT 



During the past 2 decades, civilian trauma sur- 
geons have encountered new wounding pat- 
terns characterized by high-energy transfers 
(from automaticweaponsandfastmotorvehi- 
cles) causing extensive damage to multiple 
organs and massive blood loss. These exsan- 
guinating patients, who previously would have 
died before reaching the hospital, are now 
rapidly transferred to trauma centers by 
efficient prehospital systems, presenting sur- 
geons with an unusual array of challenges. The 



conventional operative sequence for trauma, 
consisting of rapid access, bleeding control, 
and reconstruction, is inappropriate in these 
exsanguinating patients. Such definitive repair 
usually requires lengthy and complex proce- 
dures, which these critically ill patients will 
not tolerate. The result of heroic attempts at 
definitive repairs has typically been early post- 
operative death due to "irreversible shock," 
diffuse coagulopathy bleeding or multiple 
organ system failure. These considerations 
have led to the development of the damage- 
control approach, a modified operative 
sequence whereby only immediately life- 
threatening visceral injuries are addressed 
using rapid temporary lifesaving measures. 
The patient is then transferred to the surgi- 
cal intensive care unit (ICU) for rewarming 
and resuscitation, and definitive repair of the 
injuries is postponed until reoperation can be 
performed on a nonbleeding, stable patient 
with restituted physiologic parameters (see 
Table 8-1). 

Damage control represents a profound 
change in the way trauma surgeons view their 
role in the operating room. The center of 
attention has shifted from reconstruction 
of the anatomy to restitution of the injured 
patient's physiologic reserves. In other words, 
the completeness of the anatomic repair is 
temporarily sacrificed to address the physio- 
logic insult before it becomes irreversible. 
Herein lies the fundamental difference 
between the traditional approach of a single 
definitive procedure and the damage-control 
approach of a staged repair. 

Trauma surgeons were slow to adopt this 
unconventional strategy because abrupt 
termination of an "unfinished" operation and 
acceptance of a temporary and anatomically 
incomplete repair seemed to contrast with tra- 
ditional surgical values. This is why almost a 
decade passed between the original descrip- 
tion of the strategy in patients with coagu- 
lopathy by Stone, Strom, and Mullins in 1983 
and the publication of the first large series in 
the early 1990s. Gradual adoption of damage 
control as a valid alternative to the traditional 
definitive operation evolved slowly (see Table 
8-1). In the mid-1990s, the new approach was 
expanded to the management of urologic, tho- 
racic, vascular, and even limb injuries. 



ch08.qxd 4/16/04 3:31PM Page 167 



8 • DAMAGE CONTROL FOR VASCULAR TRAUMA 



167 



TABLE 8-1 

THE EVOLUTION OF "DAMAGE CONTROL" 



Period 

1983 
1982-1990 

1991 

1992 

1992-1994 

1994-1997 



1997-Present 



Key Development 

Staged approach to coagulopathy 

Sporadic technical reports 
Balloon catheter tamponade 

Largest series (200 patients) and 
philosophy explained 

"Damage control" coined 

New concept gains acceptance 

Extension outside abdomen 



Attempts to define physiological 
envelope 



References 



Stone, Strom, and Mullins 
Feliciano and colleagues (1990) 

Burch and colleagues (1992) 

Rotondo and colleagues (1993) 

Morris and colleagues (1993) 

Hirshberg et al (7) 
Wall and colleagues (1994) 
Porter and colleagues (1997) 
Scalea and colleagues (1994) 

Cosgriff and colleagues (1997) 
Cushman and colleagues (1997) 
Garrison and colleagues (1996) 



The Physiologic Envelope 

The concept of the "physiologic envelope" is 
key to understanding the rationale of damage 
control. Despite technologic advances, the 
operating room remains a physiologically 
unfavorable environment for the severely 
wounded patient. Extensive peritoneal expo- 
sure during a trauma laparotomy results in 
accelerated heat loss, which is further aggra- 
vated by massive transfusion. Hypothermia in 
turn impairs blood clotting and thus con- 
tributes to ongoing hemorrhage. Shock leads 
to metabolic acidosis and a subsequent need 
for further transfusion. The most obvious 
manifestation of the injured patient's physi- 
ologic derangement is, therefore, the triad of 
hypothermia coagulopathy and acidosis. 
Together these derangements create a self- 
propagating vicious cycle that eventually leads 
to an irreversible physiologic insult. This irre- 
versibility may present intraoperatively as 
diffuse bleeding that cannot be controlled sur- 
gically, followed by refractory ventricular 
arrhythmias and death. More commonly, the 
patient survives the operation only to exhibit 
a refractory systolic blood pressure of 60 to 
80mmHg, oliguria, peripheral vasocon- 
striction, massive swelling, progressive 



hypoxemia, and diffuse oozing from every inci- 
sion and vascular access site. Death almost 
invariably ensues within the first few postop- 
erative hours. 

Thus, the triad of hypothermia, coagu- 
lopathy, and acidosis defines the patient's 
physiologic envelope, a set of physiologic 
parameters that together mark the boundary 
between a survivable physiologic insult and 
an irreversible derangement. Termination of 
the operative procedure before this physio- 
logic envelope is breached is the essence of 
damage control. 

Hypothermiahas emerged as a central patho- 
physiologic event in exsanguinating trauma 
patients. Shocked patients with penetrating 
torso injuries lose body heat to a mean tem- 
perature of 34.5°C by the time they reach the 
operating room. The ambient temperature in 
the operating room is around 22°C, and rapid 
infusion of crystalloids or blood without a 
warming device contributes to the fast devel- 
opment of hypothermia. The open peri- 
toneal cavity itself is also a major source of 
accelerated heat loss. It has been clearly shown 
that in the severely injured patient, hypother- 
mia is harmful and adversely affects survival 
independent of injury severity. Of the three 
components of the physiologic envelope, 



ch08.qxd 4/16/04 3:31PM Page 168 



168 



II • PRINCIPLES OF OPERATIVE CARE 



hypothermia is the only one for which there 
is a well-defined threshold value. In 1987, 
Jurkovich and colleagues convincingly demon- 
strated that in severely wounded patients 
undergoing laparotomy, a core temperature 
less than 32°C is associated with 100% mor- 
tality. Based on this observation, a mathe- 
matical model of intraoperative heat loss 
during laparotomy for exsanguinating hem- 
orrhage predicts a window of opportunity of 
no more than 60 to 90 minutes before this 
threshold is reached. 

Coagulopathy typically presents as diffuse 
oozing inside and outside the operative field. 
Attention has focused on hypothermia as the 
cause of coagulopathy in trauma. Hypother- 
mia affects clotting through alteration of 
platelet function and inhibition of the coag- 
ulation cascade. In the hypothermic patient, 
platelets are sequestered in the liver and spleen 
and exhibit marked morphologic changes. 
Platelet activation is inhibited resulting in pro- 
longation of the bleeding time and other 
abnormal platelet function test results. The 
enzymes of the coagulation cascade are tem- 
perature sensitive and therefore are inhibited 
during hypothermia. However, both platelet 
dysfunction and enzyme inhibition become 
clinically important only when the core tem- 
perature drops to less than 32°C, which is well 
below the usual range seen in the severely 
injured. 

Hemodilution is another important cause 
of coagulopathy in exsanguinating patients. 
Extensive blood loss and massive replacement 
with packed cells and crystalloids combine 
to produce rapid "washout" of platelets and 
clotting factors. Because many of the pa- 
tients undergoing damage-control operations 
require massive transfusion, and because the 
actual blood volume of these patients changes 
rapidly and is difficult to quantify, dilution is 
probably an underestimated contributor to 
coagulopathy. Hypothermia and dilution also 
have been clearly shown to have an additive 
effect in causing clotting abnormalities. 

Coagulopathy in the critically injured 
patient is a clinical and not a laboratory diag- 
nosis. Standard coagulation tests often fail to 
reflect the full magnitude of the clotting dis- 
order in these patients because they are rou- 
tinely conducted at 37°C and often take too 



long to be useful guides for real-time replace- 
ment of clotting factors in the exsanguinat- 
ing patient. 

Lactic acidosis is the result of anaerobic gly- 
colysis and reflects inadequate tissue perfu- 
sion. Acidosis adversely affects myocardial 
contractility and cardiac output in animal 
models, but the full scope of its physiologic 
and metabolic effects remains unclear. Aci- 
dosis is a useful measure of the severity of shock 
and a reliable predictor of survival. Serum 
lactate levels, base deficit, and the time inter- 
val to normalization of the serum lactate have 
all been shown to closely correlate with mor- 
tality from severe trauma in both animal 
and clinical studies. However, no well-defined 
threshold value for lactic acidosis can serve 
as a marker of irreversible shock. 

Several attempts were made to better define 
the physiologic envelope. Cosgriff and 
colleagues (1997) analyzed prospectively col- 
lected physiologic data from 58 injured 
patients who received massive transfusions, 
and they identified four significant risk factors 
that predict the onset of coagulopathy: pH < 
7.10, temperature <34°C, Injury Severity Score 
>25, and systolic blood pressure <70mmHg. 
About one in four severely injured patients 
requiring massive transfusion developed coag- 
ulopathy, but when all four risk factors were 
present, the probability of developing coag- 
ulopathywas98%. In another study, Cushman 
and colleagues (1997) attempted to quantify 
the physiologic envelope in a series of 53 
patients with iliac vascular injuries. Their study 
showed that an initial pH level of less than 
7.1 and a final operating room temperature 
of less than 35 °C were the best predictors 
of imminent death. 



Practical Application of 
"Damage Control" 

The damage-control sequence consists of 
three phases: initial operation, surgical ICU 
resuscitation, and planned reoperation. The 
initial operation is typically a rapid "bailout" 
procedure in which the surgeon does only 
the absolute minimum necessary to save the 
patient's life. Rapid temporary techniques are 
used to control bleeding, prevent spillage of 



ch08.qxd 4/16/04 3:31PM Page 169 



8 • DAMAGE CONTROL FOR VASCULAR TRAUMA 



169 



intestinal content or urine, restore blood 
flow to vital vascular beds, and achieve rapid 
closure of the abdomen or chest. Time- 
consuming formal resections and recon- 
structions are deliberately avoided. 

Bleeding from solid organs (such as the 
liver) or diffusely oozing cavities (such as 
the retroperitoneum) is controlled by packing. 
Spillage of intestinal content is controlled by 
ligation or stapling of bowel injuries without 
resection, or by external tube drainage of 
duodenal, pancreatic, and common bile duct 
injuries. Similar spillage-control techniques 
have been applied to injuries of the urinary 
tract. In the chest, stapled nonanatomic lung 
resection and laying open a bullet tract 
through the lung parenchyma to control 
bleeding (instead of resection) enable rapid 
termination of the operative procedure in 
accordance with damage-control principles. 

Closure of the injured cavity is performed 
rapidly using temporary measures, such as 
skin-only closure by a running monofilament 
suture. In the presence of massive visceral 
edema that precludes skin closure without 
tension, plastic silos or absorbable mesh is used 
to temporarily accommodate and protect the 
edematous viscera. 

The second phase of the sequence is re- 
suscitation in the surgical ICU. Aggressive 
correction of hypothermia is the most impor- 
tant consideration in the early postoperative 
period. This can usually be achieved using vig- 
orous external rewarming, but arteriovenous 
rewarming can greatly expedite the process 
in severely hypothermic patients. Empirical 
replacement of blood, plasma, and platelets 
is equally important to restore normal hemo- 
stasis. Support of the cardiovascular system 
focuses initially on volume replacement. The 
early use of invasive cardiovascular monitor- 
ing (a soon as the patient's coagulopathy is 
corrected) may be a useful adjunct. To achieve 
a favorable outcome, these patients require a 
direct and massive investment of bedside time 
and continuous direct involvement of the 
trauma team in the early postoperative period. 

Not uncommonly, patients may require an 
urgent (unplanned) reoperation during the 
second phase of the damage-control sequence. 
The main indication for urgent reoperation 
is ongoing hemorrhage. This is usually the 



result of either failed hemostasis during 
the "bailout" procedure, a missed injury, or 
an iatrogenic trauma. Other indications for 
an urgent reoperation are intra-abdominal 
hypertension and limb ischemia distal to an 
indwelling temporary intraluminal shunt. 

Planned reoperation is undertaken in a 
stable patient, usually within 2 to 3 days of the 
initial "bailout" procedure. The aims at this 
stage are to perform a definitive repair of the 
injuries and to accomplish formal closure of 
the visceral cavity. 

Although most trauma cases are effectively 
managed using the traditional approach of a 
single definitive operation, the damage- 
control approach is indicated only in a small 
group of the most critically injured patients, 
and one of the major problems facing the 
surgeon is deciding when to employ it. 

Formally stated, damage controlh indicated 
when the magnitude of the visceral damage 
is such that definitive repair of all injuries 
is likely to exceed the patient's physiologic 
limits. However, this is a simplified definition 
of a complex and multidimensional dilemma. 
Making the decision early, within a few minutes 
of entering the injured cavity, is one of the 
keys to successful damage control. Garrison 
and colleagues (1996) have shown that an 
early decision to perform packing is an impor- 
tant determinant of survival in abdominal 
trauma, because deterioration in coagulation, 
low pH level, and long duration of hypoten- 
sion are all associated with a decreased chance 
of survival. 

Because no good qualitative definition of 
the point at which the physiologic insult 
becomes irreversible is available, an early deci- 
sion to "bail out" must rely on recognition of 
typical injury patterns that require damage 
control rather than on physiologic parame- 
ters. The combination of a major intra-abdom- 
inal vascular injury with hollow or solid-organ 
damage is a class injury pattern in which an 
early decision to "bail out" is often lifesaving. 
However, isolated major vascular injuries can 
usually undergo a definitive repair even in a 
patient who has sustained a massive amount 
of blood loss because bleeding is controlled, 
resuscitation can be accomplished intraop- 
eratively, and the definitive repair can 
be accomplished relatively quickly. Other 



ch08.qxd 4/16/04 3:31PM Page 170 



170 



II • PRINCIPLES OF OPERATIVE CARE 



patterns include destruction of the pancre- 
aticoduodenal complex, a high-grade hepatic 
injury, retroperitoneal or pelvic bleeding, and 
injuries to multiple visceral compartments. 



VASCULAR REPAIR 
TECHNIQUES 

The application of damage-control principles 
to vascular trauma hinges on a clear distinc- 
tion between two categories of vascular repairs: 
simple and complex. Simple repairs are rapid 
and straightforward and include lateral repair, 
ligation, and temporary intraluminal shunt 
insertion. These techniques are not time con- 
suming, do not create long suture lines, and 
can be used even in the presence of diffuse 
coagulopathic bleeding or unfavorable phys- 
iology. Complex repairs include vascular 
reconstructions such as end-to-end anasto- 
mosis, patch angioplasty, and graft interposi- 
tion. These techniques are usually poor 
options in the hypothermic coagulopathic 
patient not only because they result in ongoing 
oozing from the suture lines but also because 
they are time consuming and significantly 
prolong the "bailout" procedure. This 
unorthodox approach represents a sharp 
deviation from the standard principles of vas- 
cular reconstruction but is eminently applic- 
able to the damage-control scenario, in which 
not everything that is technically possible is 
in the patient's best interest. 

Lateral repair is feasible in the absence of 
complete transection or extensive destruction 
of the arterial wall. It is important to main- 
tain the orientation of the repair perpendic- 
ular to the axis of the vessel, to avoid stenosis. 

Ligation is an underused option in the 
severely injured patient, especially when the 
injured vessel is relatively inaccessible or a 
complex repair is required. All limb veins can 
be ligated with impunity, and certainly in the 
context of damage control, reconstructing a 
peripheral vein is unjustified. The subclavian 
and iliac veins and the inferior vena cava can 
be rapidly ligated with the acceptable price 
of postoperative limb edema. Ligation of the 
portal and superior mesenteric veins is a valid 
option in the patient in extremis, but this results 



in massive third spacing that requires very 
aggressive fluid resuscitation in the post- 
operative phase. 

Many injured arteries can also be ligated 
with impunity. The external carotid artery is 
an obvious example. In the context of pene- 
trating trauma, injury to the inaccessible retro- 
mandibular part of the internal carotid artery 
(in zone 3) is managed by ligation or balloon 
tamponade, with the calculated risk of a 
neurologic deficit weighted against the neces- 
sity of obtaining rapid hemostasis. In most 
patients, ligation of the subclavian artery does 
not result in critical ischemia of the upper 
extremity because of the ample collateral cir- 
culation around the shoulder. The amputa- 
tion rates following ligation of the femoral 
arteries were 81% for the common femoral 
and 55% for the superficial femoral artery, 
based on data from World War II (before the 
advent of fasciotomy). When a major limb 
artery is ligated during a damage — control 
procedure, it is usually prudent to proceed 
with an immediate fasciotomy. 

Ligation of the proximal suprapancreatic 
superior mesenteric artery has been reported 
as a valid technical alternative in critically 
injured patients who are unlikely to tolerate 
a lengthy reconstruction because the rich col- 
lateral blood supply from the inferior mesen- 
teric and celiac arteries will maintain midgut 
viability. The celiac axis can be ligated with 
impunity, and complex repair of renal artery 
injury in the exsanguinating patient with 
multiple injuries should not be attempted. 

An effective alternative to ligation in inac- 
cessible sites is balloon catheter tamponade, 
a simple and effective vascular damage-control 
technique. A Foley or large Fogarty balloon 
catheter is inserted into the tract of the injur- 
ing missile, and the balloon is inflated until 
hemorrhage is controlled. Balloon tampon- 
ade can be either a temporary hemostatic 
maneuver or even a definitive management 
of an inaccessible injury (Fig. 8-1). Balloons 
have been successfully used to control bleed- 
ing from the carotid artery high in the neck, 
from inaccessible pelvic vessels, or from a trans- 
fixing liver injury. 

Another useful hemostatic technique is 
packing. Although traditionally employed to 
achieve hemostasis from high-grade liver 



ch08.qxd 4/16/04 3:31PM Page 171 



8 • DAMAGE CONTROL FOR VASCULAR TRAUMA 



171 




■ FIGURE 8-1 

Balloon catheter tamponade of the distal internal carotid artery in zone 3 of the neck. 



injuries, packing is a very useful adjunct in a 
coagulopathy patient with a limb injury and 
ongoing hemorrhage from multiple muscu- 
lar bleeders that cannot be controlled directly. 



TEMPORARY SHUNTS 



Intraluminal shunts are prosthetic conduits 
placed within the vessel lumen across an 
injured segment to temporarily reestablish 
blood flow until a definitive vascular recon- 
struction can be performed. Vascular surgeons 
have used temporary shunts for at least 
3 decades. In 1971, Eger and colleagues 
described the use of a shunt in a series of 
popliteal artery injuries to maintain limb per- 
fusion while the bones are aligned before vas- 
cular repair. This series was the first modern 
report of the use of shunts in vascular trauma. 
It has recently been shown in an experimen- 
tal study that a temporary shunt provides 
approximately half the blood flow of the intact 
vessel and that increased oxygen extraction 
compensates for the lower flow. 



The choice of shunt material is a matter of 
personal preference, because any rigid smooth 
synthetic tube of appropriate caliber can serve 
as a temporary vascular conduit. The original 
description by Eger and colleagues (1971) was 
of a polyethylene tube with a side port. The 
side port facilitates access for monitoring of 
flow or flushing of the lumen. Others have 
used commercially available carotid shunts 
(such as ajavid or a Sundt shunt) or a heparin- 
bonded catheter or have improvised with a 
segment of a suction catheter or small Ar gyle's 
catheter cut to the appropriate length. 

Temporary shunts remained functional for 
as long as 24 hours in an animal model. In 
clinical practice, temporary shunts remain 
functional for many hours, and patency as late 
as 36 hours after insertion has been observed. 
The presence of coagulopathy in the critically 
injured usually prevents clotting of the shunt 
during the first postoperative hours, so early 
postoperative failure is usually the result of a 
technical error during insertion. Temporary 
shunt insertion is an excellent damage-control 
option because it is rapid, controls hemor- 
rhage from the injured vessel, and preserves 



ch08.qxd 4/16/04 3:31PM Page 172 



172 



II • PRINCIPLES OF OPERATIVE CARE 




■ FIGURE 8-2 

A temporary intraluminal shunt (Argyle's tube) 
maintaining flow across a transected brachial 
artery. ■ 



distal flow while keeping all future recon- 
structive options open for the surgeon 
(Fig. 8-2). 

Shunts are also used in the surgical repair 
of combined orthopedic and vascular injuries. 
Here the preferred sequence would be to 
achieve bone alignment before arterial recon- 
struction, but often the extremity is grossly 
ischemic and requires immediate restoration 
of distal flow. Shunt insertion allows fracture 
fixation to proceed and allows a subsequent 
vascular reconstruction once the bones are 
properly aligned. 

Temporary shunts have been used in the 
management of brachial, iliac, femoral, and 
popliteal arterial injuries. A single case of 
the successful use of a temporary shunt for 
superior mesenteric artery injury has been 
reported. Several attempts have been made 
to use an Argyle chest tube as a temporary 
shunt across an injury to the abdominal aorta 
in patients in extremis, none of whom survived 
beyond the immediate postoperative period. 

Insertion of a temporary intraluminal shunt 
begins with proximal and distal control of the 
injured segment. Typically, the injured artery 
will be completely or almost completely tran- 
sected. The injury should be carefully assessed 



and the inflow and outflow tracts to the 
damaged segment should be cleared by a 
Fogarty balloon thrombectomy. A shunt of the 
appropriate diameter is then gently inserted 
distally, flushed retrograde, and then inserted 
proximally. Special care is taken to avoid 
raising an intimal flap or causing additional 
injury to the vessel. The shunt can be secured 
in place either with heavy silk ligatures or vessel 
loops held in place by a Rummel tourniquet. 
The former technique is simpler but more 
traumatic to the arterial wall. A central heavy 
silk ligature placed around the mid-body of 
the shunt is helpful for manipulation and 
serves as a marker for proximal or distal migra- 
tion of the conduit. Once the shunt is in place, 
distal perfusion should be confirmed by pal- 
pation of a distal pulse or obtaining a Doppler 
signal distal to the injured segment. Systemic 
heparin is not administered. 

The two major postoperative concerns are 
shunt dislodgment and thrombosis. Dislodg- 
ment of the shunt is rare and usually results 
from inadequate fixation of the shunt in place. 
There is sudden gross swelling of the involved 
extremity with oozing between the skin 
sutures, indicating the presence of a rapidly 
expanding hematoma. Immediate reexplo- 
ration is indicated to obtain hemostasis and 
reinsert the shunt. 

Early shunt failure is usually caused by a 
technical problem, and much like with early 
postoperative failures of arterial reconstruc- 
tions, the cause is poor inflow, a problem with 
the shunt itself, or inadequate outflow. Poor 
inflow or outflow can be the result of an intimal 
flap, a more proximal or distal injury, or a 
residual thrombus (proximal or distal to the 
shunt). The shunt itself can also be occluded 
by tying the ligatures that fix it in place too 
tightly, by angulation from excessive length, 
or by migration of the shunt into a distal arte- 
rial branch. 



POSTOPERATIVE LIMB 
ISCHEMIA 

Limb ischemia is the major concern after 
damage-control procedures that include a vas- 
cular component. Ischemia may be the result 



ch08.qxd 4/16/04 3:31PM Page 173 



8 • DAMAGE CONTROL FOR VASCULAR TRAUMA 



173 



of intentional ligation of the injured artery, a 
clotted temporary shunt, a failed repair, or a 
missed injury. The major considerations in the 
management of limb ischemia in the damage- 
control context are the same regardless of the 
etiology. The first obvious step is the diagno- 
sis of limb ischemia. Normal peripheral pulses 
are rarely palpable in these patients, even in 
the absence of an arterial injury. Hypotension, 
hypothermia, peripheral vasoconstriction, 
and edema of the injured extremity all 
combine to make the diagnosis of limb 
ischemia quite difficult in the critically 
wounded. Therefore, it is vital to establish 
reliable vascular follow-up parameters in 
the injured extremity immediately upon the 
patient's arrival in the surgical ICU. 

Such a parameter can be a Doppler signal, 
the presence of capillary refill, or even a pulse 
oximeter that is applied to a toe in the rele- 
vant extremity. The diagnosis of acute post- 
operative ischemia is based on a change in 
this follow-up parameter and on a difference 
between the perfusion of the two extremities 
that gradually becomes apparent as the 
patient's hypothermia and hypovolemia is 
corrected. 

Because an ischemic extremity is not an 
imminent threat to life, immediate reopera- 
tion is usually not undertaken. Instead, the 
surgeon should assess the patient's overall 
physiology, clinical trajectory, and the feasi- 
bility of a vascular repair, and then formulate 
a plan of action. For example, undertaking a 
vascular reconstruction in the presence of clin- 
ically obvious coagulopathy is futile and often 
leads to further deterioration in the patient's 
already precarious condition. When the 
circumstances are unfavorable for an urgent 
complex arterial reconstruction, acceptable 
options may include watchful waiting to see 
whether the collateral circulation sustains 
limb viability, performing a fasciotomy at the 
bedside, or thrombectomy with reinsertion of 
a temporary shunt until the patient is stable 
enough to undergo a definitive arterial recon- 
struction. Late (>12 hours) occlusion of the 
shunt usually indicates that the patient's coag- 
ulopathy has been corrected and that it is time 
for a definitive repair. 

On rare occasions, an urgent reconstruc- 
tion is required for limb salvage in a patient 




■ FIGURE 8-3 

Bedside surgery in the surgical intensive care 
unit. An urgent vascular reconstruction can 
occasionally be undertaken at the bedside in 
unstable patients with limb-threatening 
ischemia following damage-control surgery. ■ 



whose physiology is so unstable that making 
a trip to the operating room is extremely risky. 
Under these unusual circumstances, simple 
peripheral arterial reconstruction can be 
undertaken at the bedside in the surgical ICU. 
This entails a serious logistic effort to mobi- 
lize operating room technology and create an 
appropriate sterile work environment at the 
bedside (Fig. 8-3). However, in the critically 
ill patient on high-dose inotropic support and 
severe acute respiratory distress syndrome 
on a nonconventional ventilatory mode, this 
may be the safer option. A classic example is 
a bedside crossover femorofemoral bypass in 
a critical patient whose iliac artery has been 
ligated or whose iliac shunt has clotted off. 



PLANNED REOPERATION 



The vascular damage-control sequence ends 
with a planned reoperation in which defini- 
tive reconstruction of the injuries is under- 
taken. The timing of a planned reoperation 
depends on the clinical circumstances, but in 
general the patient should be stable, warm, 
and with normal coagulation. If possible, a 
planned repeated laparotomy should be post- 
poned until the patient has attained a nega- 
tive fluid balance because the presence of 



ch08.qxd 4/16/04 3:31PM Page 174 



174 



II • PRINCIPLES OF OPERATIVE CARE 



swollen edematous bowel and a noncompli- 
ant abdominal wall typically precludes defin- 
itive abdominal closure. If the required 
vascular reconstruction is in the abdomen, it 
is undertaken before depacking of solid 
organs because the removal of packs may result 
in rebleeding and the need for repacking and 
rapid "bailout." 

Peripheral arterial reconstructions are 
performed in the standard fashion. The 
extreme circumstances of patients undergo- 
ing damage-control procedures often pre- 
clude formal angiographic imaging of 
the injured arterial tree before reoperation, 
and the surgeon may, therefore, elect to 
begin the vascular reoperation with an on- 
table angiogram to precisely delineate the 
anatomic conditions before reconstruction 
and to ascertain that there are no missed 
injuries proximally or distally. If a temporary 
intraluminal shunt is in place, the shunt is 
removed and a Fogarty balloon catheter is 
passed proximally and distally. The arterial wall 
is then trimmed so the sites of the ligatures 
or Rummel tourniquets securing the shunt in 
place are not incorporated into the suture line 
because the arterial wall is presumed to be 
compromised. The injury is carefully assessed 
and dEbrided, and a decision is made regard- 
ing the optimal reconstructive technique. In 
the femoral segment, the choice of conduit 
for definitive reconstruction (either vein or 
PTFE) is a matter of controversy, and despite 
concerns about an increased risk of infection 
following reoperation in the same site, there 
are no data to support preference of one 
option over the other. Fasciotomy, if not pre- 
viously performed, should be considered 
under these circumstances, because it extends 
the tolerance of the limb to ischemia and pro- 
tects against the swelling that occurs with 
reperfusion after a prolonged ischemic insult. 

The definitive vascular repair is often per- 
formed in conjunction with operative proce- 
dures in other visceral compartments. If the 
various planned repairs are performed in a 
serial fashion, the operative time is prolonged 
in a patient who is still critically ill. Thus, every 
effort should be made to shorten the opera- 
tive time by planning a multiteam simultane- 
ous operation. With correct planning of the 



operative sequence, the vascular team can 
work in parallel with other teams addressing 
abdominal, thoracic, or head injuries. 



REFERENCES 

Accola KD, Feliciano DV, Mattox KL, et al: Man- 
agement of injuries to the superior mesenteric 
artery. J Trauma 1986;26(4):313-319. 

Aucar JA, Hirshberg A: Damage control for vas- 
cular injuries. Surg Clin North Am 1997;77:853- 
862. 

Bickell WH, Wall MJJr, Pepe PE, et al: Immediate 
versus delayed fluid resuscitation for hypoten- 
sive patients with penetrating torso injuries. N 
EnglJ Med 1994;331:1105-1109. 

Ballard RB, Salomone JP, Rozycki GS, et al: "Damage 
control" in vascular trauma: A new use for 
intravascular shunts. Paper presented at the 
Twenty-Eighth Annual Meeting of the Western 
Trauma Association, Feb 22-28, 1998, Lake 
Louise, Alberta, Canada. 

Burch JM, Ortiz VB, Richardson RJ, et al: Abbre- 
viated laparotomy and planned reoperation for 
critically injured patients. Ann Surg 
1992;215:476-484. 

Coburn M: Damage control for urologic injuries. 
Surg Clin North Am 1997;77:821-834. 

Cosgriff N, Moore EE, Sauaia A, et al: Predicting 
life-threatening coagulopathy in the massively 
transfused trauma patient: Hypothermia and aci- 
doses revisited. J Trauma 1997;42:857-862. 

Cushman JG, Feliciano DV, Renz BM, et al: Iliac 
vessel injury: Operative physiology related to 
outcome. J Trauma 1997;42:1033-1040. 

Dawson DL, Putnam AT, Light JT, et al: Tempo- 
rary arterial shunts to maintain limb perfusion 
after arterial injury: An animal study. J Trauma 
1999;47:64-71. 

DeBakey ME, Simeone FA: Battle injuries of the 
arteries in World War II. Ann Surg 1 946;1 23:534- 
579. 

Eger M, Golcman L, Goldstein A, Hirsch M: The 
use of a temporary shunt in the management of 
arterial vascular injuries. Surg Gynecol Obstet 
197l;132(l):67-70. 

Feliciano DV, Burch JM, Mattox KL, et al: Balloon 
catheter tamponade in cardiovascular wounds. 
Am J Surg 1990;160:583-587. 

Garrison JR, Richardson JD, Hilakos AS, et al: 
Predicting the need to pack early for severe intra- 



ch08.qxd 4/16/04 3:31PM Page 175 



8 • DAMAGE CONTROL FOR VASCULAR TRAUMA 



175 



abdominal hemorrhage. J Trauma 1996;40:923- 
929. 

Gentiello LM, Jurkovich GJ, Stark MS, et al: Is 
hypothermia in the victim of major trauma pro- 
tective or harmful? A randomized, prospective 
study. Ann Surg 1997;226:439-447. 

Gubler KD, Gentiello LM, Hassantash Maier RV: 
The impact of hypothermia on dilutional coag- 
ulopathy. J Trauma 1994;36(6):847-851. 

Hirshberg A, Mattox KL: "Damage control" in 
trauma surgery. Br J Surg 1993;80:1501- 
1502. 

Hirshberg A, Mattox KL: Planned reoperation for 
severe trauma. Ann Surg 1995;222:3-8. 

Hirshberg A, Sheffer N, Barnea O: Computer sim- 
ulation of hypothermia during "damage control" 
laparotomy. World J Surg 1999;23:960-965. 

Hirshberg A, Stein M, Adar R: Reoperation: 
Planned and unplanned. Surg Clin North Am 
1996;77:897-907. 

Hirshberg A, Walden R: Damage control for 
abdominal trauma. Surg Clin North Am 1997; 
77:813-820. 

Hirshberg A, Wall MJJr, Mattox KL: Planned reop- 
eration for trauma: A two year experience with 
124 consecutive patients. J Trauma 1994;37:365- 
369. 

Hirshberg A, Wall MJJr, Ramchandani MK, Mattox 
KL: Reoperation for bleeding in trauma. Arch 
Surg 1993;128:1163-1167. 

Husain AK, Khandeparker JMS, Tendolkar AG, 
e t al: Temporary intravascular shunts for periph- 
eral vascular trauma. J Postgrad Med 1992;38:68- 
69. 

Johansen K Bandyk D, Thiele B, Hanser STJr: Tem- 
porary intraluminal shunts: Resolution of a man- 
agement dilemma in complex vascular injuries. 
J Trauma 1982;22(5) :395-402. 

Jurkovich GF, Greiser WB, Luterman A, Curreri 
PW: Hypothermia in trauma victims: An ominous 
predictor of survival. J Trauma 1987;27:1019- 
1024. 

Majeski JA, Gauto A: Management of peripheral 
arterial vascular injuries with a Javid shunt. Am 
J Surg 1979;138:324-325. 

Mattox KL, Hirshberg A: Vascular trauma. In 
Haimovici's Vascular Surgery: Principles and 
Techniques, 4th ed. Cambridge: Blackwell 
Science, Inc, 1996. 

Michelson AD, MacGregor H, Barnard MR, et al: 
Reversible inhibition of human platelet activa- 



tion by hypothermia in vivoand in vitro. Thromb 
Haemost 1994;71 (5):633-640. 

MorrisJAJr, Eddy VA, Blinman TA, etal: The staged 
celiotomy for trauma. Issues in unpacking and 
reconstruction. Ann Surg 1993;217:576-586. 

Patt A, McCroskey BL, Moore E: Hypothermia- 
induced coagulopathies in trauma. Surg Clin 
North Am 1988;68:775-785. 

Porter JM, Ivatury RR: In search of the optimal end 
points of resuscitation in trauma patients: a 
review. J Trauma 1998;44:908-914. 

PorterJM, Ivatury RR, Nassoura ZE: Extending the 
horizons of "damage control" in unstable trauma 
patients beyond the abdomen and gastroin- 
testinal tract. J Trauma 1997;42(3):559-561. 

Pourmoghadam KK, Fogler RJ, Shaftan GW: Lig- 
ation: An alternative for control of exsanguina- 
tion in major vascular injuries. J Trauma 1997; 
43:126-130. 

Reed RL II, Johnson TD, Hudson JD, Fischer RP: 
The disparity between hypothermic coagulopathy 
and clotting studies. J Trauma 1992;33:465-470. 

Reilly PM, Rotondo MF, Carpenter JP, et al: Tem- 
porary vascular continuity during damage 
control: Intraluminal shunting for proximal 
superior mesenteric artery injury. J Trauma 
1995;39:757-760. 

Rohrer MJ, Natale AM: Effect of hypothermia on 
the coagulation cascade. Crit Care Med 
1992;20:1402-1405. 

Rotondo MF, Schwab CW, Gonigal MD, et al: 
"Damage control": An approach for improved 
survival in exsanguinating penetrating abdom- 
inal injury. J Trauma 1993;35:375-383. 

Rotondo MF, Zonies DH: The damage control 
sequence and underlying logic. Surg Clin North 
Am 1997;77:761-777. 

Scalea TM, Mann R, Austin R, Hirschowitz M: 
Staged procedures for exsanguinating lower 
extremity trauma: An extension of a technique 
[case report] J Trauma 1994;36:291-293. 

Sriussadaporn S, Pak-art R: Temporary intravas- 
cular shunt in complex extremity vascular 
injuries. J Trauma 2002 Jun;52(6):1129-1133. 

Stone HH, Febian TC, Turkleson ML: Wounds of 
the portal venous system. World J Surg 1966;6: 
335-341. 

Stone HH, Strom PR, Mullins RJ: Management of 
the major coagulopathy with onset laparotomy. 
Ann Surg 1983;197:532-535. 

Walker AJ, Mellor SG, Cooper GJ: Experimental 
experience with a temporary intraluminal 
heparin-bondedpolyure thane arterial shunt. Br 
J Surg 1994;81:195-198. 



ch08.qxd 4/16/04 3:31PM Page 176 



176 



II • PRINCIPLES OF OPERATIVE CARE 



Wall MJ Jr, Hirshberg A, Mattox KL: Pulmonary 
tractotomy with selective vascular ligation for pen- 
etrating injuries to the lung. Am J Surg 1994;1 68: 
665-669. 

Wall MJ Jr, Soltero E: Damage control for thoracic 
injuries. Surg Clin North Am 1997;77:863-878. 



Watts DD, Trask A, Soeken K, et al: Hypothermic 
coagulopathy in trauma: Effect of varying levels 
of hypothermia on enzyme speed, platelet 
function, and fibrinolytic activity. J Trauma 
1998:44:846-854. 



chlO.qxd 4/16/04 3:34PM Page 207 




Endovascular Grafts for 
Traumatic Vascular Lesions 



NICHOLAS J. GARGIULO, III 
TAKAO OHKI 
NEAL S. CAYNE 
FRANK J. VEITH 



O INTRODUCTION 

O COIL EMBOLIZATION, INTRAVASCULAR STENTS, AND OTHER 
ENDOVASCULAR TECHNIQUES 

O ENDOVASCULAR GRAFTS FOR ARTERIAL TRAUMA: BACKGROUND 

The Montefiore Experience with Endovascular Grafts for 
Arterial Trauma 

Technique and Devices 

Results 
O SUMMARY 
O ACKNOWLEDGMENTS 



INTRODUCTION 



The advent of endovascular grafting to 
treat abdominal aortic aneurysms (AAAs) 
by Parodi, Palmaz, and Barone (1991) has 
expanded to include arterial occlusive disease, 
occluded grafts, peripheral aneurysms, and 
traumatic arterial lesions (Marin and col- 
leagues, 1995a, 1995b; Parodi, 1995) . The past 



decade has marked a new enthusiasm for the 
endovascular repair of AAAs. However, the 
value and long-term outcome have yet to be 
proven. Endoleaks, arterial injury, and late 
graft deterioration continue to complicate 
endovascular graft repair of AAAs. Since stan- 
dard open aneurysm repair remains a safe and 
reliable procedure with good long-term out- 
comes, the role of endovascular graft repair 
in low-risk patients remains to be determined. 

207 



chlO.qxd 4/16/04 3:34PM Page 208 



208 



III • DIAGNOSTIC AND INTERVENTIONAL RADIOLOGY 



On the other hand, the role of endovascu- 
lar grafts for traumatic arterial injuries appears 
to be more easily defensible, especially when 
large central vessels are involved. Vascular 
trauma within the thorax or abdomen com- 
plicates the surgical approach to a vascular 
injury. Distorted anatomy due to a large 
hematoma or false aneurysm and venous 
hypertension secondary to an arteriovenous 
fistula are just a few of the problems encoun- 
tered during an open repair of injured vessels. 
Endovascular repair is more appealing 
because it can be performed from a remote 
site and does not require direct surgical expo- 
sure of the injury site, thus reducing the mor- 
bidity and mortality rates that accompany 
open repair. Furthermore, endovascular 
repair is most beneficial for those patients who 
are critically ill from other injuries or medical 
comorbidities. 

The main endovascular techniques used 
in the treatment of vascular trauma include 
coil embolization, intravascular stents, and 
endovascular stented grafts. Traumatic vas- 
cular lesions usually have normal, healthy, 
proximal, and distal arterial segments or graft 
fixation zones for endovascular graft deploy- 
ment. This is in contrast to the complex necks 
and iliac tortuosity of AAAs. As a result, high 
technical success rates and low rates of endo- 
graft migration or leakage have been reported 
after endovascular grafting for traumatic vas- 
cular lesions. 

This chapter describes endovascular tech- 
niques that may prove helpful in vascular 
trauma and reviews our experience with 
endovascular grafts for traumatic lesions at 
Montefiore Medical Center in New York. We 
also discuss the role of endovascular grafts for 
thoracic aortic vascular injuries. 



COIL EMBOLIZATION, 
INTRAVASCULAR STENTS, AND 
OTHER ENDOVASCULAR 
TECHNIQUES 

Embolization coils have been used to treat 
relatively small traumatic arteriovenous fistu- 
las and pseudoaneurysms involving nones- 
sential vessels, such as a lumbar artery, the 



internal mammary artery, or the branches of 
the hypogastric or deep femoral arteries 
(Rosch, Dotter, and Brown, 1972; Panetta and 
colleagues, 1985) . Long-term follow-up results 
of the use of these coil-treated lesions have 
proven favorable. Placement of intravascular 
stents is useful for the repair of intimal flaps. 
Because of their porous nature, however, 
uncovered stents are not indicated for treat- 
ing arteriovenous fistulas or pseudoaneurysms 
of large vessels. Although coils and stents 
have proved to be effective in selected cases, 
most patients with vascular trauma are not 
amenable to such therapy. 

A novel method for obtaining intraluminal 
balloon control of arteries in difficult cir- 
cumstances has been previously described by 
our group (Veith, Sanchez, and Ohki, 1998). 
This technique is particularly useful when 
bleeding, scarring, or infection makes dis- 
section of proximal arteries difficult or dan- 
gerous. Through an arterial puncture distal 
to the site where proximal control is required, 
an 18-gauge needle is inserted into a normal 
artery. A guidewire is inserted through the 
needle. Over the guidewire, a 6- or 7-Fr hemo- 
static sheath and dilator is inserted. Under 
fluoroscopic guidance, a standard balloon 
catheter is passed through the hemostatic 
sheath. Radiopaque contrast is then injected 
to confirm optimal placement of the balloon 
catheter for proximal occlusion within the 
arterial tree. The sheath may then be 
retracted, and the balloon inflated until arte- 
rial inflow is occluded. Alternatively, double- 
lumen balloon catheters may be passed over 
a guidewire and angiographic techniques can 
be used to facilitate proximal balloon control. 



ENDOVASCULAR GRAFTS FOR 
ARTERIAL TRAUMA: 
BACKGROUND 

Endovascular grafts have significantly 
extended the potential of endovascular 
therapy for vascular trauma. The concept of 
endovascular grafting for traumatic arterial 
lesions was initially proposed by Dotter (1969). 
Volodos and colleagues (1991) were the first 
to clinically apply this technology by placing 



chlO.qxd 4/16/04 3:34PM Page 209 



10 • ENDOVASCULAR GRAFTS FOR TRAUMATIC VASCULAR LESIONS 



209 




II 



III! 



II 



III 



31 



■ FIGURE 10-1 

Endovascular stented graft. A 
Palmaz stent is sewn to an 
expanded PTFE graft. (From 
Ohki T, Veith FJ, Marin ML, et 
al: Endovascular approaches 
for traumatic arterial lesions. 
Semin Vase Surg 1997;10:272- 
285.) ■ 



a Dacron graft and a self-expanding stent to 
treat a thoracic aortic pseudoaneurysm in 
1986. 

Endovascular grafts have been used to treat 
almost every kind of injury at various locations 
in the body. Some patients treated with 
endovascular grafts have been hemodynami- 
cally stable. However, some of these grafts have 
also been used to treat life-threatening acute 
hemorrhage (Becker and colleagues, 1991; 
Patel and colleagues, 1996). The types of 
devices that have been reported are pre- 
dominantly a combination of a Palmaz stent 
and an expanded PTFE (ePTFE) graft (Fig. 
10-1) (Marin and colleagues, 1993, 1994; 
Becker and colleagues, 1995; Terry and col- 
leagues, 1995; Zajiko and colleagues, 1995; 
Gomez-Jorge and colleagues, 1996; Criado 
and colleagues, 1997; Dorros and Joseph, 
1997) . The use of a vein graft in combination 



with a Palmaz stent has also been reported, 
since the traumatized field is often contami- 
nated. More recently, industry made devices 
such as the Corvita endovascular graft, the 
AneuRx graft and the Wallstent graft have 
become commercially available in the United 
States. AneuRx grafts have been approved by 
the Food and Drug Administration for the 
treatment of AAAs, and the Wallstent for 
obstructive biliary lesions. Components of the 
AneuRx include the bifurcated graft and the 
proximal and distal extension cuffs. Though 
not approved for the use in traumatic lesions, 
these grafts have been used in the "off-label" 
fashion. 

The lesion, location, characteristics, site of 
arterial access, technical success rate, and the 
outcome of endovascular grafts in the treat- 
ment of vascular trauma are summarized in 
Table 10-1. These results have been encour- 



TABLE 10-1 

ENDOVASCULAR GRAFTS FOR ARTERIAL TRAUMA 



Type 



Combination of Palmaz 
Stent and Various Grafts 



Cragg Endopro 



Corvita Graft 



Stent Palmaz stent Nitinol Self-expanding braided stent 

Graft material PTFE Dacron Vein Silicone Ultrathin woven polyester Polycarbonate urethane 

fabric 

Arterial access 1 or 2 2 2 1 or 2 2 2 

1, open arteriotomy; 2, percutaneous. 

From Ohki T, Marin ML, Veith FJ: Use of endovascular grafts to treat non-aneurysmal arterial disease. Ann Vase Surg 

1997:11:200-205. 



chlO.qxd 4/16/04 3:34PM Page 210 



210 



III • DIAGNOSTIC AND INTERVENTIONAL RADIOLOGY 



o 

r 
< 



>- 

CL 

D 



o 

Z 

o 



CO 

LU 



CO 

o 

h- 
co 

DC 
<M LU 

11 

< X 
H O 



o 


£■ 


o 


+* 


_l 


< 


>- 


c 


ou 


> 


III 


<u 


2 


o 

J2 


o 
n 


3 

(0 


H 


cT 


3 


re 


O 


■R 


Q 


< 



E 

3 



O 



to 






O 








N 






CO 


CO 






"CZ 








ZS 






B 






>, 


o 













E 


CD 

c 

.2 






JZ 






o 


















CO 

o 






o 

CD 


3 

CJ 






o^ 






tc 


CD 






o 






CO 


Q. 






CD 






"cO 


"cO 






J£> 




o 


b 

E 


b 

E 


p 

G 

O 




3 


O 


o 





CD 


O 


0? 


CD 


CO 


"Sf 


LL 


LL 




O 



>. o 

-£ £ ^? 

13 -* o 

8? " r^ o 

o ld t- t- 



£■ 


y 










o 















o 


"5 










CO 














r 
< 


N 








G 


CO 














CD 


0^ 






"3- 

CO 


C-0 














o 


CD 


CO 








Z3 














re 


jz 


CO 






>, 


o 
















"5 


co 






E 

o 



c 














i_ 


o 








JO 














o 

re 

r 
o 

< 


cr- 
CO 

CO 

"cO 




JZ 

o 

G 






o 

CD 

CO 
CO 


3 

CJ 



Q. 

"cO 










o 






CJ 


% 


c>- 




b 

E 


b 

E 


o 
O 


*-9 


^9 


CO 


E 

CO 


G 

o 




L" 5 


3 


r^- 


CO 








O 






d 


o 




i- CO 


JJ. 


co 


1^ 


LL 


LL 


T— 






vf 


T~ 


T— 






C3J c\J 
CO CM 



o 
o 







CO 




■ : 


:n 


_2> 


cl) 


°3 ,2 


J_ 


i- CO 


U 



t CO 

CO CL CL ^ 

a js ffl J; 

JZ JZ o " 

CJ CJ £ ^p 

n* /rt f~ O*^ 

■*■ 



■i—^l i- *- uj xi o- 
CD ^- CD QQ LL O) o 



CO 

3? n o L^ 

CO CO t- CO 



.2 co 

IS ° 

re .4_ 

8 ° 





0) 


o 






c 
















o 








"a 

ZJ 
CD 


> 
o 




>> 


CO 




rn 


ZJ 


CL 


CO 


UJ 
CD 


( 


o 


t ) 














CD 


(h 




u 


O 


( l 






sz 


L_ 


m 




CD 


CU 












m 









Q_ CL < 



.o 

"5 

CJ 


b 


n 


CD 

c 

_0 


3 
o 

o 


"5 

CL 


CD 


a) 

CO 


o 

D 
CO 




c_ 




r 


r 


CO 




j_ 


E 
o 
O 


i> 


2 


CO 


2 


CO 


5 


b 

CL 


CD 
O 




o 

1 






chlO.qxd 4/16/04 3:34PM Page 211 



10 • ENDOVASCULAR GRAFTS FOR TRAUMATIC VASCULAR LESIONS 211 




■ FIGURE 10-2 

A, Schematic drawing of an endovascular stented graft or covered stent. A segment of expanded 
PTFE is attached to a Palmaz stent (St) using two 5-0 Prolene U stitches (S). B, Schematic drawing 
of a double-stent endovascular stented graft. The proximal stent (St) is sutured to the graft as 
described in (A). The distal end of the graft is marked with gold markers (G) for visualization under 
the fluoroscope. C, The stent graft (SG) is mounted on an angioplasty balloon (B) and placed into a 
sheath (C) before insertion. Note the presence of a dilator tip (D) at the end of the balloon catheter, 
which provides a smooth taper within the catheter. W, guidewire. (From Ohki T, Veith FJ, Marin ML, 
et al: Endovascular approaches for traumatic arterial lesions. Semin Vase Surg 1997;10:272-285.) ■ 



aging with a high technical success rate (94% 
to 100%) and a complication rate of 0% to 
7% (Table 10-2), especially when we consider 
the difficulties that could be encountered in 
treating these lesions by a direct surgical repair. 
In addition, the minimal invasiveness and 
the potential for cost-effectiveness of such 
endovascular techniques are apparent from 
the short length of stay (3.3 to 3.5 days) (Table 
10-2) . Most endovascular grafts are deployed 
in nonatherosclerotic central vessels of a 
large caliber and have excellent durability. 
Mean follow-up at 16 months revealed excel- 
lent mid-term patency rates ranging from 85 % 
to 100% depending on where deployment 
occurred. 



The Montefiore Experience 
with Endovascular Grafts for 
Arterial Trauma 

TECHNIQUE AND DEVICES 

At Montefiore Medical Center, we have 



mainly used the Palmaz stent (Cordis 
[Johnson &Johnson Company, Warren, NJ] ) 
in combination with a thin-walled ePTFE graft 
(Fig. 10-1) covering to perform arterial repairs 
of pseudoaneurysms and arteriovenous fistu- 
las (Marin and colleagues, 1993, 1994, 1995a) . 
Depending on the length of the lesion, either 
a single stent device or a doubly stented device 
was used. The stents varied between 2 and 
3 cm in length (Palmaz P-204, 294, 308) and 
were fixed inside 6 mm Gore-Tex grafts (W.L. 
Gore and Associates, Flagstaff, Ariz) by two U 
stitches. The stented graft was then mounted 
on a balloon angioplasty catheter, which had 
a tapered dilator tip firmly attached to its end 
(Fig. 10-2). The entire device was contained 
within a 10- to 12-Fr delivery system for over- 
the-wire insertion either percutaneously or 
through an open arteriotomy. 

Alternative devices included the Corvita 
stent graft (Corvita Corporation, Miami, Fla) , 
and the Wallgraft, both of which are fabricated 
from a self-expanding stent or braided wire. 
The Corvita stent graft is covered with poly- 
carbonate elastomer fibers, and the Wallgraft 
is covered with Dacron. The Corvita stent graft 



chlO.qxd 4/16/04 3:34PM Page 212 



212 



III • DIAGNOSTIC AND INTERVENTIONAL RADIOLOGY 



may be cut to the desired length in the 
operating room using a wire-cutting scissors 
and then loaded into a specially designed 
delivery sheath. This sheath has a central 
"pusher" catheter, which is used for main- 
taining the graft in position while the outer 
sheath is being retrieved. TheWallgraft comes 
in various diameters up to 14 mm and lengths 
up to 7 cm. 

RESULTS 

Each procedure was performed in the 
operating room under fluoroscopic (OEC 
9800, OEC/GE, Salt Lake City, Utah; Philips, 
BV 212, Netherlands) guidance. Most cases 
were performed under local or epidural anes- 
thesia with two cases requiring general anes- 
thesia. A total of 17 stented grafts were used 
to treat 17 patients with traumatic arterial 
lesions (Table 10-3). The etiology for these 
lesions is described in Table 10-3, with the 
majority comprising gunshot wounds (Figs. 
10-3 and 10-4) and iatrogenic injuries (Figs. 
10-5 and 10-6). 

All injuries except for one were associated 
with an adjacent pseudoaneurysm (Fig. 10-5) . 
Five patients had an arteriovenous fistula (Fig. 
10-6) , and eight patients had other associated 
injuries (Table 10-3). 

Procedural complications were limited 
to one distal embolus, which was treated 
with suction embolectomy, and one wound 
hematoma, which resolved without further 
intervention. Graftpatencywas 100% with no 
early or late graft occlusions (mean follow-up 
was 30 months [range, 6 to 46 months]). 

One patient with a left axillary subclavian 
stent graft developed compression of the stent 
at 12 months and was treated with balloon 
angioplasty. This recurred 3 months later but 
did not require any intervention. At 3-year 
follow-up, the graft was patent. A second 
patient developed stenosis at either end of his 
stent graft and was successfully treated with 
additional balloon dilation and Palmaz stent 
placement (Fig. 10-4). A third patient with 
an axillary pseudoaneurysm repaired with a 
stent graft required a vein patch to close a small 
brachial artery insertion site. 

Immediate repair of blunt thoracic aortic 



injuries to prevent rupture of the contained 
hematoma as previously described may no 
longer be necessary based on studies by Camp 
and Shackford (1997) and Maggisano and col- 
leagues (1995). These authors suggest that 
delayed repair of hemodynamically stable tho- 
racic aortic injuries reduces morbidity and 
mortality. Delayed repair allows the trauma 
team to surgically optimize the multi-injured 
patient before a major surgical insult. These 
studies have significantly influenced the role 
of endovascular grafting in the treatment 
of thoracic aortic injuries. Myriad unique 
endovascular techniques and grafts have been 
employed and reported to treat these injuries 
(White and colleagues, 1997; Lobato and col- 
leagues, 2000; Fontaine and colleagues, 2001; 
Ruchat and colleagues, 2001). 

Endovascular grafting has been employed 
in blunt and penetrating injuries of the 
abdominal aorta (White and colleagues, 1997; 
Fontaine and colleagues, 2001 ) . These include 
successful exclusion of a posterior aortic 
pseudoaneurysm between the superior 
mesenteric artery and the right renal artery 
following a gunshot wound. A Cooley VeriSoft 
vascular graft attached to the outer surface of 
an extra-large Palmaz stent was successfully 
deployed across the aortic pseudoaneurysm 
3 weeks after the initial injury. 

In addition to these homemade devices, 
industry-made devices recently became avail- 
able. These include the Talent thoracic 
endovascular graft (World Medical Corpora- 
tion, Medtronic) , the Thoracic Excluder graft 
(W.L. Gore, Flagstaff, Ariz) and the AneuRx 
graft (Fontaine and colleagues, 2001; Ruchat 
and colleagues, 2001). Neither of these has 
been approved by the FDA for commercial- 
ization in the United States; however, some 
investigators have successfully used these 
grafts to treat life-threatening thoracic aortic 
injuries on a compassionate basis. These 
industry-made devices, especially the Excluder 
graft, are much more flexible and have a lower 
insertion profile. 

Delayed repair has been reported from 1 
week to a mean of 5.4 months after the initial 
accident. There has been no evidence of 
endoleak or rupture at approximately 1-year 
follow-up. Furthermore, although the number 

Text continued on p. 218 



chlO.qxd 4/16/04 3:34PM Page 213 



10 • ENDOVASCULAR GRAFTS FOR TRAUMATIC VASCULAR LESIONS 



213 



LU 

o 

z 

LU 

x 

LU 
Q. 
X 
LU 

LU 
X 
O 
LL 
LU 



CO 



CO 
LU 



< 
LX 
LU 
H 
LX 
< 
O 



Z> 
< 
X 
H 
X 
O 

LL 
CO 





X 




O 




X 




< 




_i 




D 


CO 


O 


1 

o 


CO 

< 




■> 


UJ 


O 


CO 


Q 


< 


Z 


H 


LU 





<a 




c 




o 




4- 




a 




o 




Q. 




E 




o 




o 




>. 




o ^- 








(0 ^ 




a. 


«S 




4-1 


>•£' 


'5. 


(0 > 


(A 


4- re 


O 


W Q 


X 






<A 




<0 




0) 




O 




U 




< 




JC 


** c 


4-» ^^. 


= re 


O) £ 


<u >- 


si 


w« 


o ,_ 




4-1 1- 


ll 

■"I 


>. re 

? °- 
.2. a> 








■D 




- a 




re <u 

o .2. 




V) c 




<fl — 




< 




re 




M 




<U 




£ 




4-1 




V) 




01 




c 




< 




LL 




> 




< 




< 




Q. 




^^ 




<fl ■D 




sr <u 




a> > 




SI 


E 




<o 




'E 




«S 




o 
a) 


- 1 

o .5, 


S 


c 


■» 


a> rr 


W < C 



-d 3 





Q_ 

~ E 
c o 
a) o 

co 



a> t- *- 



n 


t- t- O r- 


h- CO 


CM 


LO 


C\J 


Cvl (M CO m 


co co 


-t 


"* 



s 

CO 



CO 


O 

CD 


F 


O 

C 

CO 


E 


F 


F 


>> 
F 


CO 


CO 


CO 






F 










F 


To 


CO 


o 




o 


o 


o 


"CO 


"m 


To 


CO 


>i 


CJ 


CO 


>i 




>. 


u 




Z3 


o 


Z5 




o 


o 


o 










F 


CJ 




F 


CO 


F 


CD 


o 

CO 


CD 


.2 


CJ 















o 

CO 


a 

CO 


o 

CO 


O 

CO 


o 

n 





o 

CO 


o 


JZ 

o 


o 





o 




CO 


0_ 




CO 


CO 


CO 


JO 


n 


o 


o 


^ 


CO 


n 


i_ 




^ 


CO 


Iz 


< 

LL 


< 

LL 
CO 


if 


< 

LL 


rf 


f 


if 


jz 


jz 


jz 


jz 




if 


jz 




X) 




if 




O 


n 


O 


O 


o 


o 












n 










CD 


LL 


LL 


_1 


_i 


LL 


_i 


_1 


_l 


LL 


LL 


LL 


LL 




_i 


LL 




_l 




LL 



CO CO CO CO 



>_ E 
>■ CO 

CO i- 


o 
E 


Q 





CO 


^ i_ JZ JZ 


o j£ 
E 3 


j__ 


JZ 


JZ 


CJ 


^ -c 00 •* 


JZ 


CM 


CO 


co 


co co ^r cm 


^r co 


■* 


T- 


CJ 



_0 



o 

LO 



CD CO C 

£ -c c 2 

P ° n B 

E E g-o 

Z3 u CO 



CO CO CO 

o o o 
o o o 



O 
Z X X X 



CO CO CO CO 

cj cj o o 
o o o o 





en 



o 

CO 





(J 



o 
z 


2 


o 

X 


CO 


to 

2 


o 
z 


o 
z 


o 
z 


o 
z 


ojo 


o 
z 


CO 


CO 


CO 


CO 


CO 


CO 


CO 


CO 


CO CO CO 



CO CO 



< 

CO 


< 


> 


< 
LL 


< 


< 


< 




CO 




CO 




< 

CO 


< 

CO 


< 


< 


< 


< 

CO 


< 


o 


O 


CO 


O 


o 


o 




o 




o 




< 


< 


CO 


CO 


O 


< 


CC 


CL 


_l 


_l 


CH 


CL 


_1 




< 




< 




cc 


cc 


CC 


en 


_i 


cc 


C 








CO 

E 


c 




















c 


c 




o 








o 




















o 


o 












zs 














C 

a 














CO 








CO 


CO 


















CO 


CO 




N 








.C3 


N 


^_, 


c 


To 


c 










_N 


N 


















o 




o 




a. 

Zi 

CO 


















CO 

CJ 


CO 

E 


„ 


CO 

o 






CO 


Q. 


CO 
CD 


Q. 


CJ 


„ 


„ 


„ 










„ 


JZ 


CD 


JB 


'en jz 


o 


Zi 




Z5 




M 


ss 


_gj 


JZ 


JZ 


M 






Z5 
















TO 














CO 


D 


CO 


Z) 


zs 


CO 


CO 


CO 


O 


<S) 


o 


Zi 


ZJ 


D 


CO 


CO 


Zi 


O 


CO 




LQ 


CO 


O 


= 


CO 


< 


TO" 


< 




CD 


LO 


CO 


O 


O 


CO 


o 

CO 






OJ 


CO 
CO 


05 


CO 




CO 




CO 




CO 


CM 


CO 


CO 


CO 


UO 


C\J 




OJ 


^3- 


CO 




CO 




h- 




TC 


OJ 


r; 


N- 


CO 



< 

CO 

< CO 

_l cc 



<c < > 

LL LL 
CO CO 



j2 
3 

CO 



J? 
3 
CO 







cc 


m 














E 


E 



















CO < 



CO n 



< . 

O < 



c 


- 


- 


b 


F 


■■'. 


' i 






j_ 


^ 






~- 


<f 


< 


ii 


< ) 


U 


a: 



o JO 











< 


< 










CO 


CO 










3 


< 










co" 
'co 


E 










O 


CO 










n 












E 

o 


Z3 
0.) 
C 










JZ 


CO 












o 






>. 




CO 


T5 






E 

o 
o 


CO 


D 


-.:■ 


o 
"a 




E 

_0 


O 
C 

a 


1. 

CO 
Q. 


o 




o 


JD 


> 




E 

o 

JZ 

o 

ZJ 

-a 




r- 


o 


Q. 


< 




E 
p 
JZ 

c 
g 


Q. 

"ca 

o 

E 


O 

o 

"D 

K 

> 

Q 


Q_ 
C 

'0 
> 


<D 




o 






o 


O 

o 




Z5 
CO 

o 




a 


CO 
D 

to 


. 7 


"co 










"co 


O 
Q. 

CO 


2 

en 


a 

JZ 

CO 

o 


E 


CO 
D 
O 
C 





C 
O 

E 

CM 


c 

CO 


JZ 

1 



N 


O 
> 

g 


a 

Z3 
CO 


W 


"a 

CD 




o 


_03 


"D 


> 


ai 


a 

CO 


cc 


?>" 


O 


O 


o 


O 


LL 


u_ 


b 


O 


h= 


X 


> 

< 


00 

_l 



chlO.qxd 4/16/04 3:34 PM Page 214 



214 



III • DIAGNOSTIC AND INTERVENTIONAL RADIOLOGY 









UK\I III \1 





■ FIGURE 10-3 

Angiographic images of a 
patient who sustained a 
gunshot wound to the right 
chest. A, An angiogram 
performed via a femoral artery 
puncture shows occlusion of 
the right subclavian artery and 
active bleeding. B, An 
occlusion balloon was placed 
to achieve hemostasis. C, 
Following hemostasis, the 
patient was taken to the 
operating room. A guidewire 
was successfully passed 
across the injured artery and 
was repaired by the insertion of 
a stented graft of 
polytetrafluoroethylene 
(expanded PTFE and Palmaz 
stent). (From Patel AV, Marin 
ML, Veith FJ, et al: 
Endovascular graft repair of 
penetrating subclavian artery 
injuries. J Endovasc Surg 
1996;3:382-388.) ■ 





■^ ^^H 






OCCLUSION 
BALLOON 












chlO.qxd 4/16/04 3:34PM Page 215 



10 • ENDOVASCULAR GRAFTS FOR TRAUMATIC VASCULAR LESIONS 



215 




■ FIGURE 10-4 

The patient is a 19-year-old man status post a gunshot wound to the chest that traversed the 
mediastinum from right to left, injuring his esophagus, trachea, and left subclavian artery. A, The 
initial angiogram shows occlusion of the left vertebral artery and a small pseudoaneurysm (p) at 
that site. He was transferred to our institution for endovascular treatment following placement of a 
covered esophageal stent to repair his tracheoesophageal fistula. B, A Corvita endoluminal graft 
was placed across the lesion and there was excellent flow through it. Continued 



chlO.qxd 4/16/04 3:34PM Page 216 



216 III • DIAGNOSTIC AND INTERVENTIONAL RADIOLOGY 




■ FIGURE 10-4 

cont'd C, A plain x-ray film demonstrates the esophageal stent and the Corvita graft in the left 
subclavian artery. D, At 4 months after graft insertion, his left radial pulse was diminished, although 
he remained asymptomatic. An angiogram was taken that revealed intimal hyperplasia throughout 
the graft, more prominent at both ends of the graft (arrows). These lesions were angioplastied and a 
Palmaz stent was placed. After the procedure, the patient had a strong radial pulse. (From Ohki T, 
Veith FJ, Kraas C, et al: Endovascular therapy for upper extremity injury. Semin Vase Surg 
1998;11:106-115.) ■ 



chlO.qxd 4/16/04 3:34PM Page 217 



10 • ENDOVASCULAR GRAFTS FOR TRAUMATIC VASCULAR LESIONS 217 




■ FIGURE 10-5 

A, This arteriogram shows a large pseudoaneurysm of the subclavian artery (arrow) just distal to the 
right vertebral artery that occurred after an attempted subclavian vein catheter insertion. B, Following 
stented graft (expanded PTFE and Palmaz stent) placement through the right brachial artery, the 
pseudoaneurysm was excluded. Vertebral artery flow was maintained (V). (From Marin ML, Veith FJ, 
Panetta TF, et al: Transluminal^ placed endovascular stented graft repair for arterial trauma. J Vase 
Surg 1994;20:466-473.) ■ 



chlO.qxd 4/16/04 3:34PM Page 218 



218 



III • DIAGNOSTIC AND INTERVENTIONAL RADIOLOGY 




■ FIGURE 10-6 

A, Preoperative angiogram of an iatrogenic arteriovenous fistula (AVF) following lumbar disk 
surgery. The patient presented with severe swelling of the left lower extremity. The left common iliac 
vein (C) is dilated secondary to the fistula. B, Completion angiogram. A PTFE graft was fixed 
proximally (p) and distally (d) with a Palmaz stent to exclude the fistula from the arterial circulation. 
Coil embolization of the internal iliac artery was performed before stent-graft insertion. (From Ohki T, 
Veith FJ: Endovascular techniques in the treatment of penetrating arterial trauma. In Yao JST, 
Pearce WH [eds]. Practical Vascular Surgery, 1st ed. Stamford, Conn, Appleton & Lange, 1999, 
pp 409-423.) ■ 



of reported cases is small, there has been no 
mention of postoperative paraplegia. These 
limited experiences suggest that endovascu- 
lar graft techniques will be better than more 
traditional open techniques of thoracic aortic 
repair requiring either bypass or the clamp- 
and-sew technique, which report postopera- 
tive paraplegia rates of 4.5% to 16.4%, 
respectively (Fabian and colleagues, 1997). 



SUMMARY 



Endovascular grafting for traumatic arterial 
lesions has become an additional tool for 
the vascular surgeon. Complex open surgical 
repair of thoracic or intra-abdominal vascu- 



lar injuries may be approached with minimally 
invasive endovascular techniques. Large 
hematomas, false aneurysms, and arteriove- 
nous fistulas that often obscure the open 
surgical field have minimal impact on 
endovascular repair. This can be performed 
by accessing vascular lesions from remote sites 
so that embolization coils, stents, or endovas- 
cular grafts may be deployed. 

Few vascular injuries may be amenable 
to coil embolization or stent placement. 
However, endovascular grafts have greatly 
expanded endovascular therapy for vascular 
trauma. These grafts have been used to treat 
myriad vascular injuries at various locations 
in the body. 

Endovascular grafts have a low morbidity 
rate, high success rate, reduced anesthetic 



chlO.qxd 4/16/04 3:34PM Page 219 



10 • ENDOVASCULAR GRAFTS FOR TRAUMATIC VASCULAR LESIONS 



219 



requirements, and a minimal dissection 
requirement in the traumatized field. These 
qualities are particularly advantageous for 
patients with central arteriovenous fistulas or 
false aneurysms, especially those critically ill 
from other coexisting injuries or medical 
comorbidities. 

Endovascular grafts and techniques will con- 
tinue to evolve and complement traditional 
open techniques in vascular trauma. Future 
development of smaller delivery systems, 
better endografts, operating rooms equipped 
with improved angiographic imaging systems, 
and a supply of endovascular equipment will 
increasingly help vascular surgeons of the 
future to manage patients with vascular trauma 
better. 



ACKNOWLEDGMENTS 

This work was supported by grants from the 
U.S. Public Health Service (HL 02990), the 
Manning Foundation, the Anna S. Brown 
Trust, the New York Institute for Vascular 
Studies, and the William J. von Liebig 
Foundation. 

REFERENCES 

Becker GJ, Benenati JF, Zemel G, et al: Percuta- 
neous placement of a balloon-expandable intra- 
luminal graft for life-threatening subclavian 
arterial hemorrhage. JVTR 1991;2:225-229. 

Becker GJ, Katzen BT, Benenati JF, et al: Endografts 
for the treatmen t of aneurysm and traumatic vas- 
cular lesions: MVI experience. J Endovasc Surg 
1995;2:380-382. 

Brandt MM, Kazanjian S, Wahl WL: The utility of 
endovascular stents in the treatment of blunt arte- 
rial injuries. J Trauma 2001;51(5):901-905. 

Camp PC, Shackford SR: Outcome after blunt trau- 
matic thoracic aortic laceration: Identification 
of a high-risk cohort. Western Trauma Associa- 
tion Multicenter Study Group. J Trauma 1997; 
43:413-422. 

Criado E, Marston WA, Ligush J, et al: Endovas- 
cular repair of peripheral aneurysms, pseudo- 
aneurysms, and arteriovenous fistulas. Ann Vase 
Surg 1997;11:256-263. 

Dorros G, Joseph G: Closure of a popliteal arteri- 
ovenous fistula using an autologous vein-covered 



Palmaz stent. J Endovasc Surg 1995;2:177- 
181. 

Dotter CT: Transluminally-placed coilspring endar- 
terial tube grafts: Long-term patency in canine 
popliteal artery. Invest Radiol 1969;4:329-332. 

Fabian TC, Richardson JD, Croce MA, et al: 
Prospective study of blunt aortic injury: Multi- 
center trial of the American Association for the 
Surgery of Trauma. J Trauma 1997;42:374-380. 

Fontaine AB, Nicholls SC, Borsa JJ, et al: Seat belt 
aorta: Endovascular management with a stent- 
graft. J Endovasc Ther 2001;8:83-86. 

Gomez-Jorge JT, Guerra JJ, Scagnelli T, et al: 
Endovascular management of a traumatic sub- 
clavian arteriovenous fistula. JVIR 1996;7:599- 
602. 

Lobato AC, Quick RC, Phillips B, et al: Immediate 
endovascular repair for descending thoracic 
aortic transection secondary to blunt trauma. J 
Endovasc Ther 2000;7:16-20. 

Maggisano R, Nathens A, Alexandrova NA, et al: 
Traumatic rupture of the thoracic aorta: Should 
one always operate immediately? Ann Vase Surg 
1995;9:44-52. 

Marin ML, Veith FJ, Cynamon J, et al: Initial expe- 
rience with transluminally placed endovascular 
grafts for the treatment of complex vascular 
lesions. Ann Surg 1995a;222:449-469. 

Marin ML, Veith FJ, Lyon RT, et al: Transfemoral 
endovascular repair of iliac artery aneurysms. 
Am J Surg 1995b;170:l79-182. 

Marin ML, Veith FJ, PanettaTF, etal: Percutaneous 
transfemoral insertion of a stented graft to repair 
a traumatic femoral arteriovenous fistula. J Vase 
Surg 1993;18:229-302. 

Marin ML, Veith FJ, Panetta TF, et al: Translumi- 
nally placed endovascular stented graft repair 
for arterial trauma. J Vase Surg 1994;20:466-473. 

Marty-Ane CH, Berthet JP, Branchereau P, et al: 
Endovascular repair for acute traumatic rupture 
of the thoracic aorta. Ann Thorac Surg 2003 
Jun;75(6):1803-1807. 

Orend KH, Pamler R, Kapfer X, et al: Endovascu- 
lar repair of traumatic descending aortic tran- 
section. JEndovasc Ther 2002 Oct;9 (5) :573-578. 

Panetta TF, Sclafani SJA, Goldstein AS, et al: Per- 
cutaneous transcatheter embolization for arte- 
rial trauma. J Vase Surg 1985;2:54-64. 

ParodiJC: Endovascular repair of abdominal aortic 
aneurysms and other arterial lesions. J Vase Surg 
1995;21:549-557. 

ParodiJC, Palmaz JC, Barone HD: Transfemoral 
intraluminal graft implantation for abdominal 
aortic aneurysms. Ann Vase Surg 1991 ;5:491-499. 



chlO.qxd 4/16/04 3:34PM Page 220 



220 



III • DIAGNOSTIC AND INTERVENTIONAL RADIOLOGY 



Patel AV, Marin ML, Veith FJ, et al: Endovascular 
graft repair of penetrating subclavian artery 
injuries. J Endovasc Surg 1996;3:382-388. 

Rosch J, Dotter CT, Brown MJ: Selective arterial 
embolization. Anew method for control of acute 
gastrointestinal bleeding. Radiology 1972;102: 
303-306. 

Ruchat P, Capasso P, Chollet-Rivier M, et al: 
Endovascular treatment of aortic rupture by 
bluntchest trauma.J Cardiovasc Surg 2001 ;42:77- 
81. 

Terry PJ, Houser EE, Rivera FJ, et al: Percutaneous 
aortic stent placement for life-threatening aortic 
rupture due to metastatic germ cell tumor. J Urol 
1995;153:1631-1634. 

Thompson CS, Rodriguez JA, Ramaiah VG, et al: 
Acute traumatic rupture of the thoracic aorta 
treated with endoluminal stent grafts. J Trauma 
2002 Jun;52(6) :1 1 73-1 1 77. 



Veith FJ, Sanchez LA, Ohki T: Technique for obtain- 
ing proximal intraluminal control when arter- 
ies are inaccessible or unclampable because of 
disease or calcification. J Vase Surg 1998;27:582- 
586. 

Volodos NL, Karpovich IP, Troyan VI, et al: Clini- 
cal experience of the use of self-fixing synthetic 
prostheses for remote endoprosthetics of the tho- 
racic and the abdominal aorta and iliac arteries 
through the femoral artery and as intraopera- 
tive endoprosthesis for aorta reconstruction . Vasa 
1991;33(suppl):93-95. 

White R, Donayre C, Walot I, e t al: Endograft repair 
of an aortic pseudoaneurysm following gunshot 
wound injury: Impact of imaging on diagnosis 
and planning of intervention. J Endovasc Ther 
1997;4:344-351. 

Zajiko AB, Little AF, Steed DL, et al: Endovascular 
stent-graft repair of common iliac artery-to- 
inferior vena cava fistula. JVIR 1995;6:803-806. 



chll.qxd 4/16/04 3:36 PM Page 223 




Penetrating Cervical 
Vascular Injury 



RAO R. IVATURY 
MICHAEL C. STONER 



O SURGICAL ANATOMY 

O INCIDENCE AND ETIOLOGY OF CIVILIAN INJURIES 

O INCIDENCE AND ETIOLOGY OF MILITARY INJURIES 

O PATHOLOGY 

O INITIAL EVALUATION AND MANAGEMENT 

O DIAGNOSIS OF VASCULAR INJURIES: ZONES OF NECK AND THE ROLE 
OF PHYSICAL EXAMINATION 

Angiography 

Color Flow Doppler 

Computed Tomographic Angiography and Magnetic Resonance 
Angiography 

O TREATMENT 

Innominate and Subclavian Vessel Injuries 

Carotid Artery Injuries 

Surgical treatment 

Surgical Treatment of Carotid Artery Injury in Patients with 
Neurologic Deficits 

Vertebral Artery Injuries 

Venous Injuries 

O SUMMARY 



223 



chll.qxd 4/16/04 3:36 PM Page 224 



224 



IV • SPECIFIC VASCULAR INJURIES 



Injuries to the arteries of the neck are rel- 
atively uncommon but are of paramount 
importance to the trauma and vascular 
surgeon because of their end organ. The inci- 
dence of arterial injury in patients with pen- 
etrating neck wounds is 12% to 13%, whereas 
the incidence of venous injury is 18% to 19% 
(Asensio and colleagues, 1991, 2001). Mor- 
tality attributable to cervical vascular injury 
has been cited to be as high as 11%. Because 
of the relatively low overall incidence, it is dif- 
ficult for any one surgeon to gain extensive 
experience in the repair and management of 
these injuries. This underscores the impor- 
tance of multicenter and registry reviews when 
determining the proper treatment of these 
trauma patients. Most of these injuries, and 
certainly the most significant, are carotid 
artery injuries. 

Ambrose Pare is credited with the first 
recorded attempt to surgically treat a carotid 
artery injury more than 400years ago (Watson 
and Silverstone, 1939) . Pare ligated the carotid 
artery of a French soldier, saving his life but 
leaving him with a profound neurologic 
deficit consisting of left-sided hemiplegia 
and aphasia. Mr. Flemming (1817), 250 
years later, successfully ligated the common 
carotid artery of a patient aboard the H.M.S. 
Tonnant. The operation was successful in 
controlling hemorrhage without neurologic 
consequences. 

Simple ligation dominated surgical decision 
until the last 50 years. The devastating 
outcome of stroke or death following ligation 
of the carotid artery led surgeons to adopt 
a conservative approach to these injuries. 
Surgery was proposed for those patients with 
severe hemorrhage, enlarging hematoma, 
airway compromise, or pseudoaneurysm. 

As with most penetrating trauma, military 
conflicts have provided the bulk of informa- 
tion about penetrating carotid artery injuries. 
In World War I, Makins (1919) reported that 
one third of 128 cases of carotid artery injury 
treated by ligation resulted in irreversible neu- 
rologic deficit. During World War II and the 
Korean conflict, there were a handful of 
reports of carotid artery repair (Lawrence and 
colleagues, 1948; Huhges, 1958). 

The Vietnam conflict provided a wealth 
of information about carotid artery trauma. 



By this time, improved diagnosis, vascular 
operative techniques, and instrumentation 
yielded significantly improved morbidity and 
mortality. The Vietnam Vascular Registry, a 
massive project, was instrumental in shaping 
the modern approach to cervical vascular 
trauma. 

The unfortunate rise in civilian penetrat- 
ing trauma has been documented in various 
series. Several factors have led to improved 
management, probably the most significant 
of which is expeditious intervention. Fogel- 
man and Stewart (1956) demonstrated a 
35% mortality rate in patients with delayed 
surgical exploration versus 6% for those 
undergoing immediate operation. The 
authors advocated prompt exploration of all 
wounds penetrating the platysma, and this 
soon became a standard of care. This aggres- 
sive approach led to a high rate of negative 
explorations and caused many surgeons to 
reconsider the indications for operation. This 
controversy continues today and is outlined 
in further detail within this chapter. 



SURGICAL ANATOMY 



The key landmark to exploration of the neck 
is the sternocleidomastoid muscle, which 
defines the anterior and posterior triangles 
and is invested by the deep cervical fascia. Inci- 
sion over the anterior aspect of the muscle 
is the preferred route of exposure for most 
carotid injuries. Thoracic and cranial exten- 
sions are easily adapted to this incision as 
warranted. 

The most superficial layer of the neck mus- 
culature is the platysma, a thin confluence of 
muscle arising from the upper portion of pec- 
torals and inserting onto the skin and subcu- 
taneous tissue of the lower face. As described 
earlier in this chapter, the platysma is an impor- 
tant landmark of historical significance defin- 
ing superficial from deep penetrating cervical 
wounds. The external jugular vein is the only 
vascular structure between this layer and the 
deep cervical fascia. 

Emerging from the jugular foramen at 
the base of the skull, the internal jugular vein 
courses downward within the carotid sheath, 



chll.qxd 4/16/04 3:36 PM Page 225 



11 • PENETRATING CERVICAL VASCULAR INJURY 



225 



along with the carotid artery and vagus nerve. 
Each component of the carotid sheath is encir- 
cled in its own connective tissue investment 
throughout the cervical region. Because of 
the considerable distensibility of these con- 
nective tissue layers, the carotid sheath tends 
to be attenuated over the jugular vein. The 
internal jugular ends at the clavicle where 
it joins with the subclavian vein to form the 
brachiocephalic vein. On the left side, an 
important posterior anatomic relationship is 
the thoracic duct, which eventually inserts at 
the confluence of the jugular and subclavian 
veins. 

In most patients, the right carotid arises 
from the brachiocephalic artery, and the 
left from the aortic arch. Each artery passes 
upward to the level of the thyroid cartilage 
and divides into internal and external 
branches. Prior to this point, the common 
carotid is without branches except for the rare 
anomalous superior thyroid artery or ascend- 
ing pharyngeal branch. At the bifurcation, the 
artery dilates, and this region is known as the 
carotid bulb. The carotid bifurcation can be 
difficult to identify in the presence of an 
extensive hematoma. An important land- 
mark in identifying the bifurcation is the loca- 
tion of the facial vein and the medial portion 
of the ansa cervicalis. At their origin, the 
common carotid arteries are relatively close, 
separated only by the trachea. As they ascend, 
thyroid, larynx, and then pharynx intervene 
between the two arteries. Sternocleidomastoid 
covers the common carotid artery, the inter- 
nal carotid artery (ICA) , and the external 
carotid artery throughout their course except 
for a small window between its anterior border 
and digastric muscle at the base of the skull 
(Fig. 11-1). 

The ICA is typically larger than the exter- 
nal and supplies the anterior part of the brain 
and the eye and sends branches to the face 
and nose. At the origin of the ICA is a pressor 
receptor, the carotid sinus, stimulation of 
which results in hypotension and bradycardia. 
Along the posterior aspect of the artery runs 
the sympathetic trunk, and the spinal mus- 
culature behind that. The esophagus is medial 
and is of paramount importance when assess- 
ing for digestive injury. The hypoglossal nerve 
courses over the anterolateral aspect and is 



an important surgical landmark. A series of 
important relationships are essential in dif- 
ferentiating the ICA from the external carotid 
artery: (1) In the vast majority of patients, the 
ICA has no cervical branches; (2) the ICA 
usually lies posterolateral to external carotid; 
and (3) asitascends, the ICA moves to a medial 
position relative to external carotid artery 
(Fig. 11-2). 

The external carotid artery, termed exter- 
nal because if its extracranial distribution, 
extends upward to the mandibular neck 
where it divides into its two terminal branches: 
the superficial temporal artery and the max- 
illary artery. The main trunk of the external 
carotid quickly gives rise to a series of branches: 
superior thyroid, ascending pharyngeal, 
lingual, facial, occipital, and posterior auric- 
ular. Of note, a portion of the external carotid 
is encompassed by the parotid salivary gland, 
an important consideration in exposure and 
repair. 

The vertebral artery is the first and largest 
branches of the subclavian artery. It ascends 
behind the common carotid artery to enter 
the vertebral foramen at the transverse process 
of the sixth cervical vertebrae. Before enter- 
ing the base of the skull, the vertebral artery 
passes behind the lateral aspect of the atlas, 
in an almost horizontal plane. Once entering 
the skull through the foramen magnum, the 
paired vertebral arteries converge, giving rise 
to the basilar artery and proceed to the circle 
of Willis as their termination (Fig. 11-3). 

It is important for the surgeon to keep in 
mind the many potential collaterals between 
the internal and external system, because 
these collaterals may be able to sustain blood 
flow after ICA or external carotid artery injury. 
One significant collateral is between the 
ophthalmic branch of internal carotid and 
the facial branch of external carotid artery. 
Second, the thyrocervical trunk rises up the 
neck between thejugular vein and the antero- 
medial boarder of sternocleidomastoid. It is 
a potential source of collateral flow to the 
external carotid artery. Perhaps the most 
important and often misrepresented collat- 
eral circulation is the circle of Willis (see Fig. 
11-3). Historical autopsy data suggest that 
up to one half of patients will have "nonstan- 
dard" circulation here (Puchades-Orts and 



chll.qxd 4/16/04 3:36 PH Page 226 



226 



IV • SPECIFIC VASCULAR INJURIES 




Post, communicating a 

Post, cerebral a. 
Int. carotid a 
Basilar a 
Vertebral aa 



Superf. temporal a. 
Basilar a 



Int. carotid a 

Occipital a 

Ascend, 
pharyngeal a. 

Vertebral a 



Ext. carotid a. 
Facial a. 
gual a. 
Superior thyroid a. 

Common carotid a. 



■ FIGURE 11-1 

Except for the intrathoracic portion of the left common carotid artery, the cervical anatomy of the 
carotid arteries and the vertebral arteries is bilaterally similar. ■ 



chll.qxd 4/16/04 3:36 PH Page 227 



11 • PENETRATING CERVICAL VASCULAR INJURY 



227 



Rights were not granted to include this figure in electronic media. 
Please refer to the printed publication. 



■ FIGURE 1 1-2 

The close proximity of important contiguous structures, such as the internal jugular vein and the 
vagus nerve, is emphasized. The cross section shows the contents of the carotid sheath and the 
associated surrounding structures. ■ 



chll.qxd 4/16/04 3:36 PH Page 228 



228 



IV • SPECIFIC VASCULAR INJURIES 



Ant. cerebral a. 
Ant. communicating a. 




Middle cerebral a. 



Post communicating a 



Post, cerebral a 



=k Basilar a. 



m FIGURE 11-3 

The standard model of the circle of Willis. 
Although normal variations within the circle are 
common, hypoplasia of various segments may 
be particularly significant with interruption of 
either internal carotid artery. This may result in 
an inadequate collateral flow. (From Strandness 
DE Jr. Collateral Circulation in Clinical Surgery. 
Philadelphia: WB Saunders, 1969.) ■ 



colleagues, 1976). Because of the redun- 
dancy, hypoplasia of an individual component 
of the circle is rarely a problem, unless carotid 
flow is interrupted and the segments of the 
circle are unable to sustain adequate flow. A 
recent magnetic resonance angiographic 
study supports this high rate of variance 
(Krabbe-Hartkamp and colleagues, 1998) . 
The authors noted that only 42% of randomly 
selected adults had complete circles, with pos- 
terior variations being the most common. 
Other minor collateral circulations exist such 
as vertebral to vertebral muscular branches, 
external carotid to external carotid, and ver- 
tebral occipital branches. 



INCIDENCE AND ETIOLOGY OF 
CIVILIAN INJURIES 



The incidence of carotid artery injuries in 
civilian series ranges from 12% to 17% of 
total penetrating neck injuries. Information 
about the precise anatomic distribution of 



these injuries is sometimes limited by the 
presentation of data within a particular study 
and the lack of a standardized grading system 
and registry database specific for vascular 
injury. Recently, Mittal and colleagues (2000) 
have suggested a grading system to standard- 
ize the assessment and reporting of cervical 
vascular injuries. Compared to military 
injuries, civilian carotid injury tends to be 
blunt or stabbing. This must be considered 
when comparing outcome data between these 
groups. 



INCIDENCE AND ETIOLOGY OF 
MILITARY INJURIES 

Historic military data report that carotid 
artery injuries represent approximately 5 % of 
all arterial injuries. The most recent and exten- 
sive database for modern military traumatic 
cervical vascular injury is the Vietnam Vascu- 
lar Registry (Rich and colleagues, 1970) . Data 
from this experience indicate that fragment 
wounds (projectiles from explosive ordinance, 
shrapnel, or debris) accountfor the vastmajor- 
ity of wounds. Gunshot wounds were found 
to be much less common (especially as com- 
pared to civilian trauma) because military 
weaponry tends to be of high velocity and 
therefore more likely to be fatal. 

The incidence of carotid arterial injury in 
wartime, based on a total number of arterial 
injuries, was reported to be highest in a World 
War I study by Makins (1919) and lowest in a 
World War II study by De Bakey and Simeone 
(1946), as indicated in Table 11—1. The 
Vietnam conflict data indicate that common 
carotid injury is a more common occurrence 
than either internal or external carotid injury. 
Obviously, the vast majority of these injuries 
are penetrating, because of the nature of 
weaponry involved. 



PATHOLOGY 

The patient who survives penetrating neck vas- 
cular injury and reaches a surgeon most likely 
has a laceration or perforation. Complete 



chll.qxd 4/16/04 3:36 PH Page 229 



11 • PENETRATING CERVICAL VASCULAR INJURY 



229 



TABLE 1 1-1 

COMPILATION OF WARTIME DATA ILLUSTRATING THE INCIDENCE OF 
CAROTID ARTERY INJURY EXPRESSED AS A PERCENTAGE OF TOTAL ARTERIAL 
INJURIES REPORTED 



Study 

Makins, 1919 (World War I) 
De Bakey and Simeone, 

1946 (World War II) 
Hughes, 1958 (Korean) 
Rich and colleagues, 

1970 (Vietnam) 



Total Arterial 


Common 


Internal 






Injuries 


Carotid 


Carotid 


Total Carotid 


Percentage 


120 






128 


10.7 


247 






10 


0.4 


304 






11 


3.6 


100 


38 


12 


50 


5.0 



disruptions of the carotid artery are almost 
always fatal, although reports exist demon- 
strating viable patients with completely tran- 
sected carotid arteries, with thrombus 
formation at the severed ends, thus alleviat- 
ing the hemorrhage (Rich and colleagues, 
1970; Harris and colleagues, 1985). 

Cerebral vasospasm appears to be an impor- 
tant factor in the pathophysiology of cervical 
vascular injury. Acute spasm of the cerebral 
arteries can occur almost immediately after 
cervical arterial injury and can severely exac- 
erbate ischemia. For the most part, however, 
spasm seems to play a late role in the poten- 
tial pathology of carotid or vertebral artery 
injury, with the peak incidence occurring 5 
to 10 days after injury (Kordestani and col- 
leagues, 1997). Acute arteriovenous fistula is 
an important entity that if improperly iden- 
tified, will have a high propensity to recur 
(Marks and colleagues, 1984). Early diagno- 
sis can be difficult on physical examination 
alone, as the classic murmur may not be 
audible for several days. Some patients may 
have a substantial thrill that is palpable along 
the course of the fistula. Missile trajectory 
or pattern is very important when one 
attempts to predict possible fistulas. A report 
from our own institution described an acute 
carotid to cavernous sinus fistula after shotgun 
blast (Fields and colleagues, 2000). Diagnos- 
tic modalities for the workup of fistulas and 
other pathology associated with cervical vas- 
cular injury are discussed in the following 
section. 



INITIAL EVALUATION 
AND MANAGEMENT 

A careful history and physical examination 
should be undertaken. Weapon characteris- 
tics are important in predicting the extent of 
injury, especially for high-velocity missile 
injury. Trajectory should be assessed from 
entrance and exit wounds, especially in trans- 
cervical wounds because of the high inci- 
dence of vital structure damage (Hirshberg 
and colleagues, 1994). Obvious signs of vas- 
cular injury include external hemorrhage, 
expanding or pulsate hematoma, decreased 
pulses, and avascular bruit or thrill. So-called 
"soft signs" of vascular injury include dimin- 
ished temporal or facial arterial pulses, signs 
of hemothorax, or pharyngeal bleeding. 
Patients with hard signs of vascular injury 
require emergent neck exploration. A thor- 
ough neurologic examination is an integral 
part of the patient's evaluation. Care should 
be taken to ascertain for signs of cord injury 
such as paralysis, paresthesia, and hyper- 
reflexia. Likewise, severe cerebral vascular 
insufficiency from carotid injury may result 
in contralateral hemiparesis. Evidence of 
cerebral infarct is important. This is discussed 
later in this chapter in the context of vascu- 
lar repair in a patient with preexisting neu- 
rologic deficit. 

Initial clinical management begins in the 
prehospital phase of trauma care. For the 
patient with penetrating cervical injury, 



chll.qxd 4/16/04 3:36 PH Page 230 



230 



IV • SPECIFIC VASCULAR INJURIES 



prompt transport to the nearest trauma 
center with skilled personnel is paramount. 
Because airway establishment is fraught with 
a variety of potential problems, patients 
should probably be intubated only if unre- 
sponsive or if their situation is deteriorating 
and a long transport time is anticipated. Exter- 
nal bleeding should be controlled with direct 
pressure. 

When the patient presents to the trauma 
bay with a penetrating neck injury, airway 
control should be the foremost concern. Most 
early deaths in these patients result from either 
airway compromise or external hemorrhage, 
both of which can be addressed with fastidi- 
ous care. The airway management algorithm 
is straightforward for the unresponsive patient 
or those presenting in extremis (Rao and 
colleagues, 1993): rapid establishment of 
orotracheal or surgical airway. The difficult 
patient is the agitated patient with obvious 
severe cervical injury. Establishment of 
orotracheal or nasotracheal airway may be 
problematic because of blood or secretions 
in the oropharynx or the patient's general 
status. 

These patients need the most experienced 
personnel in the most optimal place possible 
in the most expeditious way. In our opinion, 
these patients, if at all possible, should be 
rushed to the operating room where the most 
experienced anesthesiologist and surgeon can 
together manage the airway by carefully indi- 
vidualized interventions: conscious intubation 
or sedation and a rapid surgical airway. 

Once the patient's airway is evaluated and 
steps are taken to ensure definitive tracheal 
intubation as required, massive external bleed- 
ing needs to be controlled. Precise digital 
pressure should be applied to any bleeding 
sites. Nasopharyngeal or oropharyngeal 
packing may be necessary as a temporary 
hemostatic aid. During this time, paired large- 
bore intravenous access should be obtained 
and secured. Anteroposterior and lateral 
cervical roentgenograms and chest radi- 
ograms are obtained to assess for retained pro- 
jectile, bony injury, and thoracic or pleural 
violation. 

Outside the context of this chapter, con- 
current injury must be assessed and dealt with. 
The high incidence of aerodigestive tract 



injury with penetrating neck trauma has been 
well described and may require rapid diag- 
nosis and treatment. Severe tracheal injury 
may complicate the establishment of a defin- 
itive airway in these patients and can prove 
fatal. Delay to diagnosis of esophageal or 
pharyngeal injury is directly related to poor 
outcome and must be assessed fastidiously 
(Asensio and colleagues, 2001). 



DIAGNOSIS OF VASCULAR 
INJURIES: ZONES OF NECK 
AND THE ROLE OF 
PHYSICAL EXAMINATION 



In a landmark 1969 article, Monson and col- 
leagues (1969) arbitrarily divided the neck into 
three clinical zones (Fig. 11-4). Zone 1 is 
defined as being below to sternal notch, zone 
3 above the angle of the mandible, and zone 
2 is the intervening region. This system has 
become a standard for discussing the diag- 
nostic approach to penetrating neck vascular 
trauma because of the clinical relevance of 
injuries to anatomic locations. Zone 2 injuries 
are accessible via standard cervical approaches 
and should not present difficulty in obtain- 
ing proximal and distal vascular control. 
Zone 1 injuries by definition involve the tho- 
racic inlet, and proximal control may require 
thoracotomy or supraclavicular incisions in 
addition to a cervical dissection. Zone 3 
injuries are problematic because of the poten- 
tial difficulty in obtaining distal control. The 
widely accepted practice is thatzone 2 injuries 
are explored in the operating room, whereas 
zone 1 and 3 injuries require angiography 
because of the more extensive surgical 
approaches and potential complications. 

Surgical access and exposure for sympto- 
matic vascular injuries is relatively easy, and 
the morbidity from surgical exploration of 
zone 2 is very low. In addition, several studies 
have suggested that because zone 2 of the neck 
is amenable to physical examination, signifi- 
cant vascular injuries in this location are rarely 
occult. Sekharan and colleagues (2000) 
reviewed 145 patients with neck penetration; 
in 30 of these patients, the penetrating tra- 
jectory also traversed zone 1 or 3. Thirty-one 



chll.qxd 4/16/04 3:36 PH Page 231 



11 • PENETRATING CERVICAL VASCULAR INJURY 231 
ZONES OF NECK FOR TRAUMA 




■ FIGURE 1 1-4 

Zones of the neck. ■ 



patients (21%) had hard signs of vascular 
injury (active bleeding, expanding hematoma, 
bruit/ thrill, pulse deficit, central neurologic 
deficit) and were taken immediately to the 
operatingroom;28 (90%) of these 30 patients 
had either major arterial or venous injuries 
requiring operative repair (the false-positive 
rate for physical examination thus being 
10%). Of the 114 patients with no hard signs, 
23 underwent arteriography because of prox- 
imity of the injury to the vertebral arteries or 
because the trajectory included another zone. 
Of these 23 arteriograms, three showed 
abnormalities, but only one required opera- 
tive repair. This case had no complications 
relating to the initial delay. The remaining 91 
patients with no hard signs were observed 
without imaging or surgery for a minimum 
of 23 hours, and none had any evidence of 
vascular injury during hospitalization or 
during the initial 2-week follow-up period 
(1/114; false-negative rate for physical 



examination, 0.9%) . Based on these data, the 
authors confirmed that patients with zone 
2 penetrating neck wounds can be safely 
and accurately evaluated by physical exami- 
nation alone to confirm or exclude vascular 
injury. 



Angiography 

Four-vessel cervical angiography is the gold 
standard by which all other modalities are 
judged. The goal of angiography is to define 
the exact arterial injury and it anatomic rela- 
tions, to identify any collateral flow, to look 
for any arteriovenous communications, and 
to stratify injuries in the patient with multiple 
vascular injuries. As angiography became 
readily available in the last 30 years, this system 
has become useful to define which injuries 
require arteriography. The precise indications 
for arteriography in stable patients, however, 



chll.qxd 4/16/04 3:36 PH Page 232 



232 



IV • SPECIFIC VASCULAR INJURIES 



remain controversial. Preoperative arteriog- 
raphy is recommended in all patients with 
zone 1 penetration if they are hemodynamically 
stable and do not have evidence of active hemor- 
rhage. It is recommended because these 
injuries are frequently clinically occult and 
their exposure technically challenging. 
Angiography appears to be essential in zone 3 
neck wounds because of the potential inacces- 
sibility of distal vascular injuries. The role of 
angiography in identifying injuries in zone 2 
is more controversial, as discussed earlier in 
this chapter. 

Arteriography is more likely to be helpful 
in low-velocity gunshot wounds than in 
stab wounds, but recommendations to eliminate 
arteriography from a selective management proto- 
col in stab wounds of the neck must await further 
studies. High-velocity bullet wounds, shrapnel 
injuries, and close-range shotgun wounds 
all have exceptionally high incidences of 
significant organ injury, and arteriography 
in the stable patient without exsanguinating 
hemorrhage may be of considerable 
assistance. 

Azuaje and colleagues (2002) studied 216 
patients with penetrating neck injuries (from 
1992 to 2001). Excluding 48 emergent explo- 
rations and 16 shotgun wounds, the remain- 
ing 152 patients had injuries in zone 1 (45 
patients), zone 2 (83 patients), and zone 
3 (23 patients); 63 patients had a positive 
physical examination (e.g., hematoma, bruit, 
thrill, and bleeding) and 40 (68%) also had 
a positive angiogram. Twenty of these required 
operative repair. Of the 89 patients with a neg- 
ative physical examination, only 3 had a pos- 
itive angiogram and none of these required 
operative repair. Physical examination had 
93% sensitivity and a 97% negative predictive 
value for predicting the results of angiogram. 
The authors concluded that with careful phys- 
ical examination, angiography may not be 
necessary irrespective of zone of injury. 



Color Flow Doppler 

Recently, color flow Doppler has become an 
alternative to angiography in the diagnostic 
workup of vascular trauma. It has been studied 
to some extent in peripheral arterial injuries, 



although its use in cervical trauma is ill 
defined. A study by Fry and colleagues (1994) 
demonstrated the feasibility of ultrasound 
for the assessment of vascular injury. A pilot 
study with 15 patients receiving duplex ultra- 
sonography and concomitant angiography 
demonstrated equal accuracy of the two tech- 
niques. With these pilot data, the study was 
extended to an additional 85 patients in whom 
ultrasound was the primary imaging modal- 
ity. The authors reported that ultrasound was 
as effective as operative exploration or angiog- 
raphy in their hands. A smaller study in 1996 
demonstrated the success of ultrasonography 
as a screening modality for vascular trauma, 
with all major injuries being detected by the 
sonogram (Montalvo and colleagues, 1996) . 
These findings were confirmed by Demetri- 
ades and colleagues (1995) in their study of 
82 patients. They demonstrated that Doppler 
imaging identified 10 of the 11 injuries, for a 
sensitivity of 91% and a specificity of 98.6%. 
It appears that careful examination coupled 
with the experienced use of ultrasonography 
provides results comparable to angiography. 
Pitfalls with Doppler do exist though, impor- 
tantly, the difficulty in establishing the exact 
anatomic relationship with the resolution 
available to angiography and the difficulty 
in securing readily available equipment, an 
experienced technologist, and skilled 
interpreter. 



Computed Tomographic 
Angiography and Magnetic 
Resonance Angiography 

Currently attracting increasing attention, 
computed tomographic angiography (CTA) 
should still be considered unproven. One 
study looked at 16 patients with suspected 
traumatic carotid artery injury who underwent 
CTA. Twelve of these patients had penetrat- 
ing injuries and four had blunt injuries to the 
neck. All the CTAs were diagnostic. Positive 
findings included one complete tear of the 
right common carotid artery (confirmed by 
surgery) from a penetrating injury and one 
bilateral ICA thrombosis after blunt injury to 
the neck. Negative findings on CTA were con- 



chll.qxd 4/16/04 3:36 PM Page 233 



11 • PENETRATING CERVICAL VASCULAR INJURY 



233 



firmed by surgical exploration (Ofer and 
colleagues, 2001). In a prospective study of 
146 arteries (77 carotid, 69 vertebral) studied 
by means of conventional angiography and 
helical CTA, conventional angiograms showed 
arterial injuries in 10 (17%) of 60 patients. 
These included arterial occlusion ( n = 4) , arte- 
riovenous fistula (w=2), pseudoaneurysm 
(n= 3), pseudoaneurysm with arteriovenous 
fistula (n= 1), and normal arteries (n= 136). 
Nine of ten arterial injuries and all normal 
arteries were depicted adequately at helical 
CTA. The sensitivity of helical CTA was 90%, 
specificity was 100%, positive predictive value 
was 100%, and negative predictive value was 
98%. The sensitivity and specificity of helical 
CTA, therefore, seem high for detection of 
major carotid and vertebral arterial injuries 
resulting from penetrating trauma (Munera 
and colleagues, 2000), at least in experienced 
hands. 

Magnetic resonance angiography has been 
described as an imaging modality in this 
context as well, but experience with pene- 
trating neck trauma, technical considera- 
tions, and limited experience make it difficult 
to recommend its use (Prabhu and colleagues, 
1994; Friedman and colleagues, 1995). 



TREATMENT 

Innominate and Subclavian 
Vessel Injuries 



subclavian are best approached by an incision 
along the clavicle with extension along the 
sternocleidomastoid, if necessary. Subpe- 
riosteal resection of the mid or medial one 
third of the clavicle will allow excellent expo- 
sure. Most of these vascular injuries may be 
managed by simple closure or end-to-end anas- 
tomosis. Occasionally, a saphenous vein graft 
may be necessary. In stable patients with sub- 
clavian pseudoaneurysm or intimal injuries, 
endovascular stent grafts are an evolving 
option. 

Carotid Artery Injuries 

The general consensus now is that injuries to 
the carotid arteries are best approached by 
surgical intervention and repair, either by end- 
to-end anastomosis or graft techniques, espe- 
cially in the accessible portions of the carotid 
artery (Figs. 11-5 and 11-6). For selected 
intracranial injuries high in zone 1, endovas- 
cular stents are being used with increasing fre- 
quency, as discussed later in this chapter. 



SURGICAL TREATMENT 

The patient should be placed in a prone posi- 
tion with both arms tucked in if possible. As 
long as there is no concurrent spine injury, a 
shoulder roll should be placed to extend the 
neck and the table placed in a semi-Fowler 



Operative exposure of zone 1 vascular injuries 
are outside the scope of this chapter and are 
only briefly mentioned for completion. If the 
patient is hemodynamically unstable with a 
large hemothorax or excessive bleeding from 
the chest tube, an anterolateral thoracotomy 
(high in the third or fourth intercostal space) 
in the emergency center will allow apical 
packing and tamponade the bleeding from 
innominate or proximal subclavian vessels. In 
the operating room, a median sternotomy will 
expose the innominate. For the left subcla- 
vian artery, the incision maybe extended along 
the sternocleidomastoid or the clavicle. This 
approach is superior to the "trap-door" inci- 
sion. The second and third portions of the 




■ FIGURE 1 1-5 

Gunshot wound of the neck. Angiogram reveals 
internal carotid cutoff just distal to its origin. ■ 



chll.qxd 4/16/04 3:36 PM Page 234 



234 IV • SPECIFIC VASCULAR INJURIES 
I 




■ FIGURE 1 1-6 

A and B, Carotid artery injury at operation 
repaired by a saphenous vein interposition 
graft. ■ 

position to aid in the operative exposure. The 
entire thorax and abdomen should be pre- 
pared in case of zone 1 injury for accessing 
the abdomen for potential multicavitary 
injuries. The groin and both thighs are pre- 
pared and draped for possible saphenous vein 
harvest. Carotid exposure is obtained via a skin 
incision made along the anterior border of 
the sternocleidomastoid muscle. The carotid 
sheath and its contents are readily identifiable. 
The venous and lymphatic structures are 
retracted in a lateral direction. Proximal and 
distal exposure of the carotid arteries is 
obtained after identifying the ansa cervicalis 
and twelfth cranial nerve. Digital pressure or 
a side-biting vascular clamp can be used to 
control hemorrhage while obtaining control. 
Proximal injury to the carotid at its aortic or 
subclavian origin requires more extensive 
exposure than the simple neck incision. 



Median sternotomy is the most commonly 
employed approach. A supraclavicular inci- 
sion is useful for exposure in some cases, and 
dislocation or resection of the clavicle may 
improve the exposure, as detailed elsewhere 
in this text. 

High zone 3 injuries resulting in distal inter- 
nal carotid laceration or disruption can prove 
very problematic in exposure and control. 
Anterior subluxation of the mandible can 
improve exposure, but only by about 2 cm. 
Osteotomy of the mandibular ramus may 
provide better exposure and mobility. Place- 
ment of a Fogarty balloon catheter to provide 
distal vascular control can be lifesaving during 
these maneuvers. The carotid artery may 
require ligation. Depending on the surgeon's 
experience in operating around the base of 
the skull, intraoperative assistance from either 
a maxillofacial surgeon or a neurosurgeon 
is advisable in difficult cases. A recent series 
illustrated the management and outcomes of 
four consecutive patients, two with pseudo- 
aneurysms and two with acute occlusions, after 
injury to the distal cervical/petrous ICA from 
gunshot wounds. Preoperative assessment 
determined intracranial collateral flow pat- 
terns and established the patency of the distal 
portion of the petrous ICA. Two patients 
underwent cervical-to-petrous ICA vein bypass 
grafts without neurologic complications. Both 
grafts remained patent without evidence of 
emboli at 2 years and 3 months. The two 
patients who were managed conservatively 
died, one from a massive cerebral infarction 
and the other from intracerebral hemorrhage. 
These authors concluded that the cervical- 
to-petrous ICA vein bypass graft is a valuable 
management option that can reduce the 
potential morbidity and mortality from acute 
ischemic or delayed embolic or hemorrhagic 
infarcts (Romily, Newell, and Grady, 2001). 
This approach, however, may be of limited 
value in the management of the bleeding 
patient with a difficult surgical exposure. 
In this setting, ligation of the ICA may be 
lifesaving. 

Once proximal and distal control are 
secured, a Fogarty balloon catheter is care- 
fully passed to remove any thrombus and 
both proximal and distal ends are flushed 
with heparinized saline. A monofilament 



chll.qxd 4/16/04 3:36 PM Page 235 



11 • PENETRATING CERVICAL VASCULAR INJURY 



235 



polypropylene suture of 5-0 or 6-0 size is used 
for the repair and handled with appropriate 
vascular technique. Except for tangential 
lacerations, primary repair is often difficult, 
because the transected ends of the artery 
retract. For a simple, small laceration, inter- 
rupted lateral repair is usually possible. Larger 
lacerations require a running repair, with two 
separate sutures, each originating in an apex 
of the injury and is approximated in the 
middle. Care must be taken to inspect for and 
repair any intimal flap at this time. This can 
often be done by incorporating the intimal 
defect into the laceration repair by using a 
series of interrupted sutures. Also, the lumen 
of the artery must not be narrowed by this 
primary repair; otherwise, vein patch or inter- 
position graft will be required. 

Stab wounds or low-velocity missile injuries 
often result in simple lacerations with minimal 
devitalized tissue. Most of these arteriotomies 
can be repaired with simple suture plication 
or minimal mobilization and primary repair. 
If there is a considerable destruction of the 
carotid artery, resection of devitalized tissue 
may preclude a tension-free primary repair. 
In these cases, interposition grafting is war- 
ranted, preferably using autologous tissue. 
Saphenous vein is generally accepted to be 
the ideal conduit. If adequate autologous 
conduit is not available, synthetic material may 
be used. The choice between woven Dacron 
and ethyl polytetrafluoroethylene is based on 
surgeon's preference, because neither rep- 
resents an ideal substitute for a vein graft. If 
anatomy permits, the external carotid artery 
can be divided at a distal location and trans- 
posed to the ICA. The thyroidal and pharyn- 
geal branches should be ligated to provide 
mobility. The use of a shunt is not mandated 
by the available data. The Vietnam experience 
established the safety of carotid artery repair 
without the use of shunts, even with arterial 
occlusion lasting up to 60 minutes. In many 
series, stump pressures (pressure measured 
in the distal portion of carotid artery after 
ligation or resection) failed to predict post- 
operative neurologic deficits after carotid 
endarterectomy. Even though similar data are 
not available in civilian trauma series, the use 
of a shunt or stump pressures is a matter of 
personal choice. 



Recently, percutaneous transluminal 
placement of endovascular devices has 
become an alternative option to surgical 
repair. One series evaluated the potential for 
using flexible self-expanding uncovered stents 
with or without coiling to treat post-traumatic 
pseudoaneurysms involving the extracranial 
ICA, the subclavian artery, and other periph- 
eral artery (Assali and colleagues, 2001). 
Three patients with post-traumatic pseudo- 
aneurysms of the carotid (one patient) and 
subclavian (two patients) were treated by stent 
deployment (Fig. 11-7). Angiography demon- 
strated complete occlusion of the pseudo- 




■ FIGURE 11-7 

A and B, Subclavian artery injury just proximal 
to its origin by a gunshot wound in a patient 
with severe associated brain injury. This was 
managed by a stented graft placed by the 
interventional radiologist. (Courtesy T. Marrone 
and J. Tisnado.) ■ 



chll.qxd 4/16/04 3:36 PH Page 236 



236 



IV • SPECIFIC VASCULAR INJURIES 



aneurysms. At long-term follow-up (6 to 9 
months), all patients were asymptomatic 
without flow into the aneurysm cavity by 
Duplex ultrasound. 

Much like in the extremities, minimal 
injuries to the carotid artery (small pseudo- 
aneurysms or intimal flaps) probably have a 
benign course, and may not necessarily require 
operative repair. The natural course of many 
intimal flap injuries is unknown. Considering 
the nature and importance of the carotid cir- 
culation, however, the most judicious course 
of action may be a serial objective evaluation 
via duplex color flow ultrasonography or 
angiography. In a recent experimental study, 
only up to one third of all intimal injuries 
resolved without complication, and this figure 
underscores the need for diligent follow-up 
(Panetta and colleagues, 1992). There is no 
definitive information from the literature that 
elucidates the role of anticoagulation or 
antiplatelet agents in carotid artery injuries. 
Some authors, based on elective carotid 
endarterectomy data, advocate the use of low- 
dose aspirin or intravenous low-molecular- 
weight dextran to provide prophylaxis 
against thrombosis (Robless and colleagues, 
1999). 

Surgical Treatment of Carotid Artery 
Injury in Patients with Neurologic Deficits 

Bradley in 1973 and Thai and colleagues in 
1974 challenged the concept of repairing the 
carotid artery in the face of an established cere- 
bral infarct and suggested that the repair and 
establishment of carotid flow may convert an 
anemic infarct into a hemorrhagic infarct. 
This concept, however, was refuted by a col- 
lective series of 223 patients by Liekwig and 
Greenfield (1978). Similar data were pre- 
sented by Unger, Jorgensen, and Hoffman 
(1990), Lawrence and colleagues (1948), 
Ledgerwood, Mullins, and Lucas (1980), and 
Weaver and colleagues (1988) . Many of these 
studies concluded that the outcome was 
dismal regardless of the type of therapy in the 
patient with a fixed profound neurologic 
deficit, but that the patients did better with 
repair than ligation. It is advisable, therefore, 
to repair the injury as long as the patient's 
clinical condition permits. 



The prognosis for penetrating carotid artery 
injuries depends on the neurologic status on 
admission (Lawrence and colleagues, 1948; 
Ledgerwood, Mullins, and Lucas, 1980; Deme- 
triades and Stewart, 1985; Asensio and col- 
leagues, 1991; Demetriades and colleagues, 
1996a). The mortality rates range from 6.6% 
to 33%, with an average of 17% (Asensio 
and colleagues, 1991), mostly related to 
neurodeficits. 



Vertebral Artery Injuries 

Vertebral artery injuries fortunately are not 
common. The first clinical review of vertebral 
artery injuries is attributed to Rudolph Matas 
of New Orleans in 1893. In his landmark 
article, Matas elegantly described the difficulty 
in diagnosing and surgically managing verte- 
bral artery injuries, "A glance at the surgical 
anatomy of this vessel as it lies deeply hidden 
in the skeleton of the neck, only escaping 
at short intervals from its osseous canal, to 
become immediately invested by the very 
important and vital cervical nerves as they issue 
from the spinal foramina, will at once remind 
us of the magnitude of the purely technical 
difficulties in the way of its atypical ligation, 
and of the errors of diagnosis that must 
be incurred" (Yee and colleagues, 1995). 
Reported injuries to the vertebral artery have 
been exceedingly rare during major conflicts 
involving the United States. Although three 
vertebral artery injuries were reported in 
World War I, none were reported in either 
World War II, the Korean War, or the Vietnam 
War (De Bakey and Simeone, 1946). 

If identification and exposure of the ver- 
tebral artery injury are uncomplicated during 
neck exploration, proximal and distal surgi- 
cal ligation of the injured vessel is performed. 
If encountered at neck exploration, the fol- 
lowing steps are indicated: (1) gauze packing 
at the site of bleeding, (2) exposure of the 
subclavian artery by dividing the origin of ster- 
nomastoid from the clavicle, and (3) proxi- 
mal control of the origin of the subclavian 
artery. The neck incision is carried posterior 
to the ear with division of the attachment of 
sternomastoid and splenitis capitis muscle. 
Distal ligation may be performed by dividing 



chll.qxd 4/16/04 3:36 PH Page 237 



11 • PENETRATING CERVICAL VASCULAR INJURY 



237 



the splenitis capitis and sternomastoid attach- 
ments to the mastoid, palpation of the trans- 
verse process of the atlas, and exposing the 
vertebral artery between the axis and atlas. 
Bone wax or other hemostatic agents can be 
used to pack and compress this area, or "blind " 
application of surgical clips deep into the 
wound may staunch the bleeding. 

Because of the anatomic difficulties in 
vascular control of the vertebral artery, angio- 
graphic embolization represents an accept- 
able alternative. If the injured vertebral artery 
is discovered in the initial evaluation by 
angiography, the surgeon has a "road map" 
in planning the operative approach. Addi- 
tionally, preoperative angiography allows for 
selection of patients who may not require lig- 
ation, specifically those with vertebral artery 
narrowing or occlusions. The approach to 
patients with pseudoaneurysms, dissections, 
arteriovenous fistulas, or extravasations will 
depend on the skill and judgment of the 
trauma surgeon, as well as the availability of 
experienced neuroradiologists. 

Some series (Reid andWeigelt, 1970) noted 
several cases in which a disrupted artery, as 
seen operatively, had been diagnosed inac- 
curately as an occlusion by preoperative 
angiography. As a result, they recommended 
neck exploration for all angiographically 
diagnosed vertebral artery injuries unless 
deemed minimal. In the recent series from 
San Francisco (Yee and colleagues, 1995) , five 
of six patients with vertebral artery occlusion 
were treated by clinical observation, whereas 
one underwent embolization. No adverse 
symptoms or neurologic sequelae were seen 
in these five patients. In addition, three 
patients with angiographic narrowing were 
treated by observation without complication, 
which are findings consistent with those 
reported by others. In the series reported 
by Demetriades and colleagues (1996b), 22 
patients with vertebral artery injuries were 
reviewed. Only four patients required an emer- 
gency operation. Most of the injuries (13/22) 
were successfully managed by observation. 
Five patients were managed by angiographic 
embolization, which was successful in three. 
In three patients with an aneurysm and arte- 
riovenous fistula, proximal embolization of the 
vascular lesion was not adequate and a sub- 



occipital craniectomy was required for distal 
ligation. Neurologic sequela from vertebral 
embolization is very uncommon (Demetri- 
ades and Stewart, 1985). These data support 
the conclusion that most vertebral artery 
injuries can safely be managed without an 
operation or by angiographic embolization 
(Fig. 11-8). Surgical intervention should be 
reserved for patients with severe bleeding or 
in whom embolization has failed (Demetri- 
ades and colleagues, 1989). 



Venous Injuries 

Injuries to the innominate, subclavian, axil- 
lary, or internaljugular vein may be the source 
of severe hemorrhage. All of these veins can 
be ligated if the destruction is severe. In stable 
patients, they may be repaired. In a recent 
series of 49 consecutive patients with cervical 
and thoracic venous injuries (Nair, Robbs, and 
Muckart, 2000) , the vessels involved were inter- 
nal jugular in 25, subclavian in 15, brachio- 
cephalic in 6, and superior vena cava in 3. 
Injured veins were ligated in 25 patients and 
repaired by lateral suture in 22. No complex 
repairs were performed. There were eight 
perioperative deaths and five cases of transient 
postoperative edema (Makins, 1919). In the 
case of severe bilateral jugular venous injury, 
ligation will carry significant clinical conse- 
quences. In this setting, reconstruction with 
autologous conduit is advisable. 



SUMMARY 

The escalating civilian violence of modern 
times is contributing to increasing frequency 
of vascular injuries in the neck. Early deaths 
are related to airway compromise or exsan- 
guinating hemorrhage. If the patient arrives 
stable to the hospital, an orderly assessment 
and diagnosis may be made and the wounds 
successfully managed. Late deaths are from 
neurodeficits secondary to cerebral hypoxia. 
Advances in the management of penetrating 
vascular injuries on the horizon include non- 
invasive diagnostic testing and nonoperative 
radiologic procedures of endovascular 



chll.qxd 4/16/04 3:36 PH Page 238 



238 



IV • SPECIFIC VASCULAR INJURIES 




■ FIGURE 1 1-8 

A, Lateral view of a patient with a gunshot wound of the neck with active bleeding from the wound. 
The injury to the cervical vertebra is evident. B and C, Angiography reveals injury to the vertebral 
artery. After confirming a complete circle of Willis, this was embolized successfully. (Courtesy T. 
Marrone and J. Tisnado.) ■ 



stenting and grafting. All of this progress is 
directly attributable to the early foundations 
laid down by the Vietnam Vascular Registry. 



REFERENCES 

Asensio JA, Chahwan S, et al: Penetrating 
esophageal injuries: multicenter study of the 
American Association for the Surgery of Trauma. 
J Trauma 2001;50(2) :289-296. 

Asensio JA, Valenziano CP, et al: Management of 
penetrating neck injuries. The controversy 
surrounding zone II injuries. Surg Clin North 
Aml991;71(2):267-296. 



Assali AR, Sdringola S, Moustapha A, etal: Endovas- 
cular repair of traumatic pseudoaneurysm by 
uncovered self-expandable stenting with or 
without trans stent coiling of the aneurysm cavity. 
Cathet Cardiovasc Interv 2001;53:253-258. 

Azuaje RE, Jacobson LE, Glover J, et al: Reliability 
of physical examination as a predictor of vascu- 
lar injury following penetrating neck trauma. 
Paper presented at the 15th Scientific Assembly 
of EAST; Orlando; January 15-19, 2002. 

Bradley EL: Management of penetrating carotid 
artery injuries. An alternative approach. J 
Trauma 1973;13:248-255. 

De Bakey ME, Simeone FA: Battle injuries of arter- 
ies in World War II: an analysis of 2471 cases. 
Ann Surg 1946;123:534-579. 



chll.qxd 4/16/04 3:36 PH Page 239 



11 • PENETRATING CERVICAL VASCULAR INJURY 



239 



Demetriades D, Asensio JA, et al: Complex prob- 
lems in penetrating neck trauma. Surg Clin 
North Am 1996a;76:661-683. 

Demetriades D, Skakkides J, Fofianos C, et al: 
Carotid artery injuries (1989): experience with 
124 cases. J Trauma 1989;29:91-94. 

Demetriades D, Stewart M: Penetrating injuries of 
the neck. Ann R Coll Surg Engl 1985;67:71-74. 

Demetriades D, Theodorou D, et al: Penetrating 
injuries of the neck in patients in stable condi- 
tion. Physical examination, angiography, or 
color flow Doppler imaging. Arch Surg 
1995;130:971-975. 

Demetriades D, Theodorou D.AsensioJ, etal: Man- 
agement options in vertebral artery injuries. Br 
J Surg 1996b;83:83-86. 

Fields CE, Cassano AD, et al: Indirect carotid- 
cavernous sinus fistula after shotgun injury. 
J Trauma 2000;48 (2) :338-341 . 

Flemming D. Case of rupture of the carotid artery 
and wounds of several of its branches treated by 
tying the common trunk of the carotid itself. Med 
ChirJRevl817;3:2. 

Fogelman MJ, Stewart RD: Penetrating wounds of 
the neck. Am J Surg 1956;91:581. 

Friedman D, Flanders A, et al: Vertebral artery 
injury after acute cervical spine trauma: rate of 
occurrence as detected by MR angiography and 
assessment of clinical consequences. AJR Am J 
Roentgenol 1995;164:443-447. 

Fry WR, Dort JA, et al: Duplex scanning replaces 
arteriography and operative exploration in the 
diagnosis of potential cervical vascular injury. Am 
J Surg 1994;168:693-695. 

Golueke PJ, Goldstein AS, etal: Routine versus selec- 
tive exploration of penetrating neck injuries. A 
randomized, prospective study. J Trauma 1984; 
24:1010. 

Harris JP, Anterasian G, et al: Management of 
carotid artery transection resulting from a stab 
wound to the ear. Laryngoscope 1985;95(7, pt 
l):782-785. 

Hirshberg A, Wall MJ, et al: Transcervical gunshot 
injuries. Am J Surg 1994;167(3):309-312. 

Huhges CW: Arterial repair during the Korean War. 
Ann Surg 1958;147:555. 

Kordestani RK, Counelis GJ, et al: Cerebral arter- 
ial spasm after penetrating craniocerebral 
gunshot wounds: transcranial Doppler and 
cerebral blood flow findings. Neurosurgery 
1997;41:351-359. 

Krabbe-Hartkamp MJ, van der Grond J, et al: 
Circle of Willis: morphologic variation on three- 



dimensional time-of- flight MR angiograms. 
Radiology 1998;207:103-111. 

Lawrence KB, Sheffts LM, et al: Wounds of the 
common carotid arteries. Reports of seventeen 
cases from World War II. Am J Surg 1948;76:29. 

LeBlang SD, Nunez DBJr: Helical CT of cervical 
spine and soft tissue injuries of the neck. Radiol 
Clin North Am 1999;37:515-532. 

Ledgerwood AM: Neck: vascular injuries. In: 
Ivatury RR, Cayten CG, eds. The textbook of 
penetrating trauma. Philadelphia: Williams & 
Wilkins, 1996. 

Ledgerwood AM, Mullins RJ, Lucas CE: Primary 
repair vs. ligation for carotid artery injuries. Arch 
Surg 1980;115:488-493. 

LiekwigWGJr, Greenfield LJ: Management of pen- 
etrating carotid artery injury. Ann Surg 
1978;188:587. 

Makins GH: Gunshot injuries to the blood vessels. 
Bristol: John Wright and Sons, Ltd, 1919. 

Marks MW, Argenta LC, et al: Traumatic arteri- 
ovenous malformation of the external carotid 
arterial system. Head Neck Surg 1984;6:1054- 
1058. 

Mittal VK, Paulson TJ, et al: Carotid artery injuries 
and their management. J Cardiovasc Surg 
(Torino) 2000;41:423-431. 

Monson DO, Saletta JD, et al: Carotid vertebral 
trauma. J Trauma 1969;9:987-999. 

Montalvo BM, LeBlang SD, et al: Color Doppler 
sonography in penetrating injuries of the neck. 
AJNR Am J Neuroradiol 1996;17:943-951. 

Munera F, Soto JA, Palacio D, et al: Diagnosis of 
arterial injuries caused by penetrating trauma 
to the neck: comparison of helical CT angiog- 
raphy and conventional angiography. Radiology 
2000;216:356-362. 

Nair R, Robbs JV, Muckart DJ: Management of 
penetrating cervicomediastinal venous trauma. 
Eur J Vase Endovasc Surg 2000;19(l):65-69. 

Ofer A, Nitecki SS, BraunJ, et al: CT angiography 
of the carotid arteries in trauma to the neck. Eur 
J Vase Endovasc Surg 2001;21:401-407. 

Panetta TF, Sales CM, et al: Natural history, duplex 
characteristics, and histopathologic correlation 
of arterial injuries in a canine model. J Vase Surg 
1992;16:867-874. 

Prabhu VC, Patil AA, et al: Magnetic resonance 
angiography in the diagnosis of traumatic ver- 
tebrobasilar complications: a report of two cases. 
Surg Neurol 1994;42:245-248. 

Puchades-Orts A, Nombela-Gomez M, et al: Varia- 
tion in form of circle of Willis: some anatomical 



chll.qxd 4/16/04 3:36 PH Page 240 



240 



IV • SPECIFIC VASCULAR INJURIES 



and embryological considerations. Anat Rec 
1976;185:119-123. 

Rao PM, Ivatury RR, Sharma P, et al: Cervical vas- 
cular injuries: a trauma center experience. 
Surgery 1993;114(3):527-531. 

ReidJD, Weigelt JA: Forty-three cases of vertebral 
artery trauma. J Trauma 1988;28:1007-1012. 

Rich NM, Baugh JH, et al: Acute arterial injuries 
in Vietnam: 1,000 cases. J Trauma 1970; 
10(5):359-369. 

Robless P, Okonko D, et al: Vascular surgical society 
of great Britain and Ireland: platelet function 
during carotid endarterectomy and the 
antiplatelet effect of dextran 40. Br J Surg 
1999;86(5):709. 

Romily RC, Newell DW, Grady MS, et al: Gunshot 
wounds of the internal carotid artery at the skull 
base: management with vein bypass grafts and a 
review of the literature .J Vase Surg 2001 ;33:1001- 
1007. 



Sekharan J, Dennis JW, Veldenz H, et al: Contin- 
ued experience with physical examination alone 
for evaluation and management of penetrating 
zone 2 neck injuries: results of 145 cases. J Vase 
Surg 2000;32:483. 

Thai ER, Snyder WH III, et al: Management of 
carotid artery injuries. Surgery 1974;76:955-962. 

Unger S,Jorgensen J, Hoffman M: Carotid artery 
trauma. Surgery 1990;87:47740. 

Watson WL, Silverstone SM: Ligature of the 
common carotid artery in cancer of the head 
and neck. Ann Surg 1939;109:1. 

Weaver FA, "Vellin AE, et al: The role of arterial 
reconstruction in penetrating carotid artery 
injuries. Arch Surg 1988;123:1106-1111. 

Yee LF, Olcott EW, Knudson MM, et al: Extralu- 
minal, transluminal, and observational treatment 
for vertebral artery injuries. J Trauma 1995; 
39:480-486. 



chl2.qxd 4/16/04 3:37PM Page 241 





Blunt Cervical Vascular Injury 



WALTER L. BIFFL 
ERNEST E. MOORE 
JON M. BURCH 



O ANATOMIC CONSIDERATIONS 

O MECHANISMS OF INJURY 

O PATHOPHYSIOLOGY 

O CLINICAL PRESENTATION 

O SCREENING AND THE INCIDENCE OF BLUNT CERVICAL VASCULAR 
INJURIES 

Screening for Blunt Cerebrovascular Injuries 

Identifying the Patient at Risk 
O DIAGNOSTIC EVALUATION 
O INJURY GRADING 
O TREATMENT AND OUTCOME 
O SUMMARY AND GUIDELINES 



Blunt cervical vascular injuries (BCVIs) , 
those to the extracranial carotid and 
vertebral arteries (VAs), have histori- 
cally been considered rare, yet they are rec- 
ognized as potentially devastating events. 
Given the dearth of experience with BCVIs, 
even in busy trauma centers, there is essen- 
tially no class I literature to guide their man- 
agement. Furthermore, BCVIs present a 
unique set of challenges because (1) they often 
occur in the setting of multisystem trauma, 



particularly head injuries, and symptoms may 
be masked by depressed consciousness or 
attributed to intracranial injury; (2) they are 
typically not diagnosed until after cerebral 
ischemic injury, making it difficult to achieve 
a good outcome; (3) they may occur follow- 
ing relatively minor neck "trauma," and so 
even with a high index of awareness of BCVIs, 
patients might not be considered at risk until 
they manifest cerebral ischemia; (4) the only 
reliable diagnostic test is invasive, resource 

241 



chl2.qxd 4/16/04 3:37PM Page 242 



242 



IV • SPECIFIC VASCULAR INJURIES 



intensive, and associated with its own risks, 
and alternative noninvasive diagnostic tests 
may miss early subtle lesions; (5) the natural 
history of various injury types is unknown, and 
thus, it must be presumed that all injuries 
should be treated; and (6) treatments are 
potentially risky, there is no consensus on the 
optimal treatment for various lesions, and effi- 
cacy of treatment is largely unproven. In sum, 
BCVIs present dilemmas in risk assessment, 
screening, diagnosis, and treatment. 



ANATOMIC CONSIDERATIONS 

The left common carotid artery (CCA) orig- 
inates from the aortic arch within the thorax, 
whereas the right CCA is a terminal branch 
of the innominate artery behind the stern- 
oclavicular joint. There are no significant 
arterial branches from the CCA. It generally 
divides into the internal carotid artery (ICA) 
and external carotid artery (ECA) at the level 
of the C3-C4 disc space, corresponding to the 
superior border of the thyroid cartilage. The 
ECA does not directly supply circulation to 
the brain; thus, traumatic injuries to the ECA 
are usually well tolerated neurologically unless 
there is preexisting cerebrovascular disease. 
On the other hand, in the presence of carotid 
or VA occlusive disease, the ECA branches may 
provide critical collateral flow. The ICA can 
be separated anatomically into four segments: 
cervical, petrous, cavernous, and cerebral 
(supraclinoid) . The cervical portion has no 
named branches because it ascends ventral to 
the transverse processes of the C1-C3 verte- 
bral bodies (a relationship that is pivotal 
in the pathophysiology of many injuries, as 
described later in this chapter) . The petrous 
segment traverses the carotid canal in the 
petrous portion of the temporal bone; here, 
it is at risk of laceration in the setting of a basilar 
skull fracture. The cavernous portion (also 
called the carotid siphon because of its gentle 
S shape) is the first part of the ICA within the 
cranial vault. It is suspended between the layers 
of the dura matter that form the cavernous 
sinus. At the anterior clinoid process, the ICA 
perforates the dura and becomes the supra- 
clinoid, or cerebral, segment. The ICA divides 



terminally into the anterior and middle 
cerebral arteries. 

The VAs originate from the subclavian arter- 
ies, enter the cervical vertebral foramina at 
the level of C6, exit the transverse foramen of 
C2, and merge intradurally to form the basilar 
artery. There is considerable asymmetry to the 
point of agenesis (2% of right and 3% of left 
VAs) . The circle of Willis connects the ante- 
rior and posterior circulation but is intact and 
symmetric injust 20% of individuals. The fre- 
quency of variations in collateral circulatory 
routes may explain unusual clinical presen- 
tations of arterial injuries and underscores the 
need to image the entire cerebral circulation 
in cases of BCVI. 



MECHANISMS OF INJURY 

There are four fundamental mechanisms of 
carotid injuries. The most common is associ- 
ated with hyperextension and rotation of the 
head and neck. The lateral articular processes 
and pedicles of the upper three cervical ver- 
tebrae (C1-C3) project more anteriorly than 
those of C4-C7; thus, the overlying distal cer- 
vical ICA is prone to stretch injury during 
cervical hyperextension. Rotation at the 
atlantoaxial joint may result in anterior move- 
ment of the contralateral CI lateral mass, 
further exacerbating the stretch (Fig. 12-1). 
A direct blow to the neck may crush the artery, 
or it may be compressed between the mandible 
and the vertebral prominences in acute cer- 
vical hyperflexion injuries. Intraoral trauma 
may injure the ICA, typically seen in children 
who have fallen with a hard object (such as a 
pencil) in their mouth. Finally, basilar skull 
fractures that involve the sphenoid or petrous 
bones may result in laceration of the artery. 

The third segment of the VA, which extends 
from the level of C2 to the dura, is most com- 
monly injured by blunt trauma because of the 
increased degree of stretching and compres- 
sion (Fig. 12-2) ; the relationship between the 
VA and the cervical bodies puts the VA at risk 
when the vertebral body — particularly the 
foramen transversarium — is fractured. 

Of note, numerous case reports in the 
literature have documented BCVI following 



ohl2.qxd 4/16/04 3:37PM Page 243 



12 • BLUNT CERVICAL VASCULAR INJURY 



243 




"trivial trauma." These include virtually any 
athletic endeavor, chiropractic manipulation, 
visiting the hairdresser, "head banging" to 
music, "bottoms-up" drinking, rapid head 
turning, and everyday activities such as cough- 
ing, shaving, vomiting, nose blowing, and 
scolding a child. This is in contradistinction 
to "spontaneous" carotid dissection, which by 
definition occurs in the absence of trauma. 
Reported risk factors for spontaneous dis- 
section include hypertension, Marfan's 
syndrome, fibromuscular dysplasia, syphilis, 
arteriopathies, and Erdheim's cystic medial 
necrosis. It is the contention of some that truly 
spontaneous dissections are rare, but that such 
risk factors simply predispose patients to 
BCVI following trivial trauma. The absence 
of trauma from an individual's history does 
not exclude it as an etiology, because patients 
often consider events too insignificant (or 
embarrassing) to relate. 




PATHOPHYSIOLOGY 



■ FIGURE 1 2-1 

Rotation at the atlantoaxial joint may result 
in anterior movement of the contralateral 
C1 lateral mass, further exacerbating the 
stretch. ■ 



Regardless of the underlying mechanism of 
injury, the final common pathway of BCVI in 
most cases is intimal disruption. This exposes 
thrombogenic subendothelial collagen, pro- 
moting platelet aggregation with subsequent 
embolization, partial thrombosis with low flow, 
or complete thrombosis. In addition, the 
intimal tear offers a portal of egress for a 
dissecting column of blood. Dissection may 
result in progressive luminal narrowing and 
subsequent occlusion. Whether caused by 
thromboembolism or occlusion, the end 
result, particularly in the setting of multisystem 
trauma with hypotension, is cerebral ischemia. 
Less commonly, partial or complete transec- 
tion of the artery occurs, resulting in pseudoa- 
neurysm formation or free rupture. The 
former may increase in size to compress and 
occlude the vessel lumen; it may be the source 
of platelet thromboembolism; or it may 
rupture. Rupture may result in hemorrhage 
or arteriovenous fistula formation. 



ohl2.qxd 4/16/04 3:37PM Page 244 



244 IV • SPECIFIC VASCULAR INJURIES 




■ FIGURE 1 2-2 

The third segment of the vertebral artery, which extends from the C2 level to the dura, is most 
commonly injured by blunt trauma because of the increased degree of stretching, which occurs at 
the atlantoaxial and atlanto-occipital joints during head rotation. ■ 



CLINICAL PRESENTATION 

The clinical manifestations of BCVI depend 
on the type of injury, the involved artery, and 
collateral circulation. Premonitory signs and 
symptoms associated with the vessel injury may 
suggest the presence of BCVI before mani- 
festations of cerebral ischemia. Pain (neck, 
ear, face, or periorbital) can be present in up 
to 60% of patients and is believed to reflect 
mural hemorrhage or dissection of the vessel 
wall. Complaints of such pain are often diffi- 
cult to elicit in the multi-injured patient and 
may be attributed to other injuries; however, 
BCVI must be considered in the differential 
diagnosis of post-traumatic neck pain and 
headache. Horner's syndrome or oculosym- 
pathetic paresis (partial Horner's syndrome) 
may result from disruption of the periarter- 
ial sympathetic plexus. Pupillary asymmetry 
can have several etiologies in the injured 
patient. However, if the larger of the pupils is 
reactive and the smaller pupil is not, carotid 
injury should be suspected on the side of the 
smaller pupil. 

Systematic neurologic examination will 
help localize the distribution of cerebral 
ischemia; however, cerebral ischemic signs or 
symptoms may be absent in the acute setting, 



because of (1) the presence of collateral cir- 
culatory pathways and (2) a characteristic 
latent period between the time of injury and 
the appearance of clinical manifestations. 
Unless the vessel is immediately occluded, time 
is required for a platelet plug to form and 
either limit flow or embolize. In various 
series, 23% to 50% of patients first developed 
signs or symptoms of BCVI more than 12 hours 
after the traumatic event. In our experience, 
42% of symptomatic patients manifested 
more than 18 hours after injury and two exhib- 
ited symptoms 7 days later. Delayed recogni- 
tion may also occur in the face of multisystem 
trauma, with critical injuries demanding 
immediate attention, or head injury, which 
may preclude a meaningful neurologic exam- 
ination. To make the early diagnosis of BCVI, 
the surgeon must recognize the signs and 
symptoms in a trauma patient. These include 
(1) hemorrhage — from mouth, nose, ears 
or wound — of potential arterial origin; (2) 
expanding cervical hematoma; (3) cervical 
bruit in a patient 50 years old or younger; (4) 
evidence of cerebral infarction on computed 
tomographic (CT) scan; or (5) unexplained 
or incongruous central or lateralizing neu- 
rologic deficit, transient ischemic attack (TIA) , 
amaurosis fugax, or Horner's syndrome. 



chl2.qxd 4/16/04 3:37PM Page 245 



12 • BLUNT CERVICAL VASCULAR INJURY 



245 



SCREENING AND THE 
INCIDENCE OF BLUNT 
CEREBROVASCULAR INJURIES 

The incidence of BCVI is difficult to quantify 
because many remain asymptomatic or 
symptoms may be attributed to associated 
brain (or other) injury. However, the inci- 
dence of BCVI among blunt trauma victims 
seems to be increasing. Early multicenter 
reviews identified an incidence of BCVI of 
0.08% to 0.17% among patients admitted to 
trauma centers following blunt trauma, but 
more recent series have reported incidences 
of 0.24% to 0.44%. The argument that the 
incidence actually is increasing is supported 
by the fact that nearly all the patients in the 
series published through 1997 were sympto- 
matic at the time of diagnosis. A number of 
factors could account for this explosion of 
BCVI at our center, including (1) higher 
highway speed limits in Colorado, (2) more 
widespread use of shoulder restraints and 
airbags, and (3) our role as a regional trauma 
center in the Statewide Trauma System, with 
increasing numbers of individuals being trans- 
ferred to us following major mechanism 
injuries. However, without question, screen- 
ing has identified injuries that would other- 
wise have been overlooked. In fact, two-thirds 
of patients diagnosed with BCVI at our center 
in the 1990s were asymptomatic. 

Screening for Blunt 
Cerebrovascular Injuries 

In 1996, Fabian and colleagues suggested that 
blunt carotid injuries (BCI) were being under- 
diagnosed. This had been suspected based on 
the Western Trauma Association multicenter 
study and demonstrated in a preliminary 
prospective study of screening at our center. 
Recognizing the potential to improve neuro- 
logic outcome by identifying and treating 
carotid injuries before occurrence of cerebral 
ischemia, we instituted an aggressive policy of 
screening and recently reported an epidemic 
of BCI. Between January 1990 and June 1996, 
before screening, our incidence of BCI was 
0.1% of blunt trauma admissions — similar to 



multicenter reports. During 4.5 years with a 
formal screening protocol, the incidence of 
BCVI has approached 1% of all blunt trauma 
admission to our center. In Memphis, a high 
index of suspicion and increasingly liberal 
screening resulted in an incidence of carotid 
injuries of 0.5%. 



Identifying the Patient at Risk 

Although the Louisville group has asserted 
that BCVI cannot be predicted based on 
clinical grounds, a number of groups have 
reported higher incidences of BCVI when 
diagnostic testing is employed for specific 
injury patterns and mechanisms. We formu- 
lated our screening criteria based on a knowl- 
edge of injury mechanisms and anatomic 
considerations. The screening criteria include 
(1) an injury mechanism compatible with 
several cervical hyperextension/rotation or 
hyperflexion, particularly if associated with dis- 
placed or complex midface or mandibular 
fracture, or closed head injury consistentwith 
diffuse axonal injury of the brain; (2) near 
hanging resulting in cerebral anoxia; (3) seat- 
belt abrasion or other soft tissue injury of the 
anterior neck resulting in significant cervical 
swelling or altered mental status; (4) basilar 
skull fracture involving the carotid canal: 
and (5) cervical vertebral body fracture or 
distraction injury, excluding isolated spinous 
process fracture. This widespread screening 
approach requires a tremendous commitment 
of resources. In an attempt to allocate 
resources more effectively, we critically eval- 
uated our screening criteria, analyzing the 
injury mechanisms and patterns of all the 
patients who underwent arteriography to 
exclude BCVI over a 9-year period, to iden- 
tify independent predictors of BCVI. By mul- 
tivariate analysis, Glasgow Coma Scale score 
less than 6, petrous bone fracture, diffuse 
axonal brain injury, and Le Fort II or II frac- 
ture were identified as risk factors for carotid 
injuries. The only independent predictor of 
VA injury was cervical spine injury. In 4.5 years 
of screening, we have performed screening 
cerebral arteriography on 390 patients; of 
these, 131 (33%) have had BCVI. More than 
two thirds were asymptomatic at diagnosis. On 



chl2.qxd 4/16/04 3:37PM Page 246 



246 



IV • SPECIFIC VASCULAR INJURIES 



the other hand, a handful of patients have 
been transferred from remote facilities who 
became symptomatic from BCVI following 
"trivial trauma," who would not have met cri- 
teria for screening. We believe these criteria 
represent a starting point; however, the ques- 
tion of optimal criteria will ultimately require 
a multicenter collaborative effort. 



DIAGNOSTIC EVALUATION 

The discovery of signs or symptoms sugges- 
tive of BCVI mandates emergent diagnostic 
evaluation. The gold standard for diagnosis 
of BCVI is four-vessel biplanar cerebral arte- 
riography. Unfortunately, it is invasive and 
resource intensive. Its risks include compli- 
cations related to catheter insertion (1% to 
2% hematoma, potential arterial pseudoa- 
neurysm), contrast administration (1% to 2% 
renal dysfunction, potential allergic reac- 
tion), and stroke (<1%). Noninvasive diag- 
nostic alternatives are available for screening 
asymptomatic patients for BCVI; however, one 
must recognize that diagnostic sensitivity is 
compromised in avoiding invasive testing. 
Duplex ultrasonography is widely considered 
the modality of choice for imaging the carotid 
arteries; however, experience in diagnosing 
BCVI is limited. In the Western Trauma Asso- 
ciation multicenter review, duplex scanning 
had 86% sensitivity for ICA injury. In that 
series, the lesions missed by duplex were 
located at the base of the skull. Because most 
ICA injuries involve the artery at or near the 
base of the skull, this is a major potential weak- 
ness. Furthermore, although duplex scanning 
can provide indirect evidence of injuries by 
detecting turbulence and other flow distur- 
bances, these findings are not reliable in the 
presence of stenoses of less than 60%. Unfor- 
tunately, we have witnessed the potential for 
innocuous-appearing luminal irregularities to 
cause devastating cerebrovascular accidents 
and thus do not believe duplex scanning is 
adequate for BCVI screening. CT angiogra- 
phy (CTA) is attractive because most multi- 
system trauma patients have indications for 
CT scanning. However, our experience has 
shown that CTA has a sensitivity no better than 



that of duplex ultrasonography. To image the 
cerebral vessels in their entirety with a slice 
thickness and pitch adequate for sufficiently 
sensitive reconstruction is not practical; in 
addition, bony artifact is in the carotid canal, 
potentially obscuring injuries. Of all the non- 
invasive screening modalities, magnetic reso- 
nance angiography (MRA) holds the greatest 
promise to reliably supplant cerebral arteri- 
ography. Advantages of MRA include the capa- 
bility to simultaneously image the remainder 
of the head and neck and detect cerebral 
infraction earlier than CT scanning while 
avoiding contrast administration. Major 
impediments include a lack of timely avail- 
ability at many institutions and incompatibil- 
ity of ventilatory and orthopedic fixation 
equipment with the magnet. Recent prospec- 
tive trials have reported suboptimal accuracy 
of MRA as well as CTA. Until more rigorous 
evaluation, arteriography remains the gold 
standard. 



INJURY GRADING 



Carotid artery and VA injuries are a hetero- 
geneous mix of lesions. Several groups have 
suggested that different types of injuries 
might be managed differently; the absence 
of a formal BCVI grading scale, however, 
has been a major impediment to formulating 
sound practice guidelines. We hypothesized 
that different injury types had distinct impli- 
cations in terms of response to therapy and 
ultimate neurologic outcome. Thus, we devel- 
oped a grading scale based on the literature 
and our collective experience with 109 carotid 
injuries (Table 12-1 ) . We reported that stroke 
incidence increased with injury grade. In con- 
trast, recent analysis of our experience with 
VA injuries revealed no similar correlation 
between injury grade and posterior circula- 
tion stroke (Table 12-2). 



TREATMENT AND OUTCOME 



The optimal management of BCVI remains 
controversial. The three primary choices for 



chl2.qxd 4/16/04 3:37PM Page 247 



12 • BLUNT CERVICAL VASCULAR INJURY 



247 



TABLE 12-1 

BLUNT CAROTID AND VERTEBRAL ARTERY INJURY GRADING SCALE 



Injury Grade 

I 



IV 
V 



Description 

Luminal irregularity or dissection with <25% 

luminal narrowing 
Dissection or intramural hematoma with >25% 

luminal narrowing, intraluminal thrombus, or 

raised intimal flap 
Pseudoaneurysm 
Occlusion 
Transection with free extravasation 





AIS90 




ICD-9* 


Intracranial 


Cervicar 


900.03 


3 


3 


900.3 


3 


3 


900.03 
900.03 

900.03 


3 
4 

5 


3 
3 

4 



'Internal carotid artery injury; ICD-9 code for common carotid artery injury is 900.01. 

+ Add 1 if neurologic deficit (stroke) is not related to head injury. 

AIS-90, Abbreviated Injury Scale, 1990 revision; ICD-9, International Classification of Diseases, 9th revision. 



TABLE 12-2 

STROKE RATE AND MORTALITY OF BLUNT CAROTID AND VERTEBRAL ARTERY 
INJURIES, STRATIFIED BY INJURY GRADE 







BCAI (%) 




BVAI (%) 


Worst Injury Grade 


Stroke 


Mortality 


Stroke 


Mortality 



II 
III 

IV 

V 



3 
11 
33 
44 

100 



11 
11 
11 
22 

100 



19 
40 
13 
33 



31 


13 
11 



BCAI, blunt carotid artery injury; BVAI, blunt vertebral artery injury. 



management include observation, surgical 
therapy, and nonsurgical therapy (e.g., anti- 
coagulation and endovascular techniques) . In 
determining the treatment for an individual, 
one must consider the location and grade of 
the injury, as well as symptomatology. 

Observation cannot be considered optimal 
therapy, given the natural history of sympto- 
matic BCVI; early reports established mor- 
bidity and mortality rates of 58% and 28%, 
respectively. Extrapolating from penetrating 
trauma literature, wherein neurologic mor- 
bidity and mortality were better in those under- 
going operation, early series recommended 
surgery in the absence of completed hemi- 
plegic deficits. However, most blunt injuries 
involve the ICA at or above the base of the 



skull. Thus, inaccessibility precludes direct sur- 
gical repair. Extracranial-intracranial bypass 
has been successfully employed in select 
patients, but this remains a controversial 
concept. 

In 1996, Fabian and colleagues reported a 
large single-institution experience with carotid 
artery injuries. Anticoagulation improved 
neurologic outcome of patients presenting 
with minor and major neurologic deficits. In 
fact, logistic regression analysis identified 
heparin as the only factor independently asso- 
ciated with improved neurologic outcome. We 
similarly found that symptomatic patients who 
are anticoagulated showed a trend toward 
greater neurologic improvement at the time 
of discharge compared with those who were 



chl2.qxd 4/16/04 3:37PM Page 248 



248 



IV • SPECIFIC VASCULAR INJURIES 



not anticoagulated (Table 12-3). An impor- 
tant finding of our series was that identifica- 
tion and treatment of carotid injuries before 
the onset of symptoms appear critical in 
improving neurologic outcome. In our analy- 
sis of outcomes following VAinjuries, we again 
found that heparin improves neurologic out- 
comes (Table 12-4). Neurologic outcomes 
were better in the group as a whole, as well as 
in the subgroup suffering stroke. In addition, 
anticoagulation resulted in favorable trends 
including (1) preventing progression of 
lesions to higher injury grades (Table 12-5); 
(2) preventing neurologic deterioration from 
diagnosis to discharge; and (3) preventing 
stroke. 

Obviously, bleeding complications are a 
concern in patients with multisystem injuries. 
We have experienced a 10% incidence of 
bleeding complications with our anticoagu- 
lation protocol. Presently, we are exploring 



an alternative in lower risk patients. Specifi- 
cally, we are prospectively comparing the safety 
and efficacy of antiplatelet therapy versus sys- 
temic heparinization in the treatment of 
asymptomatic grade I BCVI (intimal irregu- 
larity without luminal stenosis, intraluminal 
thrombus, or a visible intimal flap). 

Deployment of endovascular stents is 
gaining increasing favor in the treatment of 
vascular lesions. We deploy stents to treat per- 
sistent traumatic pseudoaneurysms (grade III 
BCVI) , in an attempt to tack down the intima 
and exclude the pseudoaneurysm from the 
circulation. In addition, we stent grade II 
stenoses that threaten to occlude the vessel. 
Given the risk of stroke during manipulation 
of devices in an acutely injured artery, we rec- 
ommend waiting 7 days, if possible, before 
attempting stent placement. Until more data 
are available, we recommend full anticoagu- 
lation after stenting for BCVI. It must be 



TABLE 12-3 

NEUROLOGIC OUTCOME OF BLUNT CAROTID ARTERY INJURIES, STRATIFIED BY 
TREATMENT; COMBINED EXPERIENCE FROM DENVER AND MEMPHIS 



Outcome 

Neurologic improvement, diagnosis to discharge 
Neurologic deterioration, diagnosis to discharge 
Good neurologic outcome 
Poor neurologic outcome 





No 




Systemic 


Systemic 




Heparin (%) 


Heparin (%) 


PValu 


49 


19 


<.05 


5 


24 


<c.05 


50 


30 


<.05 


30 


60 


<.05 



TABLE 12-4 

NEUROLOGIC OUTCOME OF BLUNT VERTEBRAL ARTERY INJURIES, STRATIFIED 
BY TREATMENT 



Outcome 



Poor neurologic function (all BVAI patients) 

Poor neurologic function (stroke victims) 

Progression of injury grade 

Neurologic deterioration, diagnosis to discharge 

Stroke 



Systemic 
Heparin (%) 

6 
17 
25 
19 

14 



No 

Systemic 

Heparin (%) 

60 
100 
60 
60 
35 



P Value 

<.05 
<.05 

.18 

.11 

.13 



BVAI, blunt vertebral artery injury. 



chl2.qxd 4/16/04 3:37PM Page 249 



12 • BLUNT CERVICAL VASCULAR INJURY 



249 



TABLE 12-5 

ARTERIOGRAPHY OUTCOME OF GRADED CAROTID ARTERIAL LESIONS 



Initial 
Injury Grade 



Treatment 



Healed (5) 



Final Injury Grade (%) 



IV 



IV 



Heparin 


16(70) 


6(26) 


— 


1(4) 


— 


No heparin 


12 (67) 


4(22) 


1(6) 


1(6) 


— 


Heparin 


1(10) 


1(10) 


1(10) 


6(60) 


1(10) 


No heparin 


— 


— 


— 


— 


— 


Heparin 


1(8) 


— 


— 


1 1 (85) 


1(8) 


No heparin 


— 


— 


— 


3(100) 


— 


Heparin 


— 


— 


— 


— 


1 (100) 


No heparin 


— 


— 


— 


— 


1 (100) 



emphasized that these devices are not 
approved for these indications, and their use 
should be restricted to research protocols. 

Angiographic embolization has been 
employed widely for maxillofacial arterial 
injuries, but ICA embolization has not been 
supported because of concern of brain in- 
farction. Our experience with attempted 
embolization in the ICA distribution has been 
confined to grade V injuries; in each instance, 
embolization has proven futile. On the other 
hand, angiographic embolization has been 
promoted as an alternative to surgical ligation 
in the management ofVAinjuries. Experience 
with this technique is still limited. Another 
alternative to surgical ligation of the VA is 
endovascular balloon occlusion. 



SUMMARY AND GUIDELINES 



BCVIs are infrequently diagnosed but may be 
overlooked in many patients. Symptoms may 
be masked by central nervous system injuries, 
but the large majority are asymptomatic at the 
time of presentation. Early diagnosis and insti- 
tution of treatment appear to improve out- 
comes. Thus, the following guidelines have 
been adopted by our center. 

Emergent cerebral arteriography should be 
performed to exclude BCVI in the presence 
of the following signs or symptoms: (1) 
hemorrhage — from mouth, nose, ears, or 
wounds — of potential arterial origin; (2) 



expanding cervical hematoma; (3) cervical 
bruit in a patient younger than 50 years; (4) 
evidence of cerebral infarction on CT scan; 
(5) unexplained or incongruous central or 
lateralizing neurologic deficit, TIA, amauro- 
sis fugax, or Horner's syndrome. 

Consideration should be given to screen- 
ing individuals with injury mechanisms or 
patterns consistent with BCVI. These include 
(1) an injury mechanism compatible with 
severe cervical hyperextension/rotation or 
hyperflexion, particularly if associated with dis- 
placed or complex midface or mandibular 
fracture, or closed head injury consistentwith 
diffuse axonal injury of the brain; (2) near- 
hanging resulting in cerebral anoxia; (3) seat- 
belt abrasion or other soft tissue injury of the 
anterior neck resulting in significant cervical 
swelling or altered mental status; (4) basilar 
skull fracture involving the carotid canal; and 
(5) cervical vertebral body fracture or dis- 
traction injury, excluding isolated spinous 
process fracture. 

Our current diagnostic standard remains 
four-vessel cervical arteriography. All BCVIs 
are treated. Surgically accessible grade II, III, 
IV, and V injuries are repaired. Symptomatic 
patients with BCVI should receive some form 
of antithrombotic therapy, unless absolutely 
contraindicated by central nervous system 
injury. In asymptomatic patients with cerebral 
intraparenchymal hemorrhage or fractures in 
regions with the potential to develop an under- 
lying epidural hematoma, anticoagulation is 
not initiated until follow-up CT scan in 24 



chl2.qxd 4/16/04 3:37PM Page 250 



250 



IV • SPECIFIC VASCULAR INJURIES 



hours excludes a significant change of the 
lesion. If there has been progression of the 
brain injury, anticoagulation is held until CT 
scans are stable at 24-hour intervals. Anti- 
coagulation is not withheld for punctate 
intraparenchymal, small subarachnoid, or 
intraventricular hemorrhages. Our anticoag- 
ulation protocol is to begin a heparin infu- 
sion at 15U/kg/hr, without an initial bolus 
dose. The partial thromboplastin time (PTT) 
is measured 6 hours after therapy is started, 
and the infusion rate is adjusted to maintain 
the PTT at 40 to 50 seconds. Follow-up arte- 
riography is performed 7 to 10 days after 
injury. Healing of the injury allows discon- 
tinuation of therapy, whereas persistence 
warrants 3 months of warfarin (Coumadin) 
therapy. Progression of the lesion prompts 
alteration in therapy, including endovascular 
stent placement or a change in the anticoag- 
ulant regimen, as well as additional follow-up 
imaging. Patients undergo arteriography 
again after 3 months, to determine the need 
for further treatment. 



REFERENCES 

Biffl WL, Moore EE, Ellicott JP, et al: The devas- 
tating potential of blunt vertebral arterial 
injuries. Ann Surg 2000;231:672-681. 

Biffl WL, Moore EE, Offner PJ, et al: Optimizing 
screening for blunt cerebrovascular injuries. Am 
J Surg 1999;178:517-522. 



Biffl WL, Moore EE, Offner PJ, et al: Blunt carotid 
arterial injuries: Implications of a new grading 
scale. J Trauma 1999;47:845-853. 

Biffl WL, Moore EE, Ryu RK, et al: The unrecog- 
nized epidemic of blunt carotid arterial injuries: 
Early diagnosis improves neurologic outcome. 
Ann Surg 1998;228:462-470. 

Biffl WL, Ray, CE, Moore EE, et al: Noninvasive 
diagnosis of blunt cerebrovascular injuries: A pre- 
liminary report. J Trauma 2002;53:850-856. 

Carrillo EH, Osborne DL, Spain DA, et al: Blunt 
carotid artery injuries: Difficulties with the diag- 
nosis prior to neurologic event. J Trauma 
1999;46:1120-1125. 

Eachempati SR, Vaslef SN, Sebastian MW, Reed RL 
II: Blunt vascular injuries of the head and neck: 
Is heparinization necessary? J Trauma 1998; 
45:997-1004. 

Fabian TC, Patton JH Jr, Croce MA ,et al: Blunt 
carotid injury: Importance of early diagnosis and 
anticoagulant therapy. Ann Surg 1996;223:513- 
525. 

Giacobetti FB, Vaccaro AR, Bos-Giacobetti MA, et 
al: Vertebral artery occlusion associated with cer- 
vical spine trauma: A prospective analysis. Spine 
1997;22:188-192. 

Miller PR, Fabian TC, Croce MA, et al: Prospective 
screeing for blunt cerebrovascular injuries: 
Analysis of diagnostic modalities and outcomes. 
Ann Surg 2002;236:386-395. 

Rogers FB, Baker EF, Osier TM, et al: Computed 
tomographic angiography as a screening modal- 
ity for blunt cervical arterial injuries: Prelimi- 
nary results. J Trauma 1999;46:380-385. 



chl3.qxd 4/16/04 3:41PM Page 251 




Penetrating Thoracic 
Vascular Injury 



SCOTT A. LEMAIRE 
LORI D. CONKLIN 
MATTHEW J. WALL, JR. 



INITIAL EVALUATION AND MANAGEMENT 
Prehospital Management 

EMERGENCY CENTER EVALUATION AND MANAGEMENT 
Primary Survey and Resuscitation 

History 

Initial examination 

Intravenous access and fluid administration 

Tube thoracostomy 

Pericardiocentesis 

Emergency center thoracotomy 
Secondary Survey 
DIAGNOSTIC STUDIES 
Catheter Arteriography 
Computed Tomography 
TREATMENT OPTIONS 
Endovascular Stenting 
Surgical Repair 

Preoperative considerations 

Damage control 



251 



chl3.qxd 4/16/04 3:41PM Page 252 



252 



IV • SPECIFIC VASCULAR INJURIES 



SPECIFIC INJURIES 
Thoracic Inlet 

Subclavian artery and vein 

Innominate artery and vein 

Left common carotid artery 
Thoracic Aorta 

Ascending aorta 

Transverse aortic arch 

Descending thoracic aorta 
Other Major Intrathoracic Vessels 

Pulmonary artery and vein 

Thoracic vena cava 

Azygos vein 

Internal thoracic and intercostal arteries 
SPECIAL PROBLEMS 
Mediastinal Traverse Injuries 
Thoracic Duct Injury 
Systemic Air Embolism 
Foreign Body Embolism 
POSTOPERATIVE MANAGEMENT 



The lethality of penetrating chest 
wounds has been well recognized 
throughout history. More than 90% of 
the penetrating thoracic wounds described in 
Homer's Iliad and Virgil's Aeneid were fatal. 
The first successful repair of a penetrating tho- 
racic vascular injury did not occur until 
October 1913, when a 30-year-old Russian 
surgeon named Yustin Djanelidze closed an 
8-mm stab wound to the ascending aorta with 
three interrupted sutures. 

Currently, nearly 4% of patients with pen- 
etrating chest wounds have an injury involv- 
ing the thoracic great vessels: the aorta and 
its brachiocephalic branches, the pulmonary 
arteries and veins, the superior and intratho- 
racic inferior venae cavae, and the innomi- 
nate and azygos veins. The incidence of great 
vessel injury is substantially higher following 
gunshotwounds (5%) than after stab wounds 
(2%). 



INITIAL EVALUATION AND 
MANAGEMENT 

Prehospital Management 

Patients sustaining penetrating thoracic 
trauma should be immediately transported to 
the nearest trauma center capable of manag- 
ing thoracic vascular injuries. Intravenous 
access should be avoided in the upper extrem- 
ities, particularly on the side of injury, because 
the central venous structures may be tran- 
sected or thrombosed. Allowing mild hypoten- 
sion is preferable to aggressive attempts to 
increase blood pressure with fluid boluses, 
military antishock trousers (MAST suits) , or 
pressors. Even transient increases in blood 
pressure may dislodge a soft clot and increase 
bleeding. In a randomized trial of patients 
with penetrating truncal trauma, Bickell and 



chl3.qxd 4/16/04 3:41PM Page 253 



13 • PENETRATING THORACIC VASCULAR INJURY 



253 



colleagues (1994) compared standard fluid 
resuscitation with no preoperative fluid resus- 
citation and demonstrated a significant sur- 
vival advantage in patients who had delayed 
resuscitation. Similarly, the use of MAST suits 
in hypotensive patients sustaining penetrat- 
ing thoracic trauma is associated with 
increased mortality. These pneumatic com- 
pression devices elevate blood pressure by 
increasing afterload and are equivalent to 
placing a cross clamp distal to avascular injury, 
a clearly counterproductive maneuver. 



EMERGENCY CENTER 
EVALUATION AND 
MANAGEMENT 



Patient presentation can range from minimal 
symptoms to cardiac arrest necessitating resus- 
citative thoracotomy. Penetrating vascular 
injuries may produce intraluminal intimal 
flaps or thrombosis, arteriovenous fistulas, and 
pseudoaneurysms. The resulting clinical 
manifestations include external bleeding, 
hemothorax, cardiac tamponade, medias- 
tinal hematoma, stroke, and limb ischemia. 
The diagnosis and treatment of the life- 
threatening manifestations occur con- 
comitantly during the primary survey and 
resuscitation phase. 



Primary Survey and 
Resuscitation 



of hemorrhage at the accident scene and 
hemodynamic instability during transport. 



INITIAL EXAMINATION 

The primary survey addresses the most life- 
threatening manifestations of intrathoracic 
vascular injury by including focused attention 
to airway obstruction, massive hemothorax, 
and pericardial tamponade. As always, estab- 
lishing satisfactory airway, breathing, and cer- 
vical spine protection are the first concerns. 
An expanding upper mediastinal hematoma 
can cause stridor due to airway compression. 
The subsequent circulation assessment 
increases the focus on potential thoracic vas- 
cular injuries. Hypotension immediately raises 
the suspicion for a great vessel injury; the crit- 
ical distinction is whether the hypotension is 
due to hypovolemia or tamponade from an 
intrapericardial injury. Tracheal deviation 
away from the side of injury may indicate medi- 
astinal shift due to a massive hemothorax. 
Among the classic signs of tamponade (e.g., 
distended neck veins, pulsus paradoxus 
exceeding lOmmHg, and muffled heart 
sounds) , venous engorgement is an important 
early sign of pericardial tamponade. Tam- 
ponade should be considered in the setting 
of progressive hypotension without evidence 
of ongoing blood loss. In many trauma centers, 
immediate ultrasonography can be performed 
at the bedside in the emergency department 
to rapidly determine whether a hemoperi- 
cardium is present; this is a standard compo- 
nent of the Focused Abdominal Sonography 
for Trauma examination. 



HISTORY 



The history may provide the first clues sug- 
gesting a thoracic vascular injury. Information 
regarding the length of a knife, the firearm 
type and number of rounds fired, and the 
patient's distance from the firearm — though 
not always reliable — is important to obtain 
from the patient or accompanying persons. 
In addition to information involving the 
mechanism of injury, the emergency transport 
personnel can provide medical information 
important in evaluating the potential for a tho- 
racic great vessel injury, such as the amount 



INTRAVENOUS ACCESS AND 
FLUID ADMINISTRATION 

As a general rule, patients with suspected 
injuries to the major thoracic venous branches 
should have large-bore intravenous access 
established in the lower extremities whenever 
possible. The saphenous vein can be cannu- 
lated percutaneously or via saphenous vein 
cutdown at the ankle or in the groin; when 
placing the catheter through a cutdown, 
sterile intravenous extension tubing can be 



chl3.qxd 4/16/04 3:41PM Page 254 



254 



IV • SPECIFIC VASCULAR INJURIES 



inserted directly in the vein. If an upper 
extremity or subclavian venous catheter is 
required in a patient with a potential subcla- 
vian vascular injury, the contralateral side 
should be used for cannulation. 

The treatment of severe shock should 
include blood transfusion. However, in 
patients with mild hypotension, rapid infu- 
sions of either blood or crystalloid should be 
avoided before operation because they may 
increase the blood pressure to a point that a 
protective soft perivascular clot is "blown out" 
and fatal hemorrhage ensues. 



TUBE THORACOSTOMY 

By evacuating the initial hemothorax, place- 
ment of an appropriately sized chest tube 
(Table 13-1) restores effective breathing and 
allows an assessment regarding ongoing hem- 
orrhage. The tube is usually placed in the 
fourth or fifth intercostal space (near nipple 
level) at the anterior to mid-axillary line. 
Intrapleural blood loss that results in hypoten- 
sion is termed massive hemothorax. If a massive 
hemothorax is suspected, based either on clin- 
ical findings or on the chest radiograph find- 
ings, a repository that allows autotransfusion 
can be connected to the chest tube before 
insertion. Indications for urgent thoraco- 
tomy include (1) large initial chest tube output 
(>1500mL in adults and >20% of estimated 
blood volume in children), (2) significant 
ongoing hemorrhage (>200 to 250 mL per 
hour in adults and >1 to 2mL/kg per hour 



TABLE 13-1 

APPROPRIATE CHEST TUBE SIZES IN 
PATIENTS WITH TRAUMATIC 
HEMOTHORAX 



Age 



Ag 



Newborn 

Infants 

School age children 

Adolescents 

Adult 



Chest Tube Size 

12-16 French 
16-18 French 
18-24 French 
28-32 French 
36 French 



in children), and (3) a significant increase in 
bleeding. 



PERICARDIOCENTESIS 

If hemopericardium is present and the patient 
is hemodynamically unstable, a subxiphoid 
pericardial catheter should be placed in the 
emergency center. Intermittent removal of 
pericardial blood may prevent sudden hemo- 
dynamic deterioration while preparing the 
patient for surgery. Therefore, after insertion, 
the catheter is secured in position to allow 
repeated drainage as needed during transport 
to the operating room and induction of 
anesthesia. 



EMERGENCY CENTER THORACOTOMY 

Emergency center thoracotomy in patients 
presenting with signs of life and hemodynamic 
collapse may reveal injuries to major thoracic 
vessels. In this setting, the thoracotomy allows 
rapid resuscitation and temporary control of 
bleeding in preparation for subsequent trans- 
fer to the operating room and definitive repair. 
A pericardiotomy anterior to the phrenic 
nerve is performed to relieve pericardial tam- 
ponade and allow effective cardiac compres- 
sions. Bleeding from subclavian vessels can be 
temporized by tightly packing the thoracic 
apex or by inserting large balloon catheters 
through the wounds. Either cross clamping 
the entire hilum or twisting the lung 180 
degrees after releasing the inferior pulmonary 
ligament can control major hemorrhage from 
the pulmonary hilum. 



Secondary Survey 

The secondary survey includes a search for 
more subtle signs of vascular injury. Each 
region of the body is thoroughly examined. 
All penetrating wounds are noted and marked 
with radiopaque markers. Substantial exter- 
nal bleeding is more common after stab 
wounds than gunshot wounds. Because vas- 
cular thrombosis or an intimal flap may 



chl3.qxd 4/16/04 3:41PM Page 255 



13 • PENETRATING THORACIC VASCULAR INJURY 



255 



TABLE 13-2 

CLUES TO PENETRATING THORACIC VASCULAR INJURY 



Physical Examination 



Shock 

Superior vena cava syndrome 

Pulse or pressure disparity between right and left 

upper extremities 
Pulse or pressure disparity between upper and 

lower extremities 
Intrascapular murmur 
Hematoma at base of neck 
Signs of pericardial tamponade: 

Elevated venous pressure 

Muffled heart sounds 

Pulsus paradoxus 



Chest Radiography 



Large hemothorax 

Foreign bodies (bullets or shrapnel) or their trajectories in 

proximity to the great vessels 
A foreign body out of focus with respect to the remaining 

radiograph, which may indicate its intracardiac location 
A trajectory with a confusing course, which may indicate a 

migrating intravascular bullet 
"Missing" missile in a patient with a gunshot wound to the 

chest, suggesting distal embolization 



completely occlude an injured vessel, the 
absence of significant bleeding does not rule 
out a major vascular injury. Examination of 
the chest may reveal an expanding hematoma 
at the thoracic inlet or an intrascapular 
murmur. Thrills or bruits near the clavicles 
may indicate the presence of an arteriovenous 
fistula, which most commonly involves the 
innominate or subclavian vessels. During 
assessment of extremity circulation, the pres- 
ence of a distal pulse does not rule out a prox- 
imal injury because blood flow can continue 
while the surrounding hematoma is contained 
by perivascular tissue. Loss of an extremity 
pulse may indicate intravascular embolization 
of a bullet from an aortic injury. Unequal 
blood pressures or pulses in the upper extrem- 
ities suggest an innominate or subclavian 
artery injury. An injury involving the descend- 
ing thoracic aorta may cause pseudocoarcta- 
tion syndrome with upper extremity 
hypertension and diminished lower extrem- 
ity pulses and pressures. Clinical signs indica- 
tive of penetrating thoracic great vessel injuries 
are summarized in Table 13-2. 

As part of the secondary survey, a supine 
anteroposterior chest radiograph is per- 
formed in the emergency center after placing 
radiopaque markers on all entrance and exit 
wounds; findings that suggest an intrathoracic 
vascular injury are listed in Table 13-2. In 
many cases, the radiographic findings are suf- 



ficient to warrant immediate arteriography or 
direct transport to the operating room. 



DIAGNOSTIC STUDIES 



Unlike penetrating abdominal vascular 
trauma, the operative approach to intratho- 
racic vascular injuries varies substantially and 
depends on the location of the injury (Table 
13-3) . Therefore, imaging studies play a crit- 
ical role in diagnosing and localizing the injury 
so that the optimal approach can be planned. 



Catheter Arteriography 

In stable patients with penetrating thoracic 
trauma, catheter angiography is indicated for 
suspected innominate, carotid, or subclavian 
arterial injuries. Different thoracic incisions 
are required for proximal and distal control 
of each the brachiocephalic vessels. Arteri- 
ography, therefore, is essential for localizing 
the injury and planning the appropriate inci- 
sion. Proximity of a missile trajectory to the 
brachiocephalic vessels, even without any 
physical findings of vascular injury, is an indi- 
cation for arteriography. 

Although aortography may also be useful 
in hemodynamically stable patients with 



chl3.qxd 4/16/04 3:41PM Page 256 



256 



IV • SPECIFIC VASCULAR INJURIES 



TABLE 13-3 

RECOMMENDED INCISIONS FOR THORACIC VASCULAR INJURIES 



Injured Vessel 

Uncertain injury (hemodynamically unstable) 



Ascending aorta 
Transverse aortic arch 

Descending thoracic aorta 
Innominate artery 
Right subclavian artery or vein 
Left common carotid artery 
Left subclavian artery or vein 



Pulmonary artery 
Main/intrapericardial 
Right or left hilar 

Pulmonary vein 

Innominate vein 

Intrathoracic vena cava 



Incision 






Left anterolateral thoracotomy 

± transverse sternotomy 

± right anterolateral thoracotomy (clamshell) 

Median sternotomy 

Median sternotomy 

± neck extension 

Left posterolateral thoracotomy (fourth intercostal space) 

Median sternotomy with right cervical extension 

Median sternotomy with right cervical extension 

Median sternotomy with left cervical extension 

Left anterolateral thoracotomy (third or fourth intercostal space) 

with separate left supraclavicular incision 
± connecting vertical sternotomy ("book" thoracotomy) 

Median sternotomy 

Ipsilateral posterolateral thoracotomy 

Ipsilateral posterolateral thoracotomy 

Median sternotomy 

Median sternotomy 



suspected penetrating aortic injuries, its lim- 
itations in this setting must be recognized. If 
the laceration has temporarily "sealed off," or 
if the column of aortic contrast overlies a small 
area of extravasation, the resulting "negative" 
aortogram may foster a false sense of security. 
To maximize sensitivity, therefore, an effort 
must be made to obtain views that are tan- 
gential to possible injuries (Figs. 13-1 and 
13-2). 



Computed Tomography 

Until recently, conventional computed tomog- 
raphy (CT) had a limited role in evaluating 
vascular injuries. Although CT could demon- 
strate hemomediastinum and other sugges- 
tive signs, it did not provide the diagnostic 
capability of standard aortography. However, 
newer helical CT equipment is much faster, 
uses advanced computer analysis, and per- 
forms techniques different from first- or 
second-generation CT scanners. Not only is 
the resolution much greater than in former 
models, but the current machines also allow 
for computerized anatomic reconstruction, 
which was not available earlier. Compared to 



catheter arteriography, CT angiography 
(CTA) is faster and less expensive, and it 
eliminates complications related to arterial 
catheterization. Data regarding its reliability 
in evaluating acute injuries, however, are 
limited and few trauma centers are equipped 
for its routine use. Although prospective trials 
will be required to verify its accuracy, CTA 
with three-dimensional reconstruction is 
rapidly evolving and may replace catheter 
arteriography as the study of choice in the 
future. Although magnetic resonance angiog- 
raphy can generate similarly detailed infor- 
mation, its application in these potentially 
unstable trauma patients is not currently 
practical. 



TREATMENT OPTIONS 
Endovascular Stenting 

Evolving techniques in endovascular stenting 
are providing new options for the treatment 
of vascular trauma. Endovascular grafts can 
seal vascular lacerations from within the 
lumen without compromising blood flow. In 



chl3.qxd 4/16/04 3:41PM Page 257 



13 • PENETRATING THORACIC VASCULAR INJURY 



257 




■ FIGURE 13-1 

Missed injury by aortography. Chest radiograph (A) of a patient with a tiny puncture wound from a 
Philips screwdriver in the left second intercostal space at the sternal border. The patient arrived in 
the emergency room 30 minutes after being wounded and had stable vital signs for the following 
48 hours. Anteroposterior (B), left anterior oblique (C), and near-lateral (D) projections of the 
aortogram were each interpreted by staff radiologist as showing no injury. Subtraction aortography 
in the lateral projection (E) demonstrates tiny outpouching of the thoracic aorta anteriorly at the 
base of the innominate artery and posteriorly on the undersurface of the transverse aortic arch. 
Penetrating injury of the transverse aortic arch was confirmed intraoperatively. (From Mattox KL: 
Approaches to trauma involving the major vessels of the thorax. Surg Clin North Am 1989;69:83.) ■ 



chl3.qxd 5/3/04 4:32PM Page 258 



258 IV • SPECIFIC VASCULAR INJURIES 




FIGURE 13-1, cont'd 



principle, several aspects of vascular trauma 
make it well suited for transcatheter repair. 
These injuries occur predominately in rela- 
tively young patients without peripheral vas- 
cular occlusive disease. Furthermore, remote 
access can minimize the morbidity and tech- 
nical difficulty often associated with direct sur- 
gical repair, particularly when the traumatic 
lesion occurs in the presence of a large 
hematoma, pseudoaneurysm, or arteriove- 
nous fistula. Although direct vascular repair 
is generally successful, the wide surgical expo- 
sure that is often required can cause persis- 
tent pain and various degrees of disability. For 
example, the need for clavicular resection 
increases the morbidity of subclavian vascu- 
lar repairs. Despite the advantages of a less 
invasive approach, many trauma patients, such 
as those who are hemodynamically unstable 
or those with heavily contaminated wounds, 
will not be suitable candidates for endovas- 
cular repairs. 

Despite these caveats, endovascular tech- 
niques are being successfully applied with 
increasing frequency. Parodi and colleagues 
(1999) reported a series of 29 patients who 




A 

■ FIGURE 13-2 

Plain chest radiograph (A)oi a patient with a penetrating chest wound. 6, The aortogram 
demonstrates no apparent injury in the anteroposterior projection but reveals a defect in the anterior 
aortic wall on the left anterior oblique projection. (From Mattox KL, Wall MJ Jr, LeMaire SA: Injury to 
the thoracic great vessels. In: Mattox KL, Feliciano DV, Moore EE, eds, Trauma, 4th ed. New York: 
McGraw-Hill, 2000.) ■ 



chl3.qxd 4/16/04 3:41PM Page 259 



13 • PENETRATING THORACIC VASCULAR INJURY 



259 



underwent endovascular stent placement for 
post-traumatic false aneurysms (10 patients) 
and arteriovenous fistulas (19 patients). 
Twenty-two of these injuries were located in 
thoracic or neck vessels, that is, the subcla- 
vian artery (9 injuries), axillary artery (3 
injuries), aorta (2 injuries), common carotid 
artery (5 injuries) , and internal carotid artery 
(3 injuries). The false aneurysms or arteri- 
ovenous fistulas were closed completely by one 
or more stent-graft devices in 28 of 29 patients. 
One patient died 1 month after the stent-graft 
false aneurysm closure. Twenty-three of 
twenty-nine patients continued to demon- 
strate stent-graft patency and remained asymp- 
tomatic after 24 months mean follow-up. 
Event-free survival at 3yearswas 83%. In 2000, 
du Toit and colleagues published a series of 
10 patients who qualified for stent-graft place- 
ment. The vessels involved were subclavian 
artery (in 7 patients), carotid artery (2 
patients), and axillary artery (1 patients). 
Seven had arteriovenous fistulas and three pre- 
sented with pseudoaneurysms. On average 
follow-up of 7months, no complicationswere 
encountered. 



Surgical Repair 

Whenever possible, imaging studies are used 
to establish the diagnosis and plan the surgi- 
cal approach. Clinical deterioration before 
obtaining these studies requires immediate 
transfer to the operating room for thoraco- 
tomy; indications for urgent operation include 
hemodynamic instability, hemopericardium, 
major hemorrhage from chest tubes, and radi- 
ographic evidence of a rapidly expanding 
mediastinal hematoma. 



PREOPERATIVE CONSIDERATIONS 

It is important to inform patients and their 
families of the potential for neurologic com- 
plications, such as paraplegia, stroke, and 
brachial plexus injuries, following surgical 
reconstruction of the thoracic great vessels. 
Careful documentation of preoperative neu- 
rologic status is critical. With any suspicion of 
vascular injury, prophylactic antibiotics are 



administered preoperatively. In hemody- 
namically stable patients, fluid administration 
is limited until vascular control is achieved in 
the operating room. An autotransfusion 
device should be prepared. During the induc- 
tion of anesthesia, wide swings in blood pres- 
sure are avoided; although profound 
hypotension is clearly undesirable, hyperten- 
sive episodes can have equally devastating 
consequences. 

The operative approach to great vessel 
injury varies depending on both the overall 
patient assessment and the specific injury. The 
initial steps of patient positioning and inci- 
sion selection (see Table 13-2) are particu- 
larly important in surgery for thoracic vascular 
injuries, as adequate exposure is mandatory 
for proximal and distal control. Preparing and 
draping the patient should provide access 
from the neck to the knees to allow manage- 
ment of all contingencies. For the hypoten- 
sive patient with an undiagnosed injury, the 
mainstay of thoracic trauma surgery is the left 
anterolateral thoracotomy with the patient in 
the supine position. In stable patients, pre- 
operative arteriography may dictate an oper- 
ative approach by another incision. 

Appropriate graft materials should be avail- 
able. Although an infected prosthetic graft 
may form a pseudoaneurysm, a saphenous 
vein graft is a devitalized collagen tube sus- 
ceptible to bacterial collagenase, which can 
cause graft dissolution leading to acute 
rupture and uncontrolled hemorrhage. 
Therefore, for vessels larger than 5mm, pros- 
thetic graft material is the conduit of choice, 
especially in potentially contaminated 
wounds. However, because of patency con- 
siderations, a saphenous vein graft may need 
to be used when smaller grafts are required. 
For soft vessels, such as the subclavian artery 
and the aorta in young people, a soft knitted 
Dacron graft is useful. Antibiotic irrigation of 
the graft material may help prevent subse- 
quent infection. 



DAMAGE CONTROL 

Patients with severely compromised physio- 
logic reserve, including those in extremis and 
those with massive or multiple complex tho- 



chl3.qxd 4/16/04 3:41PM Page 260 



260 



IV • SPECIFIC VASCULAR INJURIES 



racic injuries, often require damage control 
injury management to achieve survival. The 
two approaches to thoracic damage control 
are (1) definitive repair of injuries using quick 
and simple techniques that restore survivable 
physiology during a single operation and (2) 
abbreviated thoracotomy that restores sur- 
vivable physiology and requires a planned sub- 
sequent operation for definitive repairs. 
Performing a pneumonectomy using stapling 
devices can quickly control severe hilar vas- 
cular injuries. Temporary vessel ligation or 
placement of intravascular shunts can control 
bleeding until subsequent correction of aci- 
dosis, hypothermia, and coagulopathy allows 
the patient to be returned to the operating 
room. En mass closure of a thoracotomy with 
a continuous heavy suture is more hemosta- 
tic than towel-clip closure. A plastic "Bogota 
bag" can be used as a temporary closure of a 
median sternotomy in cases with associated 
cardiac dysfunction. 



SPECIFIC INJURIES 



Thoracic Inlet 

SUBCLAVIAN ARTERY AND VEIN 

Penetrating trauma to the periclavicular 
region with injury to the innominate, sub- 



clavian, and axillary vessels continues to pose 
a challenging problem for the surgeon 
because of the significant morbidity and mor- 
tality that occurs following damage to these 
vessels. The subclavian vessels are the most 
commonly injured great thoracic vessels: 21% 
of thoracic greatvessel injuries involve the sub- 
clavian arteries and 13% involve the subcla- 
vian veins. Venous injuries have a significantly 
higher mortality than arterial injuries; in a 
series of 228 penetrating subclavian vessel 
injuries reported by Demetriades (1987), the 
overall mortality was 82% and 60%, respec- 
tively (P< .01). Approximately 61% of patients 
sustaining injuries to the subclavian vessels 
are dead on arrival to the emergency center. 
Of those who reach the hospital alive, most 
require operative intervention, with an oper- 
ative mortality of up to 16% and substantial 
surgical morbidity. External or intrathoracic 
bleeding from the subclavian vessels may be 
difficult to control with direct pressure given 
their anatomic position behind the clavicle. 
In the presence of a supraclavicular wound, 
balloon tamponade using one or two Foley 
catheters placed through the wound may 
control bleeding until the patient arrives in 
the operating room (Fig. 13-3). 

Patients with injury to the subclavian vessels 
may present with hard signs of vascular injury, 
such as absent distal pulses, expanding or pul- 
satile hematomas, or massive external hem- 
orrhage. However, a subset of patients will not 



■ FIGURE 13-3 

Balloon tamponade of subclavian 
vascular bleeding. A Foley catheter 
is inserted into the supraclavicular 
wound and is advanced as far as it 
can go. The balloon is then inflated 
and firm traction is applied to the 
catheter. The balloon compresses 
the subclavian vessels against the 
clavicle and the first rib. If there is 
persistent external bleeding, a 
second catheter is inserted and the 
balloon is inflated inside the wound 
tract, superficial to the first balloon. 
(Modifed from Demetriades D. 
Penetrating injuries to the thoracic 
great vessels. J Card Surg 
1997;12:173-180.) ■ 




chl3.qxd 4/16/04 3:41PM Page 261 



13 • PENETRATING THORACIC VASCULAR INJURY 



261 



exhibit any of these findings, and their injury 
may be found on angiograms obtained solely 
on the basis of location of injury or chest radi- 
ograph findings. 

One pitfall in subclavian injuries is failure 
to anticipate the exposure necessary for prox- 
imal control. When approaching the sub- 
clavian artery via a supraclavicular incision 
without proximal control, exsanguination 
may occur. A median sternotomy with a cer- 
vical extension is employed for exposure of 
right-sided subclavian injuries. For left sub- 
clavian artery injuries, proximal control is 
obtained through an anterolateral thoraco- 
tomy (above the nipple, third or fourth inter- 
costal space) , while a separate supraclavicular 
incision provides distal control. In the 
extremely difficult left-sided subclavian artery 
injury, a formal "book" thoracotomy incision 
may be required. This approach is associated 
with a high incidence of postoperative "causal- 
gia" type of neurologic complications; there- 
fore, its use should be limited. Although this 
carries significant morbidity, resection of the 
clavicle may also aid in obtaining proximal 
control. Alternatively, for distal injuries, a com- 
bination of supraclavicular and infraclavicu- 
lar incisions may be used to avoid the 
morbidity of clavicular resection. In obtain- 
ing exposure, it is important to avoid injur- 
ing the phrenic nerve, which is located 
anterior to the scalenus anticus muscle, and 
the brachial plexus. Many patients with sub- 
clavian injuries will present with associated 
brachial plexus injuries, so careful docu- 
mentation of preoperative neurologic status 
is important. 

In most instances, repair of the subclavian 
artery requires either lateral arteriorrhaphy 
or graft interposition. A primary end-to-end 
anastomosis is usually not possible because of 
the fragility of the artery and limited mobi- 
lization. Associated injuries to the lung should 
be managed with stapled wedge resection or 
pulmonary tractotomy. 

Achievement of adequate surgical exposure 
can be difficult and may be associated with 
postoperative neurologic complications; 
therefore, certain patients sustaining pene- 
trating injuries to the subclavian vessels may 
benefit from stent-graft treatment. The success 
of stent-graft treatment for traumatic lesions 



depends largely on patient selection. Patients 
must be hemodynamically stable, and it must 
be possible to traverse the damaged segment 
with aguidewire. To avoid endoleakswith this 
technique, the proximal-distal lumen dis- 
crepancy must not be too large, and in the 
case of injuries to the subclavian artery, all side- 
branches, which potentially participate in the 
lesion, must be embolized before stent 
deployment (Fig. 13-4). 



INNOMINATE ARTERY AND VEIN 

The innominate artery is injured in approx- 
imately 9% of patients sustaining penetrating 
thoracic vascular trauma. Injuries to the left 
innominate vein are three times more 
common than those to the shorter right 
innominate vein. These injuries are 
approached through a median sternotomy 
with a right- or left-sided extension into the 
neck. Isolated venous injuries can be managed 
with primary repair or ligation. Division or 
ligation of the innominate vein can also be 
used to enhance exposure of the underlying 
artery. 

In selected patients with only partial tears, 
the innominate artery may be primarily 
repaired using 4-0 polypropylene suture. 
More often, injuries to this vessel require 
repair via the bypass exclusion technique, 
which does not require cardiopulmonary 
bypass, hypothermia, systemic anticoagula- 
tion, or shunting. Bypass grafting is per- 
formed from the ascending aorta to the distal 
innominate artery using a Dacron tube graft 
(usually a 10-mm graft in adults) . The area of 
injury is carefully avoided until the bypass is 
completed. The proximal anastomosis con- 
nects the graft to the ascending aorta away 
from the innominate artery origin; this is 
accomplished using a partial occluding, "side- 
biting" clamp on the ascending aorta. The 
distal anastomosis requires proximal and 
distal control of the innominate artery. If the 
proximal portion of the artery is injured, a 
partial occluding clamp can be placed across 
the adjacent aorta. For distal control, a vas- 
cular clamp is placed proximal to the bifur- 
cation of the innominate artery to allow 
collateral flow from the right subclavian artery 



chl3.qxd 4/16/04 3:41PM Page 262 



262 IV • SPECIFIC VASCULAR INJURIES 




■ FIGURE 13-4 

Treatment of a pseudoaneurysm of the left subclavian artery. A, Selective left subclavian artery 
arteriogram demonstrating the pseudoaneurysm; B, complete exclusion of the lesion after stent- 
graft deployment. (From du Toit DF, Strauss DC, Blaszczyk M: Endovascular treatment of 
penetrating thoracic outlet arterial injuries. Eur J Vase Endovasc Surg 2000;19:489-495.) ■ 



to perfuse the right carotid artery. After the 
bypass is completed, the aorta is controlled 
with a partial occluding clamp and is oversewn. 
If concomitantly injured or previously divided, 
the innominate vein may be ligated with 
impunity. Alternatively, in stable patients, the 
vein can be reanastomosed. If the vein remains 
intact, a pedicled pericardial flap can be posi- 
tioned between the vein and overlying graft 
to prevent erosion. With the bypass principle, 
reconstruction of innominate vascular injury 
carries an extremely low mortality rate and 
minimal morbidity, except in patients with pre- 
operative neurologic injury or complex asso- 
ciated injuries. 



LEFT COMMON CAROTID ARTERY 



than 50% of these patients require urgent 
or semiurgent intubation because of the 
expanding hematoma. Tracheostomy is 
avoided because it may disrupt an underlying 
hematoma and cause severe bleeding. A base- 
line neurologic examination should be doc- 
umented. Intraoperative endoscopy should be 
considered to rule out associated tracheal or 
esophageal injuries. The operative approach 
for injuries of the left carotid artery mirrors 
that used for an innominate artery injury: a 
median sternotomy with a left cervical exten- 
sion added when necessary. As with other great 
vessel injuries, the use of shunts or pumps 
is unnecessary. If the artery is transected 
near its origin, repair with the bypass princi- 
ple is preferred over a primary end-to-end 
anastomosis. 



Injuries to the left common carotid artery are 
relatively uncommon when compared with 
other sites, especially since penetrating tho- 
racic outlet injuries account for less than 
5% of all civilian vascular trauma. The early 
management of these patients is critical 
and cannot be overemphasized. Immediate 
airway control is a priority because more 



Thoracic Aorta 

ASCENDING AORTA 

Although penetrating injuries involving the 
ascending aorta (see Fig. 13-2) are uncom- 
mon, they do occur more often than blunt 



chl3.qxd 4/16/04 3:41PM Page 263 



13 • PENETRATING THORACIC VASCULAR INJURY 



263 



ascending aortic injuries. Survival rates 
approach 50% for patients having stable vital 
signs on arrival at a trauma center. 

Although primary repair of anterior lacer- 
ations can be accomplished without adjuncts, 
cardiopulmonary bypass may be required if 
there is an additional posterior injury. The 
possibility of a peripheral bullet embolus must 
always be considered in these patients. 



TRANSVERSE AORTIC ARCH 

Penetrating aortic arch injuries are increas- 
ing in frequency because of the escalating use 
of firearms; however, the overall incidence of 
these injuries remains small, perhaps because 
of its short length and restricted location. The 
dominant clinical presentation is a penetrat- 
ing thoracic wound with intrathoracic hem- 
orrhage and shock. 

When approaching an injury to the trans- 
verse aortic arch, extension of the median ster- 
notomy to the neck is important to obtain 
complete exposure of the arch and brachio- 
cephalic branches. If necessary, exposure can 
be further enhanced by division of the innom- 
inate vein. When hemorrhage limits exposure, 
the use of balloon tamponade is useful as a 
temporary measure. Simple lacerations may 
be repaired by lateral aortorrhaphy. With dif- 
ficult lesions, such as posterior lacerations 
or those with concomitant pulmonary artery 
injuries, cardiopulmonary bypass is recom- 
mended. As with injuries to the ascending 
thoracic aorta, survival rates approaching 
50% are possible. 



DESCENDING THORACIC AORTA 

Penetrating injury to the descending thoracic 
aorta occurs in 21 % of patients presenting with 
wounds to the thoracic vasculature and is often 
accompanied by other organ injuries, such as 
the esophagus and heart. Patients may present 
to the emergency center with exsanguination, 
enlarging hemothorax, or bullet embolism to 
the lower extremity. 

Injuries to the descending thoracic aorta 
are ideally approached via a posterolateral tho- 
racotomy through the fourth intercostal space. 



However, these are often found during emer- 
gent exploration via anterolateral thora- 
cotomy. Although lateral aortorrhaphy is 
usually possible, the surgeon must also be pre- 
pared to perform patch graft aortoplasty or 
interposition grafting. 

While gaining proximal control at the 
upper descending thoracic aorta, care must 
be taken to avoid injuring the left recurrent 
laryngeal nerve. If itis suspected that the injury 
extends to the aortic arch or ascending aorta, 
cardiopulmonary bypass should be available 
in the operating room. If the patient has had 
previous coronary artery bypass surgery with 
use of the left internal mammary artery as 
a conduit, repair may require profound 
hypothermic circulatory arrest. 

The most feared complication of descend- 
ing thoracic aortic injury is paraplegia, which 
has been associated with perioperative 
hypotension, injury or ligation of the inter- 
costal arteries, and the complexity of the 
injury. We have advocated simple clamp and 
repair for injuries to the descending thoracic 
aorta (without the use of systemic anticoagu- 
lation or shunts), a technique that continues 
to be used with excellent results. 



Other Major Intrathoracic 
Vessels 

PULMONARY ARTERY AND VEIN 

The pulmonary artery is damaged in 16% of 
patients presenting to the emergency center 
with penetrating trauma to the thoracic 
vessels, while the pulmonary veins are injured 
in 9% . These injuries are associated with mor- 
tality rates that approach 70%. The most 
common presenting manifestation of pul- 
monary artery injuries is hypotension or 
shock in association with either massive hemo- 
thorax or hemoptysis. 

Distal pulmonary artery injuries are ideally 
approached through an ipsilateral postero- 
lateral thoracotomy. These injuries are often 
identified during an emergent exploration via 
anterolateral thoracotomy. If a major injury 
to the hilum is present, rapid pneumonectomy 
may be a lifesaving maneuver. The use of a 
large balloon catheter may control exsan- 



chl3.qxd 4/16/04 3:41PM Page 264 



264 



IV • SPECIFIC VASCULAR INJURIES 



guinating hemorrhage. The intrapericardial 
pulmonary arteries are approached via 
median sternotomy. When this approach is 
used, minimal dissection is needed to expose 
the main and proximal left pulmonary arter- 
ies. Exposure of the intrapericardial right pul- 
monary artery is achieved by dissecting 
between the superior vena cava and ascend- 
ing aorta. Although anterior injuries can be 
repaired primarily without adjuncts, repair of 
a posterior injury usually requires cardiopul- 
monary bypass. If ligation of the right or left 
pulmonary artery is required, a pneumonec- 
tomy is performed. 

Injury to the pulmonary veins is difficult 
to manage through an anterior incision. With 
major hemorrhage, temporary occlusion of 
the entire hilum may be necessary. If a pul- 
monary vein must be ligated, the appropriate 
lobe needs to be resected. Pulmonary vein 
injuries are often associated with concomitant 
injuries to the heart, pulmonary artery, aorta, 
and esophagus. 



AZYGOS VEIN 

The azygos vein is not usually classified as a 
thoracic great vessel, but because of its size 
and high flow, azygos vein injuries must be 
considered potentially fatal. Penetrating 
wounds of the chest can produce combina- 
tions of injuries involving the azygos vein, 
innominate artery, trachea or bronchus, and 
superior vena cava. These complex injuries 
have a very high mortality rate and are par- 
ticularly difficult to control if approached 
through an anterior incision. Combined inci- 
sions and approaches are often needed for 
successful repair. When injured, the azygous 
vein is best managed by suture ligature on both 
sides of the injury. Concomitant injury to the 
esophagus and bronchus should be consid- 
ered and ruled out with a combination of 
direct exploration, esophagoscopy, and bron- 
choscopy before the patient leaves the oper- 
ating room. 



THORACIC VENA CAVA 



INTERNAL THORACIC AND 
INTERCOSTAL ARTERIES 



Isolated injury to the suprahepatic or supe- 
rior vena cava is infrequently reported. Injury 
at either location has a high incidence of asso- 
ciated organ trauma and carries a mortality 
rate greater than 60%. Intrathoracic inferior 
vena cava injury produces hemopericardium 
and cardiac tamponade. Exposure of the pos- 
terior thoracic inferior vena cava is extremely 
difficult unless the patient is placed on total 
cardiopulmonary bypass with the inferior 
cannula inserted via the groin to the abdom- 
inal inferior vena cava. Repair is accom- 
plished by a right atriotomy and intracaval 
balloon occlusion to prevent air entering the 
venous cannula and limit blood return to 
the heart. The injury is repaired from inside 
the cava via the right atrium. Superior vena 
cava injuries are repaired by lateral venor- 
rhaphy. At times, an intracaval shunt is nec- 
essary. For complex injuries patch angioplasty 
or an interposition tube graft (Dacron or 
ringed polytetrafluoroethylene) can be used 
safely and is more expedient than the time- 
consuming construction of saphenous vein 
panel grafts. 



Injury to the internal thoracic (i.e., the inter- 
nal mammary) artery in a young patient can 
produce extensive hemothorax or even peri- 
cardial tamponade, simulating a cardiac 
injury. Such injuries are usually serendipi- 
tously discovered at the time of thoracotomy 
for suspected great vessel or heart injury. Per- 
sistent hemothorax can be caused by simple 
lacerations of the intercostal arteries. Because 
of difficulty in exposure, precise ligature can 
be difficult. Rapid control is best achieved by 
circumferential ligatures around the rib on 
either side of the intercostal vessel injury. 
These injuries are often missed during the 
initial operation because of arterial spasm; 
bleeding ensues later when the spasm resolves. 



SPECIAL PROBLEMS 



Mediastinal Traverse Injuries 

Because penetrating injuries that traverse the 
mediastinum are classically felt to have a high 



chl3.qxd 4/16/04 3:41PM Page 265 



13 • PENETRATING THORACIC VASCULAR INJURY 



265 



probability of injury to the thoracic great 
vessels and other critical structures, manda- 
tory exploration remains a justifiable 
approach. The evaluation of stable patients 
using less invasive means, such as combined 
aortography, bronchoscopy, echocardiogra- 
phy, and esophagoscopy, is gaining propo- 
nents. Thoracoscopic evaluation of the 
mediastinal structures is another potential 
alternative that warrants investigation. 



Thoracic Duct Injury 

Injuries to the thoracic great vessels may be 
complicated by concomitant thoracic duct 
injury, which if unrecognized may produce 
devastating morbidity because of marked 
nutritional depletion. Diagnosed by chylous 
material draining from the chest tube, this 
condition is usually treated medically. Con- 
tinued chest tube drainage, coupled with a 
diet devoid of long-chain fatty acids, usually 
results in spontaneous closure in less than 1 
month. Prolonged hyperalimentation beyond 
3 weeks has not consistently resulted in spon- 
taneous closure of thoracic duct fistula. If 
thoracotomy is required, a heavy fatty meal to 
increase the chylous flow and facilitate iden- 
tification of the fistula is given to the patient 
a few hours before surgery. The fistula is simply 
ligated. 



Foreign Body Embolism 

Because of their central location, the thoracic 
great vessels may serve as both an entry site 
and a final resting place for intravascular bullet 
emboli. These migratory foreign bodies 
present a diagnostic and therapeutic dilemma. 
As the result of intravascular embolization, 
bullets may produce infection, ischemia, or 
injury to organs distant from the site of trauma. 
Bullets and catheters can embolize to the 
pulmonary vasculature; 25% of migratory 
bullets finally lodge in the pulmonary arter- 
ies. Although small fragments, such as those 
the size of a BB, can probably be left in place 
without causing problems, larger bullet emboli 
should be removed to prevent pulmonary 
thrombosis, sepsis, or other complications. 
Nonoperative management of foreign bodies 
located in the left side of the heart should be 
performed only in selected asymptomatic 
patients, such as those presenting long after 
the initial injury and in whom imaging studies 
confirm that the foreign body is encapsulated 
by fibrous tissue. Percutaneous retrieval of 
the foreign body using transvenous catheters 
and fluoroscopic guidance may obviate the 
need for thoracotomy. Intraoperative imaging 
is necessary to rule out unsuspected migra- 
tion of the foreign body during patient 
positioning. 



Systemic Air Embolism 

A fistula between a pulmonary vein and bron- 
chiole due to a penetrating lung injury may 
result in a systemic air embolism (Fig. 13-5) . 
The fistula allows air bubbles to enter the left 
heart and embolize to the systemic circula- 
tion, including the coronary and cerebral 
arteries. Intrabronchial pressure of more 
than 60 mm Hg increases the incidence of this 
complication. Manifestations include seizures 
and cardiac arrest. Resuscitation requires 
thoracotomy, clamping of the pulmonary 
hilum to prevent further air embolization, and 
aspiration of air from the left ventricle and 
ascending aorta. Cardiopulmonary bypass 
can be considered , but very few survivors have 
been reported. 



POSTOPERATIVE 
MANAGEMENT 



A significant portion of the in-hospital 
mortality associated with great vessel injury 
is secondary to the nature of the multisystem 
trauma in this group of patients. The oper- 
ating surgeon is best qualified to direct 
postoperative management. Careful hemo- 
dynamic monitoring, with avoidance of both 
hypertension and hypotension, is critical. 
Although urinary output is a generally a good 
indicator of cardiac function, for the patient 
with massive injuries, Swan-Ganz monitoring 
is often necessary to optimize hemodynamic 
parameters and manage fluids, pressors, and 
vasodilators. 



chl3.qxd 4/16/04 3:41PM Page 266 



266 



IV • SPECIFIC VASCULAR INJURIES 




Bronchiole 



Alveolus 



m FIGURE 13-5 

Drawing depicting the mechanism of systemic air embolism following a penetrating lung injury. 
(From Baylor College of Medicine, Houston, Texas.) ■ 



Various pulmonary problems, including 
atelectasis, respiratory insufficiency, pneu- 
monia, and acute respiratory distress syn- 
drome, represent the primary postoperative 
complications in this group of patients, neces- 
sitating careful fluid administration. Associ- 
ated pulmonary contusions also contribute to 
respiratory problems. Positive end-expiratory 
pressure can be provided to hemodynamically 
stable intubated patients, to minimize atelec- 
tasis. Patient mobility is important, and ade- 
quate medication for pain relief results in 
fewer pulmonary complications. For the man- 
agement of pain related to a thoracotomy or 
multiple rib fractures, postoperative thoracic 
epidural anesthesia should be considered in 
stable patients without spinal injuries; alter- 
natively, intercostal nerve blocks can be 



performed intraoperatively and repeated in 
the intensive care unit. 

Postoperative hemorrhage may be due to 
a technical problem but is often the result of 
coagulopathy related to hypothermia, acido- 
sis, and massive blood transfusion. Coagula- 
tion studies must be carefully monitored and 
corrected with administration of appropriate 
blood products. Blood draining via chest tubes 
can be collected and autotransfused. 

The presence of a prosthetic vascular graft 
requires special attention aimed at avoiding 
bacteremia. During the initial resuscitation of 
these critically injured patients, various 
intravascular lines are often rapidly placed at 
the expense of strict sterile technique; such 
lines should be replaced after the patient 
has stabilized in the intensive care unit. 



chl3.qxd 4/16/04 3:41PM Page 267 



13 • PENETRATING THORACIC VASCULAR INJURY 



267 



Antibiotic therapy should be continued into 
the postoperative period until potential 
sources of infection are eliminated. Patients 
are counseled regarding the necessity of 
antibiotic prophylaxis during invasive proce- 
dures, including dental manipulations. 

Most late complications are related to infec- 
tions or sequelae from other injuries. Long- 
term complications specifically related to the 
vascular repair, including stenosis, thrombo- 
sis, arteriovenous fistula, graft infection, and 
pseudoaneurysm formation, are uncommon. 



REFERENCES 

Bickell WH, Wall MJ Jr, Pepe PE, et al: Immediate 
versus delayed fluid resuscitation for hypoten- 
sive patients with penetrating torso injuries. N 
Engl J Med 1994;331:1105-1109. 

Demetriades D: Penetrating injuries to the thoracic 
great vessels. J Card Surg 1997;12:173-180. 

du Toit DF, Strauss DC, Blaszcyk M, et al: Endovas- 
cular treatment of penetrating thoracic outlet 



arterial injuries. Eur J Endovasc Surg 2000; 
19:489-495. 

Demetriades D, Rabinowitc B, Pezikis A, et al: 
Subclavian vascular injuries. Br J Surg 1987;74: 
1001-1003. 

Feliciano DV: Trauma to the aorta and major 
vessels. Chest Surg Clin North Am 1997;7(2):305- 
323. 

Mattox KL, Wall MJ: Trauma of the chest: newer 
diagnostic measures and emergency manage- 
ment. Chest Surg Clin North Am 1997;7(2):213- 
226. 

Parodi JC, Schonholz C, Ferreira LM, Bergan J: 
Endovascular stent-graft treatment of traumatic 
arterial lesions. Ann Vase Surg 1999;13(2):121- 
129. 

Richardson JD, Miller FB, Carrillo EH, Spain DA: 
Complex thoracic injuries. Surg Clin North Am 
1996;76(4):725-748. 

Wall MJ, Granchi T, Liscum KR, Mattox KL: Pen- 
etrating thoracic vascular injuries. Surg Clin 
North Am 1996;76(4):749-761. 



chl4.qxd 4/16/04 3:39PM Page 269 




Blunt Thoracic 
Vascular Injury 



AURELIO ROC 


RIGUEZ 


DAV 


ID C. ELL 


OTT 






O 


BLUNT THORACIC VASCULAR TRAUMA 






o 


DEMOGRAPHICS AND PATTERN OF INJURY 






o 


PRESENTATION 






o 


DIAGNOSIS 

Aortogram 

Chest X-ray 

Computed Tomography 

Transesophageal Echocardiography 






o 


PREOPERATIVE MANAGEMENT 






o 


SURGICAL TECHNIQUE 






o 


BLUNT INJURY TO OTHER THORACIC VESSELS 
Innominate Artery 
Subclavian Artery 




Vena Cava 




Pulmonary Artery and Vein 




O 


SUMMARY 



269 



chl4.qxd 4/16/04 3:39PM Page 270 



270 



IV • SPECIFIC VASCULAR INJURIES 



BLUNT THORACIC 
VASCULAR TRAUMA 



A 30-year-old man is ejected from his pickup 
truck after falling asleep at the wheel and strik- 
ing an embankment. Brought rapidly by heli- 
copter to a level I trauma center, he is found 
by expeditious evaluation to have sustained a 
mild closed head injury, a stable pelvic frac- 
ture, and multiple rib fractures. Of more 
concern, his diagnostic peritoneal lavage 
returns grossly positive and his supine chest 
radiograph reveals a widened mediastinum 
with distortion of the aortic knob. 

How will the diagnostic evaluation proceed 
from here? Will aortography precede or 
follow laparotomy? If a ruptured thoracic aorta 
is found, will a Gott shunt, a Bio-Medicus 
pump, or a "clamp-and-sew" strategy be 
pursued? Will you perform a primary repair 
of the injured aorta or place a synthetic graft? 
What perioperative measures can you employ 
to minimize postoperative complications such 
as paraplegia? These are a few of the often 
controversial questions surrounding evalua- 
tion and treatment of the patient with trau- 
matic rupture of the thoracic aorta, which we 
attempt to address in this chapter. In addi- 
tion, the less commonly seen blunt injuries 
to other thoracic vascular structures are also 
reviewed. 

Blunt thoracic aortic injury (BTAI) is a 
major cause of morbidity and mortality in the 
United States, with one fifth of motor vehicle 
accident deaths attributable thereto. Despite 
advances in surgical technique and post- 
operative care, survival has not changed much 
since 1958, when Parmley and colleagues from 
the Armed Forces Institute of Pathology and 
Walter Reed U.S. Army Hospital provided the 
classic pathophysiologic and epidemiologic 
description of BTAI. In their series of 275 
patients, 86% died at the scene. Of those who 
initially survived, only 26% were alive at 2 
weeks. More recent series report comparable 
figures for death at the scene, whereas for 
those who survived transport to a trauma 
center, survival to discharge rates of 50% to 
75% are reported. Except those presenting 
to the emergency department in extremis or 
who have obviously ruptured, injured patients 



with BTAI who are stable enough to undergo 
thoracotomy and repair have a chance of 
survival of 85%. 



DEMOGRAPHICS AND 
PATTERN OF INJURY 

BTAI classically occurs at the aortic isthmus 
1 cm distal to the left subclavian artery (at the 
site of the ligamentum arteriosum). In the 
series by Parmley and colleagues (1958), 45% 
of all aortic injuries and 63% of those in early 
survivors were at this site, which agrees well 
with results from more recent autopsy series. 
Other sites of injury, such as ascending, 
descending, and abdominal aorta, are less 
common and are often associated with spine 
fractures. Injury to other great vessels in the 
chest, such as the innominate artery and the 
subclavian artery and vein, comprises 5% or 
less of all blunt thoracic vascular trauma. 

One prospective study of 1500 patients sus- 
taining significant blunt chest trauma found 
that using multivariate logistic regression 
analysis, BTAI was associated with high-speed 
collisions (>60 miles per hour) and higher 
Injury Severity Scores (ISSs), but not direc- 
tion of impact, ejection from the vehicle, 
sudden deceleration, or other fatalities in the 
vehicle. A second recent study assessing direc- 
tion of impact found that half of BTAI victims 
(48 of 97) sustained lateral impact collisions, 
and 83% of these were wearing restraints. As 
in motor vehicle collisions, BTAI has also been 
found to be a common cause of death among 
pedestrians struck by motor vehicles, respon- 
sible in one series for 13% of pedestrian 
deaths, with a mortality rate of 93% overall. 
In the single largest prospective, multi- 
institutional study of BTAI (274 patients) , the 
mean ISS was 42, the Glasgow Coma Scale 
(GCS) score was 12, 93% of injuries were at 
the aortic isthmus, and 46 patients (17%) 
arrived to the hospital in extremis or exsan- 
guinated from free rupture soon thereafter. 
Multiple injuries were commonplace and 
included head injury (51%), rib fractures 
(46% ) , pelvic and long bone fractures (34%) , 
and abdominal injuries (22%). Traumatic 
rupture of the aorta rarely (<30% of all cases) 



chl4.qxd 4/16/04 3:39PM Page 271 



14 • BLUNT THORACIC VASCULAR INJURY 



271 



TABLE 14-1 

ASSOCIATED INJURIES TRAUMATIC RUPTURE OF THE AORTA 







Closed 












Head 




Pulmonary 


Pelvic 




Patients 


Injury 


Abdominal 


Contusion 


Fracture 


Study 


in) 


(%) 


Injury (%) 


(%) 


(%) 



Duhaylongsod, Glower, and Wolfe (1992) 
Hilgenberg and colleagues (1992) 
Hunt and colleagues (1996) 
Kieny and Charpentier (1991) 
Fabian and colleagues (1998) 
Szwerc and colleagues (1999) 



67 


31 


37 


45 


27 


51 


40 


29 


39 


25 


144 


37 


25 


16 


26 


73 


62 


18 




25 


274 


51 


22 


38 


31 


30 


37 


53 


40 


33 



Associated injuries recorded as percent of patients afflicted. 



occurs without significant associated injuries. 
Table 14-1 lists nine recent series of patients 
with BTAI and commonly associated injuries. 
In four series, the ISS was calculated, and the 
average excluding the aortic component was 
18. 

Although free rupture of the aorta is imme- 
diately lethal, the circumferential extent of 
contained tears is not clearly associated with 
increased mortality. In the Parmley and col- 
leagues (1958) series, 24% of early survivors 
had complete circumferential tears. Again, 
this finding has been reproduced in more 
recent series. Patients with partial-thickness 
tears who survive without surgical repair 
develop a fibrous pseudoaneurysm. These may 
be discovered incidentally on routine chest x- 
ray films years after the inciting injury. Alter- 
natively, these pseudoaneurysms may become 
symptomatic or result in delayed rupture in 
one third of patients. Therefore, it is recom- 
mended that pseudoaneurysms of the thoracic 
aorta are repaired whenever found. Death 
from BTAI appears to follow a bimodal dis- 
tribution, with early deaths (<4 hours) being 
due to free rupture of the aorta and exsan- 
guination and late deaths uncommonly due 
to bleeding, but due to associated injuries and 
resultant multiorgan failure. 



PRESENTATION 



Despite the dire nature of the injury, the diag- 
nosis of BTAI is often subtle because history 



and physical examination after blunt thoracic 
trauma are neither sensitive nor specific for 
aortic injury. Certain clues should, however, 
raise the index of suspicion: appropriately 
severe mechanism (high-speed motor vehicle 
collision, pedestrian struck, or a fall from great 
height); dyspnea; dysphagia; interscapular 
pain; significant chest wall trauma (multiple 
rib fractures or steering-wheel imprint) ; new 
cardiac or interscapular murmur; left-sided 
hemothorax; left supraclavicular hematoma; 
and pseudocoarctation (relative upper extre- 
mity hypertension). In particular, left- 
sided hemothorax greater than 500 mL, left 
supraclavicular hematoma, and pseudo- 
coarctation may be signs of imminent free 
rupture. 



DIAGNOSIS 
Aortogram 

The gold standard for the diagnosis of BTAI 
has been the aortogram. Not only is conven- 
tional aortography highly sensitive and spe- 
cific for aortic injury, but it also provides 
precise anatomic localization of the injury, 
which may help guide surgical repair (Fig. 14- 
1) . Conversely, aortography is expensive, inva- 
sive, and time and resource intensive, requires 
patient transport away from the trauma bay, 
and has a morbidity rate of up to 10%, making 
it unsuitable as a general screening exami- 
nation. Digital subtraction angiography may 



chl4.qxd 4/16/04 3:39PM Page 272 



272 



IV • SPECIFIC VASCULAR INJURIES 




■ FIGURE 14-1 

Digital subtraction aortogram demonstrating a 
pseudoaneurysm of the proximal descending 
thoracic aorta. (Courtesy S. Mirvis.) ■ 



improve diagnostic accuracy while decreasing 
contrast loads. 



Chest X-ray 

Because of the limitations of aortography and 
the lack of sensitivity and/or specificity of the 
clinical presentation, all patients with blunt 
trauma should undergo an initial supine 
anteroposterior chest x-ray. The purpose of 
this study is to detect indirect evidence of BTAI 
such as mediastinal hemorrhage or bony 
fractures indicating high-energy transfer. Fre- 
quently cited signs suggestive of BTAI are listed 
in Box 14-1. 

Many of the signs listed in Box 14-1 are tech- 
nique dependent and repeated evaluation in 
the upright position or standard posteroan- 
terior projection is advocated if there is no 
clinical contraindication (Fig. 14-2). Using the 
presence of any of the aforementioned signs 
as a trigger for further evaluation, some aortic 
injuries will still be missed. In a review of the 
radiologic literature, Woodring and Dillon 
(1984) found that of 656 cases of BTAI, 7.3% 



CHEST X-RAY SIGNS SUGGESTIVE OF AORTIC RUPTURE 



Mediastinal widening > 8cm 

Mediastinal-to-chest width ratio > 0.25 

Abnormal aortic contour 

Loss of the aorticopulmonary window 

Shift of the trachea to the right 

Shift of the orogastric/nasogastric tube to the right 

Left apical cap 

Widening of the paraspinal lines 

Depression of the left mainstem bronchus 

Left pleural effusion 

Scapular fracture 

Sternal fracture 

Thoracic spine fracture 

First or second rib fracture 

Multiple rib fractures 



chl4.qxd 4/16/04 3:39PM Page 273 



14 • BLUNT THORACIC VASCULAR INJURY 



273 



Rights were not granted to include this figure in electronic media. 
Please refer to the printed publication. 



■ FIGURE 14-2 

Anteroposterior "true erect" chest radiograph showing a widened mediastinum, loss of aortic 
contour, tracheal deviation, loss of the aorticopulmonary window, and a widened left paraspinal line. 
The patient sustained an aortic rupture. (Courtesy S. Mirvis). ■ 



had none of the standard radiologic criteria 
for mediastinal hemorrhage. Other recent 
series also report a significant incidence of 
normal chest x-ray film among patients with 
BTAI. Select patients, with compelling mech- 
anism of injury, should therefore undergo 
further evaluation despite a normal screen- 
ing chest x-ray film (Fig. 14-3) . 



Computed Tomography 

The role of computed tomography (CT) in 
the evaluation of blunt thoracic trauma is con- 
tested: Does it supplement or possibly replace 
the role of chest x-ray and aortography, or is 
it merely a waste of time and resources? Over 
the last 10 years, CT has evolved from an 
adjunct to an inadequate or equivocal chest 
radiograph to an all-purpose screening and 
diagnostic tool, potentially supplanting both 
chest x-ray and aortography. Many earlier 
series have documented that standard chest 
CT with intravenous contrast can consistently 



document mediastinal hemorrhage associated 
with BTAI, reporting a zero false-negative rate 
and conclude that chest CT can safely decrease 
the need for aortography by greater than 50 % . 
The latest generation of spiral and helical 
CT scanners have greatly increased the accu- 
racy of making a diagnosis of BTAI through 
noninvasive means. Used with a dynamic bolus 
of contrast and three-dimensional recon- 
struction algorithms, CT aortography can 
perhaps obviate the need for most conven- 
tional aortograms. A recent series by Gavant 
and colleagues applied this technique to 
1518 patients with blunt chest trauma over an 
11-month period. Of these, 127 patients had 
abnormal CT scans and subsequently under- 
went conventional aortography. CT sensitiv- 
ity for BTAI was 100% versus 94% for 
conventional aortography, and specificity was 
82% for the CT versus 96% for conventional 
aortography. A follow-up study with 38 tho- 
racic aortic and great-vessel injuries demon- 
strated that CT aortography can not only 
accurately diagnose BTAI but also provide 



chl4.qxd 4/16/04 3:39PM Page 274 



274 



IV • SPECIFIC VASCULAR INJURIES 




1. High speed collision (>60mph), ejection from vehicle, airplane crash, 
fall (>2 stories), multiple severe injuries 



■ FIGURE 1 4-3 

Suggested algorithm for radiographic evaluation of patients sustaining significant blunt thoracic 
trauma. ■ 



sufficient anatomic detail (previously only 
available through conventional aortography) 
to guide management. In our hands, CT was 
superior to angiography in the diagnosis of 
BTAI. Mirvis and colleagues have also docu- 
mented the steady progress made with tho- 
racic CT in the diagnosis of BTAI, in an earlier 
series showing thatnonhelical CT can reliably 
(100% sensitivity and negative predictive 
value) pick up the presence of mediastinal 
hematoma, but in the latest series showing the 
same reliability and accuracy with helical CT 
in demonstrating the actual aortic injury, 
precluding the need for aortography. Three 
prospective series similarly add to the growing 
body of evidence that helical CT is equal 
or superior to aortography in the diagnosis 
of BTAI, that this technology represents a 
significant improvement over previous non- 



helical CT, and that up to 95% of angiogra- 
phy scans can be obviated through use of CT 
(Fig. 14-4). 

The preceding discussions on the merits of 
CT and aortography in the diagnosis of BTAI 
must be viewed in light of their major draw- 
back: Both require transport away from the 
trauma bay and therefore are only useful in 
a hemodynamically stable patient. Fabian and 
colleagues point out that CT has the advan- 
tage in this regard; compared to aortography, 
helical CT is faster, easier, more available, and 
less invasive. Nonetheless, like any other tech- 
nologic imaging innovation, results depend 
on user experience. The near-perfect diag- 
nostic accuracy cited may not be universally 
reproducible at facilities where the volume of 
trauma patients is insufficient to allow ade- 
quate experience with this technique to 



chl4.qxd 4/16/04 3:39PM Page 275 



14 • BLUNT THORACIC VASCULAR INJURY 



275 




■ FIGURE 14-4 

Axial dynamic thoracic computed 
tomogram revealing a mediastinal 
hematoma anteriorly and an aortic 
intimal flap. The patient had 
sustained an aortic rupture. 
(Courtesy S. Mirvis). ■ 




■ FIGURE 14-5 

Transesophageal echocardiogram 
revealing an intimal flap of disrupted 
descending thoracic aorta. (From 
Brooks SW, Young JC, Cmolik B, et al: 
J Trauma 1992:32:761-766.) ■ 



accrue. In such situations, the time-tested 
modality of aortography may represent the 
diagnostic test of choice. 

Transesophageal 
Echocardiography 

Since its introduction in the early 1980s, trans- 
esophageal echocardiography (TEE) has 
become the study of choice for various cardiac 
diseases and is increasingly used for the 
evaluation of the thoracic aorta. TEE is well 
suited for this role because of the close 
anatomic proximity between the esophagus 
and the thoracic aorta. 

In a representative study by Smith and col- 
leagues (1995), TEE was attempted in 101 
blunt trauma patients but only completed in 
93. TEE diagnosis of BTAI was corroborated 



with aortography and surgery and/or autopsy. 
Overall, TEE sensitivity was 100% and speci- 
ficity was 98% with one false-positive result. 
In these and other authors' hands, TEE is both 
sensitive and specific in the diagnosis of 
BTAI. Also, TEE provides precise anatomic 
localization of the site of injury and does not 
require patient transport, making it better 
suited for the unstable patient (Fig. 14-5) . On 
the negative side, TEE cannot be used in all 
trauma patients (7% in this study) because 
of patient combativeness, cervical spinal or 
maxillofacial injury, and airway difficulty. In 
addition, results are operator dependent, and 
other authors have not been able to reliably 
reproduce such excellent results, with sensi- 
tivity for BTAI only 57% in one series. In ana- 
lyzing 10 series totaling 407 patients 
undergoing TEE to diagnose BTAI, Ben- 
Menachem (1997) found this test's sensitiv- 



chl4.qxd 4/16/04 3:39PM Page 276 



276 



IV • SPECIFIC VASCULAR INJURIES 



ity for BTAI to be 86% and inferior to that of 
aortography. To summarize, the diagnostic 
accuracy of TEE is open to question, it pos- 
sesses "blind spots" to include the ascending 
aorta, the aortic arch, and its branches, and 
certain patients cannot tolerate the procedure. 
However, it possesses a few advantages over 
other techniques, including portability, rapid- 
ity, and its safety profile. Until incontrovert- 
ible documentation of superior accuracy, 
TEE might best be reserved for the following 
circumstances: 

• Evaluation of unstable patients who are poor 
risks for transport to the CT scanner or the 
angiography suit 

• Evaluation of patients in the operating room 
during emergent laparotomy for intra- 
abdominal hemorrhage 

• Evaluation of the morbidly obese patient 
whose weight may exceed table limits of the 
CT scanner or angiography suite 



PREOPERATIVE MANAGEMENT 

Multisystem injuries are common in patients 
with BTAI; thus, keeping the entire clinical 
scenario in perspective is important when dis- 
cussing management. It has been noted that 
hypotension in patients with BTAI is generally 
not due to the aortic injury because bleeding 
from the aorta is rapidly fatal. Therefore, as 
in all trauma patients, it is imperative to dis- 
cover and address the etiology of hypotension. 
Primary and secondary surveys will reveal sites 
of external hemorrhage and sites of possible 
occult blood loss such as pelvic and long bone 
fractures. Chest x-ray and/or tube thoracos- 
tomy will expose significant intrapleural hem- 
orrhage. Intra-abdominal hemorrhage is best 
evaluated in this situation by diagnostic peri- 
toneal lavage (DPL) , CT, or abdominal ultra- 
sonography. The advantages of DPL and 
ultrasonography reside in their ability to be 
performed quickly, in both unstable and stable 
patients. The use of abdominal CT takes longer 
and requires a stable patient but gives more 
information and can be performed coincident 
to CT of the chest, if already planned. In cases 
of combined thoracic and abdominal injury, 
laparotomy should follow thoracotomy when 



signs of imminent BTAI rupture exist such as 
left hemothorax, pseudocoarctation, or supr- 
aclavicular hematoma. Otherwise, laparotomy 
should generally precede aortography and/or 
thoracotomy. 

A subset of patients with BTAI and severe 
associated injuries are poor candidates for 
immediate aortic repair. Initial nonoperative 
management can allow sufficient physiologic 
recovery for delayed aortic repair. Akins and 
colleagues (1981) suggest the following cri- 
teria for delayed repair: 

• Major intracranial injury 

• Extensive burns 

• Severe respiratory failure 

• Extensively contaminated wound 

• Sepsis 

Similarly, some authors have advocated non- 
operative therapy for patients with minimal 
aortic injury (e.g., aortic intimal irregularity 
without extravasation of contrast). These 
patients should be followed with serial angio- 
graphy or TEE to document resolution of their 
aortic injury. Afterload reduction and (3- 
blockade are useful medical adjuncts, tem- 
porizing patients with delayed operative and 
primary nonoperative management by mini- 
mizing aortic wall stress. 

Increasing experience with delayed opera- 
tive management of BTAI is resulting in an 
overall paradigm shift and change in man- 
agement strategy for this injury. There is now 
substantial evidence supporting that except- 
ing BTAI patients presenting in extremis, in- 
hospital rupture can be effectively prevented 
by keeping the systolic blood pressure below 
140 mm Hg. Most of these data have been 
accrued through retrospective analysis of 
prospective medical protocols using a com- 
bination of esmolol, labetalol, and sodium 
nitroprusside, and although outcomes were 
simply compared to historical controls, virtu- 
ally no deaths resulting from in-hospital 
rupture of BTAI have occurred in these series 
when adequate medical control of blood pres- 
sure is ensured. This allows for a planned, con- 
sidered, and prepared approach to the repair 
of this problematic injury, ensuring optimal 
patient physiologic condition and optimal hos- 
pital resource support, rather than the sense 



chl4.qxd 4/16/04 3:39PM Page 277 



14 • BLUNT THORACIC VASCULAR INJURY 



277 



of hurry and panic that can be engendered 
from the perceived need to thwart a poten- 
tial intrathoracic "time bomb." 

It is further recommended that adequate 
sedation be used at the time of endotracheal 
intubation, should this be required, and that 
fluid resuscitation be particularly judicious, 
to minimize wall stress on the attenuated aorta. 
Pneumatic antishock garments (PASGs), a 
once common appliance of patients with mul- 
tiple trauma brought to trauma centers, are 
more clearly detrimental. No benefit to these 
devices has been demonstrated clinically in 
the management of BTAI and in a porcine 
model of aortic injury, 100% of pigs with and 
0% of pigs without PASG died of their injury. 

Approximately one half of patients with BTAI 
will also have a closed head injury (see Table 
14-1). Optimally, a head CT scan should be 
obtained before BTAI repair so craniotomy can 
be planned if needed. Head CT findings will 
also bear on surgical technique; for example, 
systemic heparinization is contraindicated in 
the presence of intracranial hemorrhage. 



SURGICAL TECHNIQUE 

Most patients with blunt thoracic vascular 
injury are best served by repair via a left 
posterolateral thoracotomy, because this 
approach gives the best exposure to the aortic 
isthmus where most injuries occur. There are 
two main technical variables in the surgical 
repair of a classic (aortic isthmus) BTAI (Fig. 
14-6) , as follows: 

• "Clamp-and-sew" technique versusshunt and 

• Interposition graft versus primary repair 

The clamp-and-sew technique is less 
complex and more expedient than shunting. 
Proponents of this technique argue that it is 
superior to shunting in all cases of BTAI 
because it does not require systemic hepar- 
inization, does not have adjunct-associated 
complications (e.g., insertion site hemor- 
rhage or aortic dissection), and does not 
require special equipment. Arguably, mor- 
bidity and mortality rates are similar between 
clamp-and-sew and shunt techniques. In short, 
the key technical points in the clamp-and-sew 
technique are as follows: 



SURGICAL MODALITIES 

AWlf 

Primary Repair/ >. 





Partial Left 
Heart Bypass 



Centrifugal 
Pump Head 



■ FIGURE 14-6 

Options in operative repair techniques for rupture 
of the descending thoracic aorta. A, Primary 
or direct repair. B, Graft interposition without a 
mechanical adjunct (clamp and sew). Use of a 
mechanical adjunct in aortic repair: A, Aortoaortic 
(Gott's) shunt. B, Left atrial femoral artery bypass 
with a Bio-Medicus centrifugal flow pump. ■ 

1. Position the patient in the right lateral 
decubitus position and create a standard 
left posterolateral thoracotomy. 

2. Identify left vagus and phrenic nerves; 
retract left vagus. 

3. Circumferentially dissect left subclavian 
artery and secure with an umbilical tape. 

4. Perform sharp and blunt dissection 
between the left subclavian and left 
common carotid arteries and place umbil- 
ical tape around the proximal aorta. 

5. Circumferentially dissect descending 
aorta below hematoma and secure with 
an umbilical tape. 

6. Ligate and/or control intervening inter- 
costal arteries. 

7. Administer intravenous mannitol 

8. First cross-clamp aortic arch, followed by 
descending aorta and subclavian artery. 



chl4.qxd 4/16/04 3:39PM Page 278 



278 



IV • SPECIFIC VASCULAR INJURIES 



9. Open hematoma and control back- 
bleeding. 

10. Debride devitalized aortic wall. 

11. Anastomose aorta with or without inter- 
vening prosthetic graft. 

12. Back-bleed and vent aorta before secur- 
ing suture line. 

13. Notify anesthesiologist, then release 
subclavian and descending aorta clamps, 
followed by aortic arch clamp. 

Shunt techniques of BTAI repair are similar 
to "clamp and sew" with the exception that a 
conduit is inserted proximal and distal to the 
aortic injury. Benefits of this arrangement 
include afterload reduction of the left ven- 
tricle and preserved systemic perfusion during 
aortic cross-clamp. The least complicated of 
these techniques involve passive conduits 
such as the heparin-bonded Gott shunt. This 
is typically inserted into the left ventricle or 
ascending aorta proximally and the femoral 
artery distally. Passive conduits such as the Gott 
shunt rely on cardiac output and therefore 
may not provide adequate distal perfusion 
if cardiac performance is impaired. Active 
shunts employing centrifugal or roller pumps 
are cardiac output independent but are at the 
risk of stealing perfusion from the cerebral 
and coronary circulation. In a trauma setting, 
centrifugal pumps have the advantage over 
roller pumps of not requiring systemic 
heparinization. Like passive conduits, the 
distal insertion of these shunts is usually in 
the femoral artery and the proximal insertion 
is in the left atrial appendage. Shunts may 
decrease the incidence of ischemic spinal cord 
injury, systemic acidosis, and renal injury, and 
although the evidence in the trauma litera- 
ture on this subject has in the past been 
divided, more recent series support that the 
incidence of postoperative paraplegia is lower 
at centers that use partial left heart bypass with 
a centrifugal flow pump. A recent prospec- 
tive nationwide survey of trauma centers per- 
forming BTAI repair found that although 
mortality was no different between the two 
groups, the incidence of postoperative para- 
plegia was 16.4% among the 73 patients 
treated with the clamp-and-sew technique, 
versus 2.9% among the 69 patients treated 



using centrifugal pump bypass (P<.004). 
Citing similar results, another recent study 
showed by multivariate regression analysis that 
the factors independently predicting postop- 
erative paraplegia included older age, oper- 
ative technique (clamp and sew), clamp time 
greater than 30 minutes, and the occurrence 
of intraoperative hypotension. 

Choice of primary repair versus use of pros- 
thetic graft is largely dictated by the physical 
characteristics of the native aorta and the 
extent of aortic disruption. The advantages 
of primary repair include shorter cross-clamp 
times, decreased risk of infection, and less 
intercostal artery sacrifice (possibly con- 
tributing to ischemic spinal cord injury). In 
the pediatric population, primary repair 
abates the need for reoperation to upsize an 
outgrown aortic prosthesis. Generally, pros- 
thetic grafts are better suited for instances 
when the edges of the torn aorta are widely 
distracted making a tension-free anastomosis 
impossible. One prominent series, however, 
reports 32 consecutive aortic injuries repaired 
primarily despite up to 5-cm separation of 
the torn aortic edges. Another recent series 
wherein primary repair was the preferential 
mode of therapy for BTAI reports mortality 
and paraplegia rates comparable to that those 
reported with use of the centrifugal pump 
bypass. Use of both Dacron and polytetraflu- 
oroethylene (PTFE) have been described in 
the literature, and both provide similarly good 
results. 

Still in its infancy, endovascular repair of 
BTAI appears to be a promising area for study. 
One published series of nine patients sus- 
taining BTAI reports TEE-guided endolumi- 
nal stent repair from 1 to 8 months after injury 
with excellent results: no mortality, no rupture, 
no occlusions, and no need for revision after 
the initial procedure. 



BLUNT INJURY TO OTHER 
THORACIC VESSELS 



Innominate Artery 

Innominate and proximal carotid artery 
injury is rare, comprising less than 5% of all 



chl4.qxd 4/16/04 3:39PM Page 279 



14 • BLUNT THORACIC VASCULAR INJURY 



279 



blunt thoracic vascular trauma, although the 
innominate artery is probably the most 
common thoracic vessel injured by blunt 
trauma after the thoracic aorta. Focal neuro- 
logic deficits may be present with this injury, 
but most injuries are picked up through 
screening chest radiography, CT, and 
aortography. 

Optimal exposure is via median sternotomy, 
plus or minus right neck extension, and repair 
is advocated over ligation because of the 
concern for cerebral ischemia and resultant 
neurologic injury. Because most blunt innom- 
inate injuries occur at its aortic insertion and 
native tissue at that location may be compro- 
mised, anastomosis at this location should 
be avoided. Rather, the preferred approach 
involves the construction of a Dacron or PTFE 
bypass graft proximal to the injury by anas- 
tomosis to the aorta using a partial-occlusion 
clamp. No heparin, shunt, or cardiopul- 
monary bypass is generally employed. The 
bypass graft is sutured distally to uninjured 
vessel or vessels, and the aorta at the site of 
the innominate insertion is oversewn with 
pledgeted sutures. Outcome is generally excel- 
lent, and mortality should be 0% to 10%, based 
on hemodynamic condition on admission and 
concomitant injuries (which are common) . 
Of note, endoluminal stent repair has also 
been performed successfully for blunt innom- 
inate artery injury. 



Subclavian Artery 

Approximately 1 % to 5% of blunt thoracic vas- 
cular trauma involves the subclavian artery. 
In contrast to BTAI, shoulder harness seat belts 
have been implicated in the pathogenesis of 
this injury. Signs concerning for subclavian 
injury include supraclavicular hematoma, 
pulse deficit, brachial plexus injury, clavicu- 
lar fracture, and bruit (although none are par- 
ticularly sensitive or specific) . Pulse deficit is 
most characteristic but may be absent because 
of extensive collateral flow often present 
about the shoulder. Diagnosis of subclavian 
artery injury, as in BTAI, is best made with 
aortography. Optimal surgical exposure is 
debated (see Table 14-2), although most 
favor median sternotomy for right subclavian 
and proximal left subclavian lesions, and left 
supraclavicular incision for mid-subclavian to 
distal subclavian injuries of the left side. Recon- 
struction is generally done with prosthetic 
graft. Concomitant subclavian vein injury 
occurs in up to 20% of patients and can often 
be treated with simple ligation. If collateral 
venous return is compromised, reconstruction 
of the subclavian vein is recommended and 
can be performed with a saphenous vein inter- 
position graft or an end-to-end subclavian 
jugular vein bypass. 

One variant of blunt subclavian vessel injury 
occurs in the syndrome of scapulothoracic 



TABLE 14-2 

OPTIMAL AND ALTERNATIVE EXPOSURE FOR THORACIC VASCULAR INJURIES 



Injured Vessel 



Optimal Exposure 



Alternate Exposure 



Ascending aorta 

Right subclavian 

Innominate 

Proximal common carotid 

Left subclavian 

Aortic arch 

Aortic isthmus 

Descending aorta 

SVC 

Suprahepatic IVC 



MS 

P: MS with supraclavicular ext. 

MS 

MS 

D: L. supraclavicular 

MS 

L. posterolateral thoracotomy 

L. posterolateral thoracotomy 

MS 

Thoracoabdominal 



D: R. supraclavicular 



P: L. anterolateral thoracotomy 



D, distal; IVC, inferior vena cava; L, left; MS, median sternotomy; P, proximal; R, right; SVC, superior vena cava. 



chl4.qxd 4/16/04 3:39PM Page 280 



280 



IV • SPECIFIC VASCULAR INJURIES 



dissociation. In this devastating injury, a 
strong torsion or rotational force is applied 
to the shoulder joint, shearing the scapula and 
shoulder girdle from the chest wall, resulting 
in extensive complex fractures of the upper 
extremity and avulsion of the brachial plexus 
and subclavian vessels as they emerge from 
the thoracic cavity. Patients present in shock 
with massive swelling of the ipsilateral chest 
and neurovascular deficiency of the arm. For 
survival, treatment requires early recognition, 
control of bleeding, reversal of shock, and 
usually, substantial amputation of the involved 
extremity. 



Vena Cava 

Blunt injury to the thoracic vena cava, 
though less common than BTAI, has similar 
mortality. This difference in incidence 
between arterial and venous injuries is most 
likely due to differences in vessel wall plas- 
ticity. Surgical approach to the vena cava, and 
in particular the suprahepatic vena cava, is 
problematic and may require heroic measures 
such as total hepatic vascular occlusion and 
atriocaval shunting. When feasible, primary 
repair with lateral venorrhaphy is preferred 
even though this may result in vessel 
narrowing;. 



Pulmonary Artery and Vein 



will ensure that few of these injuries are missed. 
Chest helical CT is increasingly becoming both 
the screening and the diagnostic test of choice 
in centers that serve significant numbers of 
blunt trauma patients; however, chest radi- 
ography and aortography remain gold stan- 
dards in the diagnostic algorithm. 

Attentive medical management, based 
around the use of intravenous (3-blockers to 
keep systolic blood pressure below 140 mm Hg, 
should be employed early in patients with 
probable BTAI and may prevent in-hospital 
rupture. Technique of operative repair is con- 
troversial, and no single nationwide standard 
of care has emerged for definitive operative 
strategy. However, the preponderance of 
medical evidence increasingly points to 
decreased incidence of paraplegia after BTAI 
repair when heparin-less centrifugal pump 
bypass is employed. 

Because of the common occurrence of 
BTAI, the proclivity of its presence and repair 
to result in mortality or morbidity, and the 
wide diversity of management options that 
have been used for care, it is strongly recom- 
mended that each trauma center that deals 
with this injury consider the use of a clinical 
practice guideline for BTAI thatwould encour- 
age a single safe standard for the diagnosis 
and management of this injury. An excellent 
example of such a practice guideline has been 
developed and published by the Eastern Asso- 
ciation for the Surgery of Trauma. 



Blunt trauma to the pulmonary artery and vein 
is rare. Patients with these injuries present in 
shock from hemorrhage and/or cardiac tam- 
ponade. The treatment of choice for these 
injuries is lateral repair, but if the tear in the 
vessel wall is not amenable to this, pneu- 
monectomy is an option. 



SUMMARY 

BTAI is a common and often lethal injury. For 
those who survive the initial insult, patients 
with this injury are best served by expeditious 
evaluation and prompt repair. A high index 
of suspicion, based on mechanism of injury, 



REFERENCES 

Albrink MH, Rodriguez E, England GJ, et al: Impor- 
tance of designated thoracic trauma surgeons 
in the management of traumatic aortic transec- 
tion. Southern Med J 1994;87:497-501. 

Akins CW, Buckley MJ, Daggett W, et al: Acute trau- 
matic disruption of the thoracic aorta: A 10-year 
experience. Ann Thorac Surg 1981;31:305-309. 

AliJ,VanderbyB,PurcellC:The effect of the pneu- 
matic antishock garment (PASG) on hemody- 
namics, hemorrhage, and survival in penetrating 
thoracic aortic injury. J Trauma 1991 ;31 :846-851 . 

Attar S, Cardarelli MG, Downing SW, et al: Trau- 
matic aortic rupture: Recent outcome with 
regard to neurologic deficit. Ann Thorac Surg 
1999;67:959-965. 



chl4.qxd 4/16/04 3:39PM Page 281 



14 • BLUNT THORACIC VASCULAR INJURY 



281 



Axisa BM, Loftus IM, Fishwick G, et al: Endovas- 
cular repair of an innominate artery false 
aneurysm following blunt trauma. J Endovasc 
Ther 2000;7:245-250. 

Baker SP, O'Neill B, Haddon W, et al: The Injury 
Severity Score: A method for describing patients 
with multiple injuriesandevaluatingemergency 
care. J Trauma 1974;14:187-196. 

Ben-Menachem Y: Rupture of the thoracic aorta 
by broadside impacts in road traffic and other 
collisions: Further angiographic observations 
and preliminary autopsy findings. J Trauma 
1993;35:363-367. 

Ben-Menachem Y: Assessment of blunt aortic- 
brachiocephalic trauma: Should angiography be 
supplanted by transesophageal echocardiogra- 
phy? J Trauma 1997;42:969-972. 

BormanKR,Aurbakken CM, Weige It JA: Treatment 
priorities in combined blunt abdominal and 
aortic trauma. Am J Surg 1982;144:728-732. 

Brundage SI, Haruff R, Jurkovich GJ, et al: The 
epidemiology of thoracic aortic injuries in pedes- 
trians. J Trauma 1998;45:1010-1014. 

Buckmaster MJ, Kearney PA, Johnson SB, et al: 
Further experience with transesophageal 
echocardiography in the evaluation of thoracic 
aortic injury. J Trauma 1994;37:989-995. 

Buscaglia LC, Walsh JC, Wilson JD, et al: Surgical 
management of subclavian artery injury. Am J 
Surg 1987;154:88-90. 

Clark DE,Zieger MA, Wallace KL,etal: Bluntaortic 
trauma: Signs of high risk.J Trauma 1990;30:701- 
705. 

Cohn SM, Burns GA, Jaffe C, et al: Exclusion of 
aortic tear in the unstable trauma patient: The 
utility of transesophageal echocardiography. 
J Trauma 1995;39:1087-1090. 

Cooper C, Rodriguez A, Omert L: Blunt vascular 
trauma. Curr Probl Surg 1992;29:291-357. 

Cox CS, Allen GS, Fischer RP, et al: Blunt versus 
penetrating subclavian artery injury: Presenta- 
tion, injury pattern, and outcome. J Trauma 
1999;46:445-449. 

Demetriades D, Gomez H, Velmahos GC, et al: 
Routine helical computed tomographic evalua- 
tion of the mediastinum in high-risk blunt 
trauma patients. Arch Surg 1998;133:1084-1 088. 

Duhaylongsod FG, Glower DD, Wolfe WG: Acute 
traumatic aortic aneurysm: The Duke experience 
from 1970 to 1990.J Vase Surg 1992;15:331-343. 

Dyer AD, Moore EE, like DN, et al: Thoracic aortic 
injury: How predictive is mechanism and is chest 
computed tomography a reliable screening tool? 



A prospective study of 1561 patients. J Trauma 
2000;48:673-682. 

Ebraheim NA, An HS, Jackson WT, et al: Scapu- 
lothoracic dissociation. J Bone Joint Surg 1988; 
70:428-432. 

Fabian TC, Davis KA, Gavant ML, et al: Prospec- 
tive study of blunt aortic injury: Helical CT is 
diagnostic and antihypertensive therapy reduces 
rupture. Ann Surg 1998;227:666-676. 

Fabian TC, Richardson JD, Croce MA, et al: 
Prospective study of blunt aortic injury: Multi- 
center trial of the American Association for the 
Surgery of Trauma. J Trauma 1997;42:374-380. 

Feczko JD, Lynch L, PlessJE, et al: An autopsy case 
review of 142 nonpenetrating (blunt) injuriesof 
the aorta. J Trauma 1992;33:846-849. 

Fenner MN, Fisher KS, Sergei NL, et al: Evalua- 
tion of possible traumatic aortic injury using 
aortography and CT, Am Surg 1990;56:497- 
499. 

Finkelmeier BA, Mentzer RM, Kaiser DL, et al: 
Chronic traumatic thoracic aneurysm. Influence 
of operative treatment on natural history: An 
analysis of reported cases, 1950-1980. J Thorac 
Cardiovasc Surg 1982;84:257-266. 

Fisher RG, Oria RA, Mattox KL, et al: Conserva- 
tive management of aortic lacerations due to 
blunt trauma. J Trauma 1990;30:1562-1566. 

Forbes AD, Asbaugh DG: Mechanical circulatory 
support during repair of thoracic aortic injuries 
improves morbidity and prevents spinal cord 
injury. Arch Surg 1994;129:494-497. 

GavantJL, Flick P, Menke P, et al: CT aortography 
of thoracic aortic rupture. AJR Am J Roentgenol 
1996;166:955-961. 

Gavant ML, Menke PG, Fabian T, et al: Blunt trau- 
matic aortic rupture: Detection with helical CT 
of the chest. Radiology 1995;197:125-133. 

Hajarizadeh H, Rohrer MJ, Cutler BS: Surgical 
exposure of the left subclavian artery by median 
sternotomy and left supraclavicular extension. 
J Trauma 1996;41:136-139. 

Hilgenberg AD, Logan DL, Akins CW, et al: Blunt 
injuries of the thoracic aorta. Ann Thorac Surg 
1992;53:233-239. 

Hoff SJ, Reilly MK, Merrill WH, et al: Analysis of 
blunt and penetrating injury of the innominate 
and subclavian arteries. Am Surg 1994;60:151- 
154. 

Hunick MGM, Bos JJ: Triage of patients to angi- 
ography for detection of aortic rupture after blunt 
chest trauma: Cost-effectiveness analysis of using 
CT. AJR Am J Roentgenol 1995;165:27-36. 



chl4.qxd 4/16/04 3:39PM Page 282 



282 



IV • SPECIFIC VASCULAR INJURIES 



HuntJP, Baker CC, Lentz CW, et al: Thoracic aorta 
injuries: Management and outcome of 144 
patients. J Trauma 1996;40:547-556. 

Johansen K, Sangeorzan B, Copass MK: Traumatic 
scapulo thoracic dissociation: Case report. J 
Trauma 1991;31:147-149. 

Johnston RH, Wall MJ, Mattox KL: Innominate 
artery trauma: A thirty-year experience. J Vase 
Surg 1993;17:134-140. 

Katyal D, McLellan BA, Brenneman FD, et al: 
Lateral impact motor vehicle collisions: Signifi- 
cant cause of blunt traumatic rupture of the tho- 
racic aorta. J Trauma 1997;42:769-772. 

Kearney PA, Smith DW, Johnson SB, et al: Use of 
transesophageal echocardiography in the eval- 
uation of traumatic aortic injury. J Trauma 1993; 
34:696-701. 

Kieny R, Charpentier A: Traumatic lesions of the 
thoracic aorta: A report of 73 cases. J Cardiovasc 
Surg 1991;32:613-619. 

Kram HB, Wohlmuth DA, Appel PL, et al: Clinical 
and radiographic indications for aortography in 
blunt chest trauma. J Vase Surg 1987;6:168-176. 

Kudsk KA, Bongard F, Lim RC: Determinants of 
survival after vena caval injury: Analysis of a 14- 
year experience. Arch Surg 1984;1 1 9: 1009-1 01 2. 

Madayag MA, Kirshenbaum KJ, Nadimpalli SR, et 
al: Thoracic aortic trauma: Role of dynamic CT 
Radiology 1991;179:853-855. 

Mattox KL: Symposium: New approach in vascu- 
lar trauma. J Vase Surg 1988;7:725-729. 

Mattox KL, Feliciano DV, Burch J, et al: Five thou- 
sand seven hundred sixty cardiovascular injuries 
in 4459 patients: Epidemiologic evolution 1958 
to 1987. Ann Surg 1989;209:698-707. 

Mattox KL, Holzman M, Pickard LR, et al: 
Clamp/repair: A safe technique for treatment 
of blunt injury to the descending thoracic aorta. 
Ann Thorac Surg 1985;40:456-463. 

McCroskey BL, Moore EE, Moore FA, etal: Aunified 
approach to the torn thoracic aorta. Am J Surg 
1991;162:473-476. 

Minard G, Schurr MJ, Croce MA, et al: A prospec- 
tive analysis of transesophageal echocardiogra- 
phy in the diagnosis of traumatic disruption of 
the aorta. J Trauma 1996;40:225-230. 

Mirvis SE, Pais So, Gens DR: Thoracic aortic rup- 
ture: Advantages of intraarterial digital subtrac- 
tion angiography. AJR Am J Roentgenol 1986; 
146:987-991. 

Mirvis SE, Shanmuganathan K, Buell J, et al: Use 
of spiral computed tomography for the assess- 



ment of blunt trauma patients with potential 
aortic injury. J Trauma 1998;45:922-930. 

Mirvis SE, Shanmuganathan K, Miller BH, et al: 
Traumatic aortic injury: Diagnosis with contrast- 
enhanced thoracic CT. Five-year experience at 
a major trauma center. Radiology 1996;200:413- 
422. 

Morgan PW, Goodman LR, Aprahamian C, et al: 
Evaluation of traumatic aortic injury: Does 
dynamic contrast-enhanced CT play a role? Radi- 
ology 1992;182:661-666. 

Myers SI, Harward TRS, Cagle L: Isolated subcla- 
vian artery disse c tion af te r blun t trauma . Surge ry 
1991;109:336-338. 

Nagy K, Fabian T, Rodman G, et al: Guidelines for 
the Diagnosis and Management of Blunt Aortic 
Injury. Eastern Association for the Surgery of 
Trauma: Available at http://www.east.org. 2000. 

Omert L, Rodriguez A, Simon B, et al: Blunt vas- 
cular torso trauma: A review of 310 cases. Panam 
J Trauma 1991;2:102-111. 

Parmley LF, Mattingly TW, Manion WC, et al: Non- 
penetrating traumatic injury of the aorta. Cir- 
culation 1958;17:1086-1101. 

Pate JW, Fabian TC, Walker W: Traumatic rupture 
of the aortic isthmus: An emergency? World J 
Surg 1995;19:119-126. 

Pate JW, Gavant ML, Weiman DS, et al: Traumatic 
rupture of the aortic isthmus: Program of selec- 
tive management. WorldJ Surg 1999;23:59-63. 

Pre'tre R, Chilcott M, Murith N, et al: Blunt injury 
to the supra-aortic vessels. BrJSurgl997;84:603- 
609. 

Ramadan F, Rutledge R, Oiler D, et al: Carotid 
artery trauma: A review of contemporary trauma 
center experiences. J Vase Surg 1995;21:46-56. 

Raptopoulos V: Chest CT for aortic injury: Maybe 
not for everyone. AJR Am J Roentgenol 1994; 
162:1053-1055. 

Razzouk AJ, Gundry SR, Wang N, et al: Repair of 
traumatic aortic rupture: A 25-year experience. 
Arch Surg 2000;135:913-918. 

Read RA, Moore EE, Moore FA, et al: Partial left 
heart bypass for thoracic aorta repair: Survival 
without paraplegia. Arch Surg 1993;128:746-752. 

Richardson P, Mirvis SE, Scorpio R, et al: Value of 
CT in determining the need for angiography 
when findings of mediastinal hemorrhage on 
chest radiographs are equivocal. AJR Am J 
Roentgenol 1991;156:273-279. 

Richardson JD, Smith JM III, Grover FL: Manage- 
ment of subclavian and innominate artery 



chl4.qxd 4/16/04 3:39PM Page 283 



14 • BLUNT THORACIC VASCULAR INJURY 



283 



disruption due to blunt trauma. Am J Surg 
1977;134:780. 

Richardson JD, Wilson ME, MillerFB: The widened 
mediastinum: Diagnostic and therapeutic pri- 
orities. Ann Surg 1990;211:731-737. 

Rodriguez A, Elliott D: Blunt traumatic rupture of 
the thoracic aorta. In Maull et al. (eds) : Advances 
in Trauma and Critical Care. St. Louis, Mosby, 
1993;8:145-181. 

Rousseau H, Soula P, Perreault P, et al: Delayed 
treatment of traumatic rupture of the thoracic 
aorta with endoluminal covered stent. Circula- 
tion 1999;99:498-504. 

Saletta S, Lederman E, Fein S, et al: Trans- 
esophageal echocardiography for the initial 
evaluation of the widened mediastinum in 
trauma patients. J Trauma 1995;39:137-142. 

Schmidt CA, Smith DC: Traumatic avulsion of arch 
vessels in a child: Primary repair using hypother- 
mic circulatory arrest (case report) . J Trauma 
1989;29:248-250. 

Schmidt CA, Wood MN, Razzouk AJ, et al: Primary 
repair of traumatic aortic rupture: A preferred 
approach. J Trauma 1992;32:588-592. 

Sharma S, Reddy V, Ott G, et al: Surgical manage- 
ment of traumatic aortic disruption. Am J Surg 
1997;173:416-418. 

Smith MD, Cassidy JM, Souther S, et al: Trans- 
esophageal echocardiography in the diagnosis 
of traumatic rupture of the aorta. New Engl J 
Med 1995;332(6):356-362. 

Sweeney MS, Young DJ, Frazier OH, et al: Trau- 
matic aortic transections: Eight-year experience 
with the "Clamp-Sew" technique. Ann Thorac 
Surg 1997;64:384-389. 

Szwerc MG, Benckart DH, Lin JC, et al: Recent clin- 
ical experience with left heart bypass using a 
centrifugal pump for repair of traumatic aortic 
transection. Ann Surg 1999;230:484-492. 



Townsend RN, ColellaJJ, Diamond DL: Traumatic 
rupture of the aorta critical decisions for trauma 
surgeons. J Trauma 1990;30:1 1 69-1 1 74. 

Turney SZ, Rodriguez A: Injuries to the great tho- 
racic vessels. In Turney SZ, Rodriguez A, Cowley 
RA (eds): Management of Cardiothoracic 
Trauma. Baltimore, Williams & Wilkins, 1990: 
229-260. 

Vignon P, Gueret P, Vedrinne JM, et al: Role of trans- 
esophageal echocardiography in the diagnosis 
and management of traumatic aortic disruption. 
Circulation 1995;92:2959-2968. 

Von Oppell UO, Dunne TT, De Groot KM, et al: 
Spinal cord protection in the absence of collat- 
eral circulation: Metaanalysis of mortality and 
paraplegia. J Card Surg 1994;9:685-691. 

Wahl WL, Michaels AJ, Wang SC, et al: Blunt tho- 
racic aortic injury: Delayed or early repair? 
J Trauma 1999;47:254-259. 

Walls JT, Boley TM, Curtis JJ et al: Experience with 
four surgical techniques to repair traumatic 
aortic pseudoaneurysm. J Thorac Cardiovasc 
Surg 1993;106:283-287. 

Weiman DS, McCoy DW, Haan CK, et al: Blunt 
injuries of the brachiocephalic artery. Am Surg 
1998;64:383-387. 

Williams JS, Graff JA, Uku JM, et al: Aortic injury 
in vehicular trauma. Ann Thorac Surg 1994; 
57:726-730. 

WoodringJH, Dillon ML: Radiographic manifes- 
tations of mediastinal hemorrhage from blunt 
chest trauma. Ann Thorac Surg 1984;37:l7l- 

178. 

Zeiger MA, Clark DE, Morton JR: Reappraisal of 
surgical treatment of traumatic transection of 
the thoracic aorta.J Cardiovasc Surg 1990;31 :607- 
610. 



chl5.qxd 4/16/04 3:41PM Page 285 




Wounds of the Heart 



MATTHEW J. WALL, JR 
DAVID RICE 
ERNESTO SOLTERO 



O HISTORY 

O INCIDENCE 

O CLINICAL PATHOLOGY 

O PATHOPHYSIOLOGY 

O PREHOSPITAL MANAGEMENT 

O EMERGENCY CENTER 

O EMERGENCY CENTER PROCEDURES 

O OPERATIVE MANAGEMENT OF CARDIAC INJURIES 

Incisions 

Aortic Occlusion 

Cardiac Manipulation 

Hemorrhage Control 

Complex Injuries 

Cardiac Septal Injuries 

Cardiac Valvular Injury 

Intrapericardial Inferior Vena Cava Injury 
O SUMMARY 



HISTORY 



The heart has always been regarded as 
the sustainer of life, and its wounds have 



been approached through the ages with 
awe and apprehension. Homer provided 
antique literature with many references 
to heart wounds. Their fatality was 
clear. 

285 



chl5.qxd 4/16/04 3:41PM Page 286 



286 



IV • SPECIFIC VASCULAR INJURIES 



The insulting victor with disdain bestrode 
The prostrate prince and on his bosom trod; 
Then drew the weapon from his panting heart, 
The reeking fibers clinging to the dart; 
From the wide wound gushed out a stream of 

blood 
And the soul issues in the purple flood. 

Beall, Gasior, and Brickeret (1971) 



While originally recommending pericardio- 
centesis, Beall, in 1961, advocated aggressive 
use of thoracotomy and direct repair even in 
the emergency center (EC) . Currently, this is 
the most common approach, with pericar- 
diocentesis being used only rarely for tem- 
porary decompression of cardiac tamponade, 
if indicated, before direct cardiac repair. 



Attempts to treat wounds of the heart have 
been recorded as early as the first century ad, 
when Galen described therapy based on 
anatomic and experimental study through 
surgery of the pericardium. Ambroise Pare 
tried to dispel the general belief that cardiac 
injuries were usually fatal in his sixteenth 
century reports. Yet even in 1709, Boerhaave 
wrote that all heart wounds resulted in death. 

Larrey is often credited with the first suc- 
cessful decompression of the pericardium in 
1810 during the Napoleon wars, but it is only 
in the last 100 years that treatment of heart 
wounds has been repeatedly beneficial to the 
patient. Until 1896, pericardiocentesis, either 
alone or combined with phlebotomy, was the 
only method of surgical treatment for heart 
wounds. Those treated were usually small pen- 
etrating wounds of the pericardium. In 1896, 
however, Cappelen attempted to repair a heart 
by suturing a myocardial laceration. Although 
this operation failed, in the same year Rehn, 
in Frankfurt, was successful in relieving a 
cardiac tamponade and in suturing a knife 
wound of the heart. Rehn's accomplishment 
is generally regarded as the first actual repair 
of a heart wound, although in 1908, Matas 
reported that Farina had performed a similar 
operation, also in 1896. On September 14, 
1902, Dr. Hill of Montgomery, Alabama, 
became the first American physician to suc- 
cessfully repair a cardiac injury. 

Thus it is interesting that H.M. Sherman in 
1902 noted, "The road to the heart is only 2 
to 3 cm in a direct line, but it has taken surgery 
nearly 2400 years to travel it," Considerable 
controversy continued regarding the best 
approach in managing penetrating cardiac 
trauma. In 1943, Blalock and Ravitch still 
advocated pericardiocentesis as a form of 
definitive treatment for cardiac tamponade 
secondary to penetrating wounds of the heart. 



INCIDENCE 



In 1908, Matas noted that there had been 
160 reported cases of heart wounds after the 
operations of 1896. Recent reports show an 
increasing incidence of recognized cardiac 
trauma. The greater number of gunshot 
wounds in many urban centers is associated 
with the rise in cases of penetrating heart 
wounds, and true blunt cardiac trauma is 
associated with high-speed transportation. 

Parmley, Mattingly, andManion (1958) eval- 
uated 456 postmortem cases of penetrating 
wounds of the heart and aorta but stressed 
that the true incidence of cardiac trauma had 
not been established. Assessing numbers is 
complicated by the high early mortality rate 
of patients with these injuries. Isaacs (1959) 
reported, for instance, that from 1937 to 1959, 
more than 50% of the 133 patients were dead 
on arrival at Johns Hopkins Hospital. Only 86 
of the 459 patients analyzed by Sugg (1968) 
arrived alive in the EC of Parkland Hospital 
in Dallas. 

Other reports of note were those of 
Griswold and Drye (1954), who found 108 
cardiac wounds at Louisville General Hospi- 
tal in 20 years (1933 to 1953); Naclerio 
(1964) , who recorded 249 penetrating wounds 
in 13years (1950 to 1963); Wilson and Bassett 
(1966), who saw 200 patients and 205 wounds 
in 16.5 years (1949 to 1965); and Beall and 
colleagues (1972) who had 269 patients with 
penetrating cardiac injuries in Houston within 
20 years (1951 to 1971). Similarly, in Atlanta, 
Symbas, Harlaftis, and Waldo (1976) treated 
102 patients with penetrating cardiac heart 
wounds, between 1964 and 1974. In 1997, Wall 
and Mattox reported 711 heart injuries over 
a 30-year period in Houston, of which 60 were 
complex. 



chl5.qxd 4/16/04 3:41PM Page 287 



15 • WOUNDS OF THE HEART 



287 



The true incidence of cardiac trauma in the 
military experience is difficult to ascertain. 
On the battlefield, many cardiac wounds are 
immediately fatal. This is emphasized by one 
of the typical case reports from World War I. 
Dixon and McEwan (1916) reported one 
wounded heart in a series of 123 wounds of 
the thorax. These authors proclaimed, "prob- 
ably nearly all cardiac wounds produced 
death from hemorrhage too quickly to allow 
the patients being removed alive even to a 
short distance from the battlefield." Interest 
in wounds of the heart increased greatly 
during World War II. Harken (1946) reported 
a unique experience in removing foreign 
bodies from the heart and adjacent major 
vessels in 134 patients. 

There was one major report of injuries to 
the heart during the Korean conflict. Valle 
(1955) reported an incidence of 4.2% of 
injuries to the heart and mediastinum: 117 
injuries in a group of 2811 chest casualties 
treated at Tokyo Army Hospital from August 
1950 to March 1953. In this group, however, 
there were only 19 cases of foreign bodies in 
the heart and 42 pericardial effusions. The 
remainder of the injuries were to the medi- 
astinum and structures adjacent to the heart. 

Cardiac trauma during the Vietnam War 
has not been completely documented. 
Gielchinsky and McNamara (1970) reported 
10 heart injuries at the 24th Evacuation Hos- 
pital, an incidence of 2.8%. The records of 
nearly 120 patients with cardiac wounds in 
Vietnam are included in the long-term follow- 
up effort in the Vietnam Vascular Registry. 
Specifically, details of 96 cardiac injuries were 
evaluated by Geer and Rich (1972). Most of 
these injuries occurred between 1968 and 
1970. At least 21 different surgical facilities 
participated in the care of patients with 
cardiac injuries (Table 15-1). 



CLINICAL PATHOLOGY 



Penetrating wounds of the pericardium and/ 
or the myocardium caused by sharp instru- 
ments or low-velocity missiles are the most 
frequent types of injuries reported. Recent re- 
ports emphasize the vulnerability of the right 



TABLE 15-1 

ETIOLOGY OF CARDIAC TRAUMA 
IN VIETNAM 



Wounding 
Agent 



No. of Patients 



Deaths 



Fragment 


71 


74.0 


7 


Gunshot 


11 


11.5 


1 


Flechette 


3 


3.1 





Stab 


3 


3.1 





Unknown 


8 


8.3 


2 


Total 


96 


100 


10 



From T.M. Geer and N.M. Rich, Vietnam Vascular Registry, 
unpublished data, 1972. 

ventricle because of its anterior location. Our 
service found that the site of injury among 
patients with penetrating wounds of the peri- 
cardium was the right ventricle in 40% of 
patients, the left ventricle in 40% of patients, 
the right atrium in 24%, and the left atrium 
in3% (multiple injuries included) (Fig. 15-1). 

The World War II combat experience as 
described by Samson (1948) varies somewhat 
in that the left ventricle was involved more 
often than the right ventricle. This is the excep- 
tion, however, because the location of cardiac 
wounds in the Vietnam experience again 
emphasizes the predominance of wounds of 
the right ventricle (Table 15-2). As alluded 
to earlier, most of these wounds are pene- 
trating wounds. 

True blunt cardiac trauma usually results 
in diffuse contusion of the myocardium. 
However, the extent of cardiac injuries sec- 
ondary to blunt trauma to the chest may range 

TABLE 15-2 

CARDIAC TRAUMA IN VIETNAM: 
LOCATION OF WOUNDS 



Site 


No. 




Right ventricle 


40 


44.9 


Left ventricle 


22 


24.7 


Right atrium 


7 


7.9 


Left atrium 


5 


5.6 


Unknown 


15 


16.9 


Total 


89 


100 



From T.M. Geer and N.M. Rich, Vietnam Vascular Registry, 
unpublished data, 1972. 



chl5.qxd 4/16/04 3:41PM Page 288 



288 



IV • SPECIFIC VASCULAR INJURIES 




©Baylor College of Medicine 1997 

■ FIGURE 1 5-1 

Distribution of injuries to the four chambers of 
the heart (multiple injuries included). The 
posterior nature of the relatively protected left 
atrium probably accounts for its low incidence 
of injury. ■ 



from minor subepicardial or subendocardial 
hemorrhage to actual rupture of the 
myocardium. When there is sufficient force 
involved in a nonpenetrating injury to cause 
actual cardiac laceration, a fatal outcome fre- 
quently occurs. Patients who have had blunt 
trauma to the chest with cardiac trauma of 
varying degrees may have electrocardio- 
graphic changes, dysrhythmias, cardiac failure, 
cardiac tamponade, or hemothorax. Cardiac 
injuries from blunt trauma to the chest wall 
may or may not be associated with rib frac- 
tures or obvious chest wall deformity. When 
Parmley and colleagues (1958) reviewed 546 
autopsies in patients who had nonpenetrat- 
ing traumatic cardiac injuries, they found that 
353 of the 546 patients died of rupture of the 



heart. Of these 353 patients, 106 had multi- 
ple chamber ruptures. 

In addition to the injuries of the myocar- 
dial surface, other more unusual types of 
injuries can be seen to the valves, the inter- 
ventricular or interatrial septum, the coronary 
vessels, and the conduction system of the 
heart. Representative case reports of these 
unusual injuries include the removal of a wire 
lodged in the interventricular septum by 
Kleinsasser (1961); two patients with pene- 
trating wounds of cardiac valves, one with 
mitral insufficiency and the other with tri- 
cuspid insufficiency (Pate and Richardson, 
1969); three patients with intracardiac lesions 
including an aortic right ventricular fistula 
(Hardy and Timmis, 1969) ; and coronary arte- 
rial injuries (Tector and colleagues, 1973). 
Patients have developed left ventricular 
aneurysms after penetrating wounds, as 
reported in the civilian experience by Kakos 
and colleagues (1971) and in the military 
experience by Aronstam and colleagues 
(1970). There have been, in addition, more 
recent series by Demetriades (1990), 
Thandroyen (1981), and Wall (1997). In the 
later, 60 patients had complex heart wounds 
out of a total of 711 into the hemothorax. 

Associated pathology frequently accompa- 
nies cardiac wounds. This is emphasized by 
the report of Ricks and colleagues, who found 
that concomitant organ injury was associated 
with a striking rise in the mortality from 12% 
when there was injury of one associated organ 
to 69% with two or more associated organ 
injuries in their 31 patients with gunshot 
wounds of the heart. All but 1 of the 31 patients 
had one or both lungs injured together with 
the associated cardiac wound. Sugg and col- 
leagues found that 30 survivors of penetrat- 
ing heart wounds had no associated injuries. 
However, 33 patients who survived penetrat- 
ing wounds of the heart had a total of 84 asso- 
ciated injuries. 



PATHOPHYSIOLOGY 

Injuries to the heart can be divided into 
simple and complex. Simple injuries to the 
myocardium that result in bleeding from 



chl5.qxd 4/16/04 3:41PM Page 289 



15 • WOUNDS OF THE HEART 



289 



an injured chamber can present two ways. If 
the injury through the pericardium is so small 
that the bleeding is contained, tamponade 
physiology results. The pericardium does not 
distend acutely and can prevent the passive 
filling of the heart. Thus the patient essen- 
tially has an empty beating heart. If the injury 
through the pericardium is large, the patient 
may exsanguinate either externally or into the 
hemithorax. How much blood is in the left 
pleural cavity is often helpful to note during 
an empiric exploration for cardiac injury 
because the approach may be altered based 
on whether exsanguination or tamponade 
physiology is present. Most cardiac injuries are 
simple lacerations and can be managed with 
direct repair. However, complex cardiac 
injuries that involve the coronary arteries, 
cardiac valves, subvalvular apparatus, or the 
cardiac septum, though rare, present a dif- 
ferent challenge. Injuries to the coronary 
arteries can result in an area of ischemic 
myocardium. Treatment options are based on 
the distribution and amount of ischemic 
myocardium at risk. Injuries to the atrioven- 
tricular valves often result in regurgitation, 
which is commonly diagnosed postopera- 
tively when a new murmur is noticed. Signif- 
icant injury to the aortic valve is not well 
tolerated in the acutely hypotensive patient, 
and most of these patients die before arrival 
at the hospital, so they are rarely seen. Cardiac 
septal injuries from penetrating trauma often 
initially are small and diagnosed postopera- 
tively as a new murmur. Thus the common 
scenario is that many of the valvular and septal 
injuries are detected postoperatively after 
the acutely bleeding cardiac injury is con- 
trolled and are repaired subacutely at a later 
operation. 



PREHOSPITAL MANAGEMENT 



These patients most commonly present with 
a pattern of injury of penetrating trauma with 
proximity to the heart with either hypovolemia 
or tamponade physiology. Early consideration 
of the possibility of a cardiac injury with appro- 
priate transport to a center that can manage 
it may be lifesaving. If the patient has a sys- 



tolic blood pressure more than 80 mm Hg and 
is awake, ancillary measures to artificially 
elevate the blood pressure may not be helpful. 
Thus time should not be wasted on large- 
volume crystalloid resuscitation or the place- 
ment of pneumatic antishock trousers. In a 
patient in extremis, endotracheal intubation 
to control ventilation and maximally oxy- 
genate the remaining circulating blood may 
be one of the few efficacious prehospital 
maneuvers. 



EMERGENCY CENTER 

The diagnosis of a cardiac injury in the EC 
is based on a high index of suspicion. A pen- 
etrating injury in the area of the middle third 
of the chest between the nipples laterally and 
from the xiphoid to the sternal notch verti- 
cally is a common presentation. The patients 
often present in extremis and the cardiac 
injury is diagnosed on empiric exploration 
during EC thoracotomy. Low-energy mecha- 
nisms such as stab wounds can be problem- 
atic because tamponade physiology may not 
immediately develop. Awake patients usually 
have a profound anxiety. There are also 
reports of tamponade manifesting 2 to 3 days 
after the injury. Muffled heart sounds, dis- 
tended neck veins, and hypotension are clas- 
sically described. However, distended neck 
veins and an elevated central venous pressure 
are common in the anxious patient, and 
muffled heart sounds are difficult to detect 
in the noisy EC. Few diagnostic studies are 
usually needed and only delay transfer to the 
operating room for definitive therapy. The 
problematic patients are the ones who present 
hemodynamically stable with no signs of tam- 
ponade that are being investigated for prox- 
imity. These patients most benefit from 
monitoring and further investigation. 

Chest radiographs are commonly obtained. 
This is often unhelpful for the diagnosis of 
cardiac injury because these patients usually 
have a normal mediastinum. Their primary 
efficacy is to detect a pneumothorax or hemo- 
thorax. Wound clips marking entrance and 
exitwounds can be helpful, although missiles 
often do not follow straight lines between these 



chl5.qxd 4/16/04 3:41PM Page 290 



290 



IV • SPECIFIC VASCULAR INJURIES 



clips. Unexplained missile trajectories can be 
problematic and may represent either a 
missile bouncing off bony structures or an 
intravascular missile that has embolized. 

One diagnostic procedure that has had a 
significant impact on the diagnosis of cardiac 
injury has been ultrasound in the EC by the 
surgical team. Blood around the heart may 
be readily seen and diagnosed before the 
hemodynamic affects of tamponade. In skilled 
hands and the appropriate body habitus, the 
use of ultrasound has probably superseded 
other diagnostic entities such as central venous 
pressure monitoring and subxiphoid peri- 
cardial window. Formal echocardiography 
(either transthoracic or transesophageal) has 
little use in the hypotensive patient and only 
delays therapy. Their primary use may be as 
a second study or as a follow-up study after 
operative repair of a cardiac injury to docu- 
ment wall motion, septal integrity, and valvu- 
lar function. 



EMERGENCY CENTER 
PROCEDURES 

A patient with a penetrating wound to the 
chest often requires a tube thoracostomy for 
a concomitant hemothorax or pneumo- 
thorax. In the hypotensive patient with a 
suspected cardiacwound, an empiric tube tho- 
racostomy may be used to rule out a tension 
pneumothorax, which can present in a similar 
manner. Chest tubes should be placed no 
lower than the nipple level (to avoid the 
diaphragm) in the midaxillary line directing 
the tube posteriorly. After developing a tunnel 
and dividing intercostal muscles, the pleural 
space should be entered bluntly with the 
finger to avoid injuring the lung or the 
heart. During tube thoracostomy, the lung, 
diaphragm, and pericardium should be pal- 
pated. This is often referred to as a digital 
thoracotomy. Before the wide use of ultra- 
sound, balloting the pericardium could be 
used to detect a tamponade and provide an 
indication for operation. In a patient with 
suspected stab wounds to the heart, the tube 
thoracostomy incision is often placed slightly 
more anterior so the apex of the heart can 



be more readily palpated. Balloting the heart 
to detect tamponade is a subtle maneuver 
and should be done during each tube 
thoracostomy. 

Pericardiocentesis is often recommended 
in some resuscitation courses to temporize 
tamponade. Unfortunately, it is an unreliable 
procedure that often results in significant 
iatrogenic injuries. Even when a catheter is 
successfully placed, the clotted blood is unre- 
liably removed and may result in a false sense 
of security. Thus with the availability of ultra- 
sound, the use of pericardiocentesis as a diag- 
nostic maneuver has practically disappeared. 
At best, it may be a temporizing maneuver en 
route to the operating room. 

EC thoracotomy is one of the original 
damage control procedures in surgery. Many 
patients with cardiac injuries are often pre- 
morbid on arrival and will not survive the trip 
to the operating room. Using EC thoracotomy 
to bring techniques of definitive care to the 
EC has resulted in survivors. The primary 
thrust of EC thoracotomy for cardiac injuries 
is accessing the heart, relieving the tampon- 
ade, and controlling bleeding. Once this is 
accomplished, the patient can be moved to 
the operating room for completion of the 
procedure. 

The EC thoracotomy is performed with the 
patient supine after abducting the left arm. 
The incision is made immediately below the 
nipple in the male patient or beneath the 
breast tracking up to the fourth intercostal 
space in the female patient. The incision is 
made from the sternum to the posterior axil- 
lary line following the rib. Intercostal muscles 
are divided entering the chest in one area and 
the intercostal incision extended with the scis- 
sors. The rib retractor is placed with the rack 
toward the table and the retractor widely 
opened. The pleural cavity is inspected for the 
amount of blood to determine exsanguina- 
tion versus tamponade and the pericardium 
inspected. If tamponade is present and the 
heart is still beating, the pericardium is 
grasped between clamps anterior to the 
phrenic nerve and the pericardium is opened 
with the scissors. This is extended superiorly 
and inferiorly evacuating the clot and may 
result in a return of cardiac output. The heart 
is brought into the left side of the chest and 



chl5.qxd 4/16/04 3:41PM Page 291 



15 • WOUNDS OF THE HEART 



291 



inspected for injuries. For injuries to the right 
side of the heart, the incision may need to be 
extended across the chest with a Gigli saw, 
Lebsche knife, or sternal saw. If relief of tam- 
ponade does not result in return of perfusion, 
the aorta is cross clamped with an aortic clamp 
just distal to the left subclavian artery, being 
careful to avoid the esophagus. 

Immediate control of the injury is obtained 
with the surgeon'sfinger. A4-0 polypropylene 
suture on a large needle is then used to rapidly 
close the laceration in a gentle running 
fashion. The suture may be tied by an assis- 
tant while the surgeon continues to hold the 
heart. Because the incidence of needle stick 
during EC cardiorrhaphy approaches more 
than 30% , the skin stapler has often been used 
to achieve rapid vascular control and mini- 
mize the incidence of injury to the surgical 
team (Fig. 15-2) . After repair, warm saline is 
poured on the heart and the patient is rapidly 
transferred to the operating room for defin- 
itive repair. Pitfalls during the EC thoracotomy 
involve taking too long to perform it, not 
making a large enough incision initially, injur- 
ing the heart and lung while opening the 



chest, and injuring segmental vessels or the 
esophagus during aortic cross clamping. The 
outcomes of EC thoracotomy hinge on patient 
selection. A significant number of patients who 
have signs of life after an isolated stab wound 
to the heart can be salvaged. Recent data by 
Moore suggest that if a sustainable blood pres- 
sure is not obtained in the EC, further efforts 
in the operating room may be futile. 



OPERATIVE MANAGEMENT OF 
CARDIAC INJURIES 



Incisions 

For abdominal trauma, the midline laparo- 
tomy offers almost universal exposure. 
However, there are multiple incisions that can 
be made to manage chest trauma. The two 
most common incisions employed to manage 
cardiac injuries are left anterolateral thora- 
cotomy with possible extension across the 
sternum or median sternotomy. Each incision 
has advantages and disadvantages. The 




© Baylor College of Medicine 1 997 



■ FIGURE 1 5-2 

Acute management of cardiac 
injuries. A, Laceration of the left 
ventricle. B, Continuous fine 
polypropylene suture placed 
for both repair and hemostasis. 
C, A Foley balloon catheter can 
be useful to achieve 
hemostasis in the beating heart 
before placement of sutures. D, 
Because of the high incidence 
of needle stick during 
cardiorrhaphy, the skin stapler 
may be useful acutely in the 
emergency center for initial 
control. ■ 



chl5.qxd 4/16/04 3:41PM Page 292 



292 



IV • SPECIFIC VASCULAR INJURIES 



median sternotomy, the standard elective 
cardiac incision, is a midline incision that is 
relatively bloodless. Though easy to perform, 
it has the disadvantage of being difficult to 
clamp the aorta for resuscitation. In addition, 
efforts to retract the heart to repair a poste- 
rior injury may result in intractable ventricu- 
lar fibrillation in the cold, irritable injured 
heart. In many centers the left anterolateral 
thoracotomy is the incision of choice for 
cardiac trauma. It is easily made with a 
minimal number of instruments and offers 
excellent exposure of the heart and descend- 
ing thoracic aorta. In most cases the patient 
is positioned supine with the arms out so they 
are available to the anesthesia service. For 
right-sided injuries, a transsternal extension 
into the opposite chest is helpful. Left antero- 
lateral thoracotomy has the additional advan- 
tage in that injuries to the posterior heart can 
be more readily visualized from this viewpoint 
with less retraction and manipulation. 



Aortic Occlusion 

Occlusion of the descending thoracic aorta 
may be helpful as a resuscitative maneuver. 
The aorta is cross clamped just distal to the 
left subclavian origin, being careful to avoid 
injury to the esophagus. It is useful to dissect 
anterior and posterior to the aorta and actu- 
ally encircle it with a finger before applying 
the clamp to ensure accurate positioning. This 
ensures that blood is preferentially diverted 
to the brain and heart. As the patient is resus- 
citated, the aortic cross clamp can be gradu- 
ally weaned and removed. It is extremely 
common after hemorrhage is controlled for 
patients to be over-resuscitated and the heart 
may become distended. One technique to 
decompress the heart is to remove the aortic 
cross clamp and vent the heart into the sys- 
temic circulation momentarily. 



Cardiac Manipulation 

The cold, empty injured heart can be 
extremely irritable. Even minor manipulation 
can cause significant dysrhythmias. Unfortu- 
nately, in the cold patient, ventricular 



fibrillation is often refractory. Thus all mani- 
pulations of the heart should be gentle and 
retraction minimized. 

It has been learned from elective cardiac 
surgery that significant retraction of the heart 
can be performed if done slowly and in such 
a manner that the heart can fill. Because the 
filling of the heart is passive, it is important 
not to compress the cardiac chambers as it is 
retracted. Gentle manipulation with a drag- 
ging motion can often allow one to completely 
invert the heart out of the chest and still main- 
tain cardiac output while allowing access to 
the posterior aspect of the left atrium. If the 
heart is not beating in an organized rhythm, 
manual cardiac compression may be required. 
This is best performed with a two-handed tech- 
nique gently alternating between allowing the 
heart to fill and compressing it from apex to 
base. Cardiac compressions often are per- 
formed with one hand by some doctors. Unfor- 
tunately, the distended right ventricle can be 
extremely thin walled and may be injured by 
the thumb or fingers. In the bradycardic heart, 
manual compression may serve to prevent it 
from becoming distended by manually emp- 
tying the chambers. Sutured temporary epi- 
cardial leads can be helpful also. 



Hemorrhage Control 

Some injuries may be extremely difficult to 
manage because of massive hemorrhage. 
These may often be managed by cardiac inflow 
occlusion to empty the heart before repair. 
The superior and inferior vena cava can be 
pinched between the fingers or clamped with 
vascular clamps and the heart allowed to 
empty. The injury can then be visualized and 
the repair performed. If after a left-sided injury 
is repaired there is concern about intracar- 
diac air, the patient should be placed in a head- 
down position and the ascending aorta vented 
while a cardiac rhythm is restored. Inflow 
occlusion allows a short interval of an empty 
beating heart and the repair must be accom- 
plished before its arrest. 

Inflow occlusion can also be extremely 
useful in repairing the distending heart or 
repairing a thin soft aorta. To avoid placing 
undo tension as the stitches are tied down, 



ohl5.qxd 4/16/04 3:41PM Page 293 



15 • WOUNDS OF THE HEART 



293 



inflow occlusion can be accomplished to tem- 
porarily decrease the blood pressure while 
tying down the repair. 

The right atrium is a commonly injured 
cardiac structure. Upon diagnosis of a right 
atrial injury, it is helpful to extend the inci- 
sion across the sternum for better visualiza- 
tion. Initially, the injury is controlled with the 
finger. Other adjuncts that may be useful are 
a partial occluding clamp or a Foley catheter 
placed through the injury for temporary 
control (Fig. 15-2) . A 4-0 polypropylene or 5- 
polypropylene suture is then used in a simple 
continuous manner to close the injury and 
effect repair. The repair of atrial injuries 
close to the superior vena cava-right atrial 
appendage junction may involve the sinoatrial 
node and result in dysrhythmias. 

Injuries to the left atrium can be difficult 
to manage. The heart is gently and slowly 
retracted while avoiding compression and per- 
mitting passive filling of the heart. All instru- 
ments and sutures should be prepared before 
retracting the heart, and sutures can often be 
placed in a back-hand manner. It may be 
helpful for the surgeon to retract with one 
hand and sew with the other so the cardiac 
performance can be monitored and the heart 
returned to the chest before arrest. It may take 
multiple episodes of suturing to close these 
injuries. As described earlier, inflow occlusion 
can decrease bleeding through a posterior 
injury so it can be more readily visualized. 
Repairs in the empty heart should raise the 
surgeon's suspicion of intracardiac air, and 
the patient can be placed head down and the 
aorta vented before restitution of inflow. 

Whereas the left ventricle is a thick mus- 
cular structure, the anterior wall of the right 
ventricle is only approximately 5 mm thick. 
Repair of the right ventricle can be initially 
managed with digital pressure, followed by 
repair with a fine suture. Pledgeted sutures 
are not routinely needed though may be used 
if the repair fails or the heart distends. Avoid- 
ing over-resuscitation and inflow occlusion 
may be helpful adjuncts in these repairs. 
Injuries to the left ventricle are performed in 
a similar manner, although these are often on 
the posterior surface and require some 
measure of retraction. If possible, the ventri- 
cle should be carefully inspected to identify 



injury to adjacent coronary arteries. It should 
be remembered that the heart is a relatively 
soft muscle and a gentle technique with a fine 
suture often gets better results. Again, initial 
control with a finger or a Foley balloon 
catheter to arrest hemorrhage is often helpful. 
With no intravenous access available, the Foley 
catheter can be connected to an intravenous 
catheter for direct infusion. It is our prefer- 
ence not to aggressively resuscitate the heart 
before repair because the empty bradycardic 
heart is ideal for the placement of sutures. 
Overzealous crystalloid resuscitation results 
in an overdistended heart that not only fails 
to hold stitches but also fails to beat well after 
repair. Administration of pressor drugs before 
repair results in a rapidly beating empty heart 
that is extremely difficult to sew. Multiple 
injuries and complex lacerations portend a 
poorer prognosis. 

Complex Injuries 

Anterior stab wounds often occur immedi- 
ately adjacent to the left anterior descending 
coronary artery. If the coronary artery is unin- 
jured, it is important to repair the laceration 
without compromising the coronary artery. 
Deep mattress sutures beneath the coronary 
artery will permit repair while avoiding the 
coronary artery. When a coronary artery is 
injured, decision making is guided by its loca- 
tion and the amount of myocardium at risk. 
Small secondary branches of the coronary 
arteries can usually be ligated. The patient can 
then be observed for dysrhythmias or the 
development of an akinetic area of the heart. 
If there is any concern, many will place a hor- 
izontal mattress suture and use a snare tourni- 
quet and observe the heart before tying the 
suture down. If a significant area of the heart 
becomes akinetic and fails, then coronary 
artery bypass may be indicated. If a small area 
of the heart becomes akinetic and cardiac 
function is borderline after ligation, the place- 
ment of an intra-aortic balloon pump may tem- 
porize the injury and the patient may be 
treated similar to those having had a small 
myocardial infarction. Though uncommon, 
most coronary artery injuries that require 
emergent bypass are proximal injuries of the 



chl5.qxd 4/16/04 3:41PM Page 294 



294 



IV • SPECIFIC VASCULAR INJURIES 



■ FIGURE 1 5-3 

Coronary artery bypass grafting 
for injury involving the left 
anterior descending coronary 
artery. A, Anterior stab wound 
to the chest. B, Injury involves 
the proximal left anterior 
descending coronary artery, 
resulting in a large area of 
ischemic myocardium. C, 
Hemostasis is initially obtained 
with continuous suture of the 
laceration. D, Coronary artery 
bypass from the ascending 
aorta to the distal left anterior 
ascending coronary artery 
using cardiopulmonary bypass. 
Fortunately, this is seldom 
needed in most patients. ■ 




©Baylor College ol Medicine 1997 



primary branches such as the left anterior 
descending or the right coronary artery (Fig. 
15-3). Although off-pump methods would 
seem attractive, these patients often require 
cardiopulmonary bypass to support the failing 
heart. Because this is an emergent lifesaving 
activity, the saphenous vein is most commonly 
used as the conduit. 



Cardiac Septal Injuries 

The most common presentation for cardiac 
septal injuries is when a new murmur is noted 
postoperatively in the intensive care unit. Most 
patients who survive cardiac repair who have 
a septal injury often have small injuries that 
may not be hemodynamically significant. The 
diagnosis is confirmed with echocardiography. 
A saturation run during catheterization of the 



right side of the heart may be diagnostic and 
can be used to calculate shunt fraction. If indi- 
cated, most of these patients undergo car- 
diopulmonary bypass in a subacute fashion 
often weeks after the initial injury. A shunt 
fraction of more than 2 : 1 is often used as an 
indication for surgery. Smaller injuries with 
smaller shunt fractions may be observed. The 
patient however should be counseled about 
the risk of endocarditis if they undergo other 
invasive procedures. 

These injuries are repaired using car- 
diopulmonary bypass usually via a median ster- 
notomy. For anterior stab wounds resulting 
in ventricular septal defects, the injury may 
be repaired through the previous myocardial 
repair. Although ventriculotomy is avoided in 
elective cardiac surgery, this area is often 
scarred from the initial injury and offers excel- 
lent exposure of the septal injury (Fig. 15-4) . 



chl5.qxd 4/16/04 3:41PM Page 295 



15 • WOUNDS OF THE HEART 



295 




©Baylor College ol Medicine 1997 



■ FIGURE 1 5-4 

Repair of traumatic ventricular 
septal defect. A, The acute 
management of the injury to the 
surface of the heart is with 
either continuous or pledgeted 
sutures controlling the 
hemorrhage. Later in the 
intensive care unit, a murmur is 
detected and the ventricular 
septal defect is diagnosed. B, 
These injuries are most 
commonly repaired subacutely 
often weeks after the initial 
injury. Because of the scarring 
from the initial injury, 
ventriculotomy can be 
performed through the original 
scar. C, This results in good 
visualization of the septal 
wound. These can be repaired 
with either (D) interrupted 
pledgeted sutures or (E) larger 
defects closed with a Dacron 
patch. ■ 



Knowledge of the conduction system of the 
heart and counseling the patient preopera- 
tively about the risk of conduction defects is 
helpful. Smaller defects can be closed pri- 
marily, but often the use of a prosthetic mate- 
rial such as Dacron may be required. Small 
ventricular septal defects located near the apex 
of the heart can be extremely difficult to local- 
ize and repair. Atrial septal defects can be 
approached via the standard incision in the 
right atrium. Again, knowledge of the path of 
the conduction system can help avoid iatro- 
genic injuries. 



Cardiac Valvular Injury 



esophageal echocardiography may be helpful 
to adequately visualize the subvalvular appa- 
ratus of the mitral valve. The injuries may 
involve the leaflets of the valve or the sub- 
valvular apparatus. Although it is often hoped 
that a simple repair can be performed, upon 
exploration, the valve often is found to be 
totally destroyed and in need of being 
replaced. Replacement is performed via 
median sternotomy and the standard 
approach using cardiopulmonary bypass. 
Injury to the right-sided heart valves is not 
common. The indications for operation are 
the same as those for elective cases. Choice of 
technique and prosthesis depends on the 
pathology encountered at exploration. 



Similar in presentation to septal injuries, 
most cardiac valvular injuries in survivors are 
detected as a new murmur postoperatively in 
the intensive care unit. They are usually 
injuries of the mitral or tricuspid valve and 
are evaluated with echocardiography. Trans- 



Intrapericardial Inferior Vena 
Cava Injury 

One injury that requires acute cardiopul- 
monary bypass is a posterior injury to the 



chl5.qxd 4/16/04 3:41PM Page 296 



296 



IV • SPECIFIC VASCULAR INJURIES 



■ FIGURE 1 5-5 

Unusual injury of posterior 
intrapericardial inferior vena 
cava. A, These are often from 
transaxial gunshot wounds. 6, 
The injury is extremely difficult 
to visualize and access. C, 
Cardiopulmonary bypass is 
instituted with cannulation of 
the superior vena cava directly 
and the inferior vena cava via 
the groin for venous drainage. 
D, Total cardiopulmonary 
bypass allows the right atrium 
to be opened and the injury 
repaired from within. E, Larger 
injuries may require the use 
of a Dacron or pericardial 
patch. ■ 




©Baylor College of M 



intrapericardial inferior vena cava. This area 
is extremely difficult to access, is a short vessel, 
and difficult to visualize posteriorly. One tech- 
nique available to address it is to cannulate 
the groin for cardiopulmonary bypass and 
place a superior vena caval cannula for total 
cardiopulmonary bypass. The superior vena 
cava is snared around the cannula and the 
inferior vena cava is clamped immediately 
above the liver. The injury is accessed by 
opening the right atrium and repairing it from 
inside. Though extremely uncommon, this is 
one of the few areas in which cardiopulmonary 
bypass may assist in managing cardiac injuries 
acutely (Fig. 15-5). 



SUMMARY 

Injuries to the heart have fascinated trauma 
surgeons for ages. As with many other injuries, 



a repair that was originally thought to be futile, 
with advances in technique, has resulted in 
significant salvage. Cardiac injuries can 
present as either tamponade or exsanguina- 
tion and can be classified as either simple or 
complex. Simple injuries primarily involve the 
myocardium, and complex injuries involve the 
coronary arteries, cardiac septa, and cardiac 
valves. The EC thoracotomy is one of the 
original damage-control procedures and 
has resulted in a significant salvage rate for 
patients with low-energy penetrating injuries 
to the heart. Ultrasound has significantly aided 
in the diagnostic accuracy of patients who 
present hemodynamically stable. Cardiac 
injuries are managed in the operating room 
via left anterolateral thoracotomy with exten- 
sion across the sternum. Cross clamping of 
the descending thoracic aorta could be a sig- 
nificant adjunct and various maneuvers such 
as digital compression, Foley catheter occlu- 
sion, stapling, and a partial occluding clamp 



chl5.qxd 4/16/04 3:41PM Page 297 



15 • WOUNDS OF THE HEART 



297 



may provide initial hemostasis. The avoidance 
of resuscitation until repair is complete, the 
avoidance of cyclic hyper-resuscitation and the 
avoidance of overdistention of the heart can 
result in improved outcomes. It is important 
to evaluate the patient postoperatively with 
physical examination and echocardiography 
to document wall motion and assess the 
patient for occult septal and valvular injuries. 
Most cardiac injuries can be managed by the 
trauma surgeon without any specialized 
cardiac technique. The need for cardiopul- 
monary bypass is extremely rare and is seen 
in fewer than 1 % of these patients. 



REFERENCES 

Surgical approach and initial management of 
patients with cardiac injuries 

Asensio JA, Stewart BM, Murray J, et al: Penetrat- 
ing cardiac injuries. Surg Clin North Am 
1996;76:685. 

Beall AC Jr, Ochsner JL, Morris GC, et al: Pene- 
trating wounds of the heart. J Trauma 1961 ;1:195. 

Ivatury RR, Shah PM, Ito K, et al: Emergency room 
thoracotomy for the resuscitation of patients with 
"fatal" penetrating injuries of the heart. Ann 
Thorac Surg 1981;32:377. 

Mattox KL, Beall AC, Jordan GL, et al: Car- 
diorrhaphy in the emergency center. J Thorac 
Cardiovasc Surg 1974;68:886. 

Diagnosis and management of complex cardiac 
injuries to the coronary arteries, septa, and 
valves 

Demetriades D, Charalambides C, Sareli P, 
Pantanowitz D: Late sequelae of penetrating 
cardiac injuries. Br J Surg 1990;77:813-814. 



Fallahnejad M, Kutty ACK, Wallace HW: Sec- 
ondary lesions of penetrating cardiac injuries: 
A frequent complication. Ann Surg 1980; 
191:228-233. 

Symbas PN, DiOrio DA, Tyras DH, et al: Penetrat- 
ing cardiac wounds: Significant residual and 
delayed sequelae. J Thorac Cardiovasc Surg 
1973;66:526-532. 

Wall MJ, Mattox KL, Chen C-D, Baldwin JC: Acute 
management of complex cardiac injuries. 
J Trauma 1997;42(5):905-912. 

Management of intracardiac injuries 

Asfaw I, Thorns NW, Arbulu A: Interventricular 
septal defects from penetrating injuries of the 
heart: A report of 12 cases and review of the lit- 
erature. J Thorac Cardiovasc Surg 1975;69:450- 

457. 

Espada R, Whisennand HH, Mattox KL, Beall AC 
Jr: Surgical management of penetrating injuries 
to the coronary arteries. Surgery 1975;78:755- 
760. 

Thandroyen FT, Matisonn RE: Penetrating thoracic 
trauma producing cardiac shunts. J Thorac Car- 
diovasc Surg 1981;81:569-573. 

Whisennand HH, Van Pelt SA, Beall AC Jr, et al: 
Surgical management of traumatic intracardiac 
injuries. Ann Thorac Surg 1979;28:530-536. 

Ventricular aneurysms after cardiac injury 

Aronstam EM, Strader LD, Geiger JP, Gomez AC: 
Traumatic left ventricular aneurysms. J Thorac 
Cardiovasc Surg 1970;59:239-242. 

Morales RA, Garcia F, Grover FL, Trinkle JK: 
Aneurysm of the left ventricle after repair of a 
penetrating injury. J Thorac Cardiovasc Surg 
1973;66:632-635. 



chl6.qxd 4/16/04 3:40 PM Page 299 




Injury to Abdominal Aorta and 
Visceral Arteries 

DAVID V. FELICIANO 



O GENERAL 
Incidence 
Pathophysiology 
Clinical Presentation 
Areas of Abdominal Vascular Injuries 
O OPERATION 

Supramesocolic Area of Zone 1 

Suprarenal Abdominal Aorta 

Celiac Trunk 

Superior Mesenteric Artery 

Renal Artery 
Inframesocolic Area of Zone 1 

Infrarenal Abdominal Aorta 
Zone 2 or Upper Lateral Retroperitoneum 

Renal Artery 
Complications 



299 



chl6.qxd 4/16/04 3:40 PM Page 300 



300 



IV • SPECIFIC VASCULAR INJURIES 



GENERAL 



Incidence 

The incidence of injuries to the abdominal 
aorta, celiac trunk or major branches, supe- 
rior mesenteric artery, and renal artery is sur- 
prisingly high in urban trauma centers in the 
United States. This is a reflection of rapid trans- 
port by prehospital emergency medical ser- 
vices and the large number of patients who 
are treated for penetrating wounds, parti- 
cularly those caused by low-velocity civilian 
handguns. 

In one recently published review from the 
Grady Memorial Hospital in Atlanta, Georgia, 
300 patients with 205 abdominal arterial and 
284 abdominal venous injuries were treated 
at laparotomy during a 10-year period. Of 
interest, the mechanism of injury was a pen- 
etrating wound in 86.7% of patients, with 
abdominal gunshot wounds (78%) account- 
ing for the majority. The group of patients 
with abdominal arterial injuries included 77 
(37.5%) with injuries to the abdominal aorta, 
18 (8.8%) with injuries to the renal artery, 16 
(7.8%) with injuries to the superior mesen- 
teric artery, and 16 with injuries to the celiac 
trunk or major branches. This group there- 
fore accounted for 62% of all abdominal 
arterial injuries treated. 

Another recently published review from the 
Los Angeles County Hospital described 302 
patients with 238 abdominal arterial and 266 
abdominal venous injuries that were treated 
at laparotomy during a 6-year period. Of inter- 
est, the mechanism of injury was a penetrat- 
ing wound in 88% of patients, with abdominal 
gunshot wounds (81%) accounting for the 
majority. Patients with injuries to the aorta, 
celiac trunk or major branches, superior 
mesenteric artery, and renal artery accounted 
for 57% of all abdominal arterial injuries 
treated. 

In contrast to the aforementioned reports, 
injuries to all abdominal vessels have been 
uncommon in reviews of military conflicts. 
This low incidence reflects the greater wound- 
ing power of high-velocity military weapons 
and the longer delays to definitive operation 
that occur in all war zones. In the report by 



DeBakey and Simeone of 2471 arterial injuries 
during World War II, only 49 (2%) occurred 
in the abdomen. In similar fashion, the report 
by Hughes of 304 arterial injuries from the 
Korean War included only 7 (2.3%) that 
occurred in the abdomen. Finally, the report 
by Rich and colleagues of 1000 arterial injuries 
treated in the Vietnam conflict described only 
29 (2.9%) involving abdominal vessels. 



Pathophysiology 

Penetrating injuries to the abdominal aorta 
or visceral branches most commonly cause 
lateral wall defects with intraperitoneal bleed- 
ing or expanding pulsatile retroperitoneal or 
mesenteric hematomas. Aless common injury 
is complete transection of a visceral artery with 
secondary bleeding, an expanding hematoma, 
or complete thrombosis of both ends of the 
vessel. On occasion, the track of a missile may 
be in proximity to a visceral vessel and cause 
a thrombosis because of disruption of the 
intima from a blast effect. The rarest injury 
related to a penetrating wound is the creation 
of an upper abdominal arteriovenous fistula 
involving the hepatic artery and portal vein, 
the superior mesenteric vessels, or the renal 
vessels. 

Bluntinjuries to the abdominal aorta orvis- 
ceral branches are most commonly caused by 
deceleration, a direct anterior crushing mech- 
anism (lap-type seatbelt), or a posterior blow 
to the spine. Deceleration or direct anterior 
blows have most commonly caused either 
thrombosis of the infrarenal abdominal aorta, 
superior mesenteric artery, or renal artery or 
lateral wall defects in the superior mesenteric 
artery at the base of the mesentery. Posterior 
blows to the spine have most commonly 
caused an intimal flap and secondary throm- 
bosis of the infrarenal abdominal aorta. 



Clinical Presentation 

In patients with either a penetrating or a 
blunt mechanism of injury, the clinical pre- 
sentation will depend on several factors. The 
first of these is the type of aortic or visceral 
arterial injury. In patients with defects in the 



chl6.qxd 4/16/04 3:40 PM Page 301 



16 • INJURY TO ABDOMINAL AORTA AND VISCERAL ARTERIES 



301 



lateral wall, hemorrhage will occur and lead 
to hypotension with or without peritonitis. 
With complete transection of a visceral vessel 
and hemorrhage, the presentations will be the 
same. Should transection lead to thrombosis 
of both ends of the visceral vessel, a rare event 
in my experience, only abdominal pain (supe- 
rior mesenteric artery) or hematuria (renal 
artery) may be present. The same presenta- 
tions along with ischemia of both lower 
extremities (abdominal aorta) would occur if 
blunt trauma, an intimal flap, and secondary 
arterial thrombosis were present. 

Clinical presentation is affected by the pres- 
ence or absence of retroperitoneal or mesen- 
teric tamponade, as well. In patients with 
defects in the lateral wall of the abdominal 
aorta or visceral arteries, retroperitoneal or 
mesenteric tamponade is the most common 
finding at a subsequent laparotomy. All 
patients who have arterial injuries and tam- 
ponade are still hypotensive at some point in 
the preoperative period — in the field, in the 
emergency center, or in the operating room 
as general anesthesia is initiated. In contrast 
to patients with abdominal venous injuries and 
tamponade, any improvement in blood pres- 
sure secondary to the infusion of crystalloid 
solutions and blood is transient. If retroperi- 
toneal or mesenteric tamponade does not 
occur and there is active hemorrhage into the 
peritoneal cavity, a confused or moribund 
patient with profound hypotension, clear- 
cut peritonitis, and a tight abdomen is the 
presentation. 



Areas of Abdominal 
Vascular Injuries 

As has been discussed in numerous other texts, 
it is often helpful to describe the approaches 
to abdominal vascular injuries in a "geographic 
zone" fashion. Zone 1 includes the midline 
retroperitoneum and base of the mesentery, 
zone 2 is the upper lateral retroperitoneum 
(renal vessels), and zone 3 is the pelvic 
retroperitoneum (iliac vessels). Because they 
are uncommon, injuries to the vessels in the 
porta hepatis or retrohepatic area are usually 
described separately, as well. 



Zone 1, the topic of discussion in this 
chapter, is best divided into suprameso colic and 
inframeso colic areas, because the operative 
approach is different for each, as is described. 
A midline supramesocolic area of hematoma 
or hemorrhage is likely to contain an injury 
to the suprarenal abdominal aorta, celiac 
trunk, proximal superior mesenteric artery, 
proximal renal artery, superior mesenteric 
vein, or obviously the pancreas. A midline 
inframesocolic area of hematoma or 
hemorrhage is likely to contain an injury to 
the infrarenal abdominal aorta, left renal vein, 
or inferior vena cava. 

As a general rule, all hematomas in zone 1 
(either supramesocolic or inframesocolic) 
from either penetrating or blunt trauma are 
opened by the surgeon using techniques to 
be described. Hematomas from penetrating 
wounds in zones 2, 3, and in the porta hepatis 
are opened, as well. In contrast, hematomas 
from blunt trauma that are located in zones 
2 and 3 or in the retrohepatic area are opened 
only if they are pulsatile, expanding rapidly, 
or have already ruptured. 



OPERATION 

Supramesocolic Area of Zone 1 

SUPRARENAL ABDOMINAL AORTA 

The presence of a hematoma in the midline 
supramesocolic area will usually give the 
surgeon time to obtain proximal control of 
the supraceliac abdominal aorta. Such a 
hematoma is more likely to be present when 
the injury to the abdominal aorta is in the 
diaphragviatic aorta (in the aortic hiatus, itself) 
rather than in the visceral aorta (origins of 
visceral arteries) (Fig. 16-1). 

The left-sided medial mobilization maneu- 
ver is the preferred operative approach. This 
maneuver includes division of the retroperi- 
toneal attachments and reflection of the left 
colon, left kidney, spleen, tail of the pancreas, 
and fundus of the stomach to the midline (Fig. 
16-2) . The advantage of this technique is that 
it allows visualization of the entire abdominal 
aorta from the aortic hiatus of the diaphragm 



chl6.qxd 4/16/04 3:40 PM Page 302 



302 IV • SPECIFIC VASCULAR INJURIES 

Divisions of Suprarenal Aorta 



Diaphragmatic 
aorta 



Visceral 
aorta 




© Baylor College of Medicine 1987 



■ FIGURE 16-1 

Penetrating wounds of the diaphragmatic 
abdominal aorta may be tamponaded by 
muscle fibers of the aortic hiatus, and wounds 
of the visceral abdominal aorta are obviously 
more complex. (From Baylor College of 
Medicine, 1987.) ■ 



to the aortic bifurcation (Fig. 16-3). Dis- 
advantages include the time required to 
complete the maneuver (4 to 5 minutes in 
inexperienced hands); risk of damage to the 
spleen, left kidney, or posterior left renal artery 



Plane of Dissection 




■ FIGURE 16-2 

Left medial mobilization maneuver is initiated 
by dividing lateral retroperitoneal attachments 
of left colon, left kidney, spleen, tail of 
pancreas, and fundus of stomach. (From 
Feliciano DV: Truncal vascular trauma. In 
Callow AD, Ernst CB [eds]: Vascular Surgery. 
Theory and Practice. Stamford, Conn, Appleton 
& Lange, 1995, pp 1059-1085.) ■ 




■ FIGURE 16-3 

Completion of left medial mobilization 
maneuver with all left-sided intra-abdominal 
viscera elevated to the midline. ■ 



during the maneuver; and anatomic distor- 
tion that results when the left kidney is rotated 
anteriorly. One alternative is to leave the left 
kidney in its fossa, thereby eliminating poten- 
tial damage to or distortion resulting from 
rotation of this structure. 

There are three significant obstacles to com- 
pleting the maneuver once the viscera are 
mobilized. These include the length of the 
hiatal muscle fibers surrounding the diaphrag- 
matic aorta, the dense nature of the celiac 
plexus of nerves connecting the right and left 
celiac ganglia, and the thickened lymphatic 
tissue around the aorta at this level. Although 
it is possible to peel the hiatal muscle fibers 
away and dissect through the celiac plexus of 
nerves and lymphatics, these maneuvers may 
be too time consuming in profoundly hypoten- 
sive patients (Fig. 16-4). It is much easier to 
transect the left crus of the aortic hiatus of 
the diaphragm at the 2-o 'clock position to 
allow for exposure of the distal descending 
thoracic aorta above the hiatus. With the distal 
descending thoracic aorta or abdominal aorta 
in the hiatus exposed, the supraceliac aortic 
clamp can be applied without difficulty. 

An alternative approach in the patient with 
a supramesocolic hematoma is to perform an 
extensive Kocher maneuver, elevate the Cloop 
of the duodenum and the head of the pan- 
creas to the left, and incise the retroperitoneal 
tissue to the left of the inferior vena cava. This 



chl6.qxd 4/16/04 3:40 PM Page 303 



16 • INJURY TO ABDOMINAL AORTA AND VISCERAL ARTERIES 



303 



Superior mesenteric a 



Left renal a 




» Baylor College of Med I 



■ FIGURE 1 6-4 

View of suprarenal abdominal aorta and major 
branches after left-sided medial mobilization 
maneuver and removal of all neural and 
lymphatic tissue. Note the fold in the visceral 
abdominal aorta created by mobilization of the 
left kidney and renal artery. (From Baylor 
College of Medicine, 1986.) ■ 



will expose the suprarenal abdominal aorta 
between the celiac axis and the superior 
mesenteric artery. The disadvantage of this 
approach is that the exposure obtained is 
below the level of any wounds to the 
supraceliac aorta in the hiatus. 

If active hemorrhage is coming from the 
supramesocolic area of the abdominal aorta, 
the surgeon may obtain temporary control 
manually or with one of the aortic compres- 
sion devices (Fig. 16-5). If this compression 
prevents exposure and repair of the aortic 
injury, the next maneuver is to divide the lesser 
omentum manually, retract the stomach and 
esophagus to the left, and digitally separate 
the muscle fibers of the crura from the 
supraceliac aorta to obtain the same exposure 
as described for the left-sided medial mobi- 
lization maneuver, but anteriorly and more 
quickly. Distal control of the aorta in this loca- 
tion is awkward because of the presence of 
the visceral vessels. In young patients with 
injury confined to the supraceliac aorta, the 
celiac axis should be ligated and divided to 
allow for more space for the distal aortic clamp 
and subsequent vascular repair. 

With small perforating wounds to the aorta 
at this level, lateral aortorrhaphy with 3-0 or 
4-0 polypropylene suture is preferred. If two 
small perforations are adjacent to one another, 




© Baylor College of Medicine 1 987 



■ FIGURE 16-5 

Aortic compression device applied to 
supraceliac abdominal aorta for temporary 
proximal control superior to wound in visceral 
abdominal aorta. (From Baylor College of 
Medicine, 1987.) ■ 



they should be connected and the defect 
closed in a transverse fashion with the 
polypropylene suture. When closure of the 
perforation (s) results in significant narrow- 
ing or if a portion of the aortic wall is missing, 
patch aortoplasty with polytetrafluoroethylene 
(PTFE) is indicated in the patient who is 
hemodynamically stable without hypothermia, 
significant acidosis, or an intraoperative 
coagulopathy. The other option is to resect a 
short segment of the injured aorta and 
perform an end-to-end anastomosis. This is 
difficult because of the limited mobility of both 
ends of the aorta at this level. 

On rare occasions, patients with extensive 
injuries to the diaphragmatic or supraceliac 
aorta will require insertion of a synthetic vas- 
cular conduit or spiral graft after resection of 
the area of injury. Many of these patients have 
associated gastric, enteric, or colonic injuries. 
Therefore, much concern has been expressed 
about placing a synthetic conduit, such as a 



chl6.qxd 4/16/04 3:40 PM Page 304 



304 



IV • SPECIFIC VASCULAR INJURIES 



12-, 14-, or 16-mm woven Dacron, albumin- 
coated Dacron, or PTFE prosthesis, in the 
aorta. The data in the American literature 
describing young patients with injuries to 
nondiseased abdominal aortas do not support 
the concern about infection occurring in 
Dacron interposition grafts, and there are few 
data relating to the use of PTFE grafts in pen- 
etrating trauma to the abdominal aorta. 
Despite the available data, some authors con- 
tinue to recommend an extra-anatomic bypass 
when injury to the abdominal aorta would 
require replacement with a conduit in the 
presence of gastrointestinal contamination. 
To lower the risk of infection in a prosthetic 
patch or graft inserted into the abdominal 
aorta at any level, one should not perform 
repairs of the intestine and the aorta simul- 
taneously. Once the perforated bowel with 
occlusion clamps applied has been packed 
away and the surgeon has changed gloves, the 
aortic prosthesis is sewn in place with 3-0 or 
4-0 polypropylene suture. After appropriate 
flushing of both ends of the aorta and removal 
of the distal aortic clamp to flush air out from 
the graft, the proximal aortic clamp should 
be removed very slowly as the anesthesiolo- 
gist rapidly infuses fluids. If a long aortic clamp 
time has been necessary, the prophylactic 
administration of intravenous bicarbonate is 
indicated to reverse the "washout" acidosis 



from the previously ischemic lower extremi- 
ties. The retroperitoneum is then copiously 
irrigated with an antibiotic solution and 
closed in a watertight fashion with an 
absorbable suture. At this point, the injuries 
to the gastrointestinal tract are repaired. 

The survival rate of patients with injuries 
to the suprarenal abdominal aorta is approx- 
imately 30% (Table 16-1). Combined injuries 
to the suprarenal aorta and inferior vena cava 
had a 100% mortality rate in the large series 
from the Ben Taub General Hospital in 1987. 



CELIAC TRUNK 

When branches of the celiac trunk are 
injured, they are often difficult to repair 
because of the surrounding dense neural and 
lymphatic tissue and the small size of the vessels 
in a patient in shock with secondary vaso- 
constriction. Therefore major injuries to 
either the left gastric or the proximal splenic 
artery should be ligated. The common hepatic 
artery may have a larger diameter than the 
other two vessels, and an injury to this vessel 
may occasionally be amenable to lateral arte- 
riorrhaphy, end-to-end anastomosis, or the 
insertion of a saphenous vein or prosthetic 
graft. In general, one should not worry about 
ligating the common hepatic artery proximal 



TABLE 16-1 

SURVIVAL WITH INJURIES TO THE SUPRARENAL ABDOMINAL AORTA 



Reference 

Arch Surg 109:706, 1974 

Am J Surg 128:823, 1974 

Ann Surg 42:1, 1976 

J Trauma 22:481, 1982 

J Trauma 22:672,1982 

Surg Gynecol Obstet 160:313, 1985 

Am J Surg 154:613, 1987 

Am Surg 58:622, 1992 

J Trauma 50:1020, 2001 

Diaphragmatic 

Visceral 

Suprarenal 

Pararenal 
Overall 



No. Patients 


No. Survivors 


% Survival 


17 


5 


29.4 


28 


10 


35.7 


5 


4 


80.0 


3 


3 


100.0 


9 


4 


44.4 


15 


7 


46.6 


74 


21 


28.4 


4 





0.0 


9 


1 


11.1 


9 


1 


11.1 


13 





0.0 


5 


1 


20.0 


191 


57 


29.8 



chl6.qxd 4/16/04 3:40 PM Page 305 



16 • INJURY TO ABDOMINAL AORTA AND VISCERAL ARTERIES 



305 



to the origin of the gastroduodenal artery, 
because the extensive collateral flow from the 
inferior pancreaticoduodenal artery in the 
midgut will maintain the viability of the liver. 
If the entire celiac trunk is injured, it is best 
to ligate all three vessels and make no attempt 
at repair. Ligation of the celiac trunk has never 
caused any short-term morbidity or mortality 
in properly resuscitated patients. 



SUPERIOR MESENTERIC ARTERY 

Injuries to the superior mesenteric artery may 
occur at several levels. In 1972 Fullen and col- 
leagues described an anatomic classification 
of injuries to the superior mesenteric artery 
that has been used only infrequently by sub- 
sequent authors in the trauma literature. If 
the injury to the superior mesenteric artery 
is beneath the pancreas (Fullen zone I), the 
pancreas may on rare occasions have to be 
transected between Glassman or Dennis 
intestinal clamps to control the bleeding point. 
Because the superior mesenteric artery has 
few branches at this level, proximal and distal 
vascular control is relatively easy to obtain once 
the overlying pancreas has been divided. 
Another option is to perform medial rotation 
of the left-sided intra-abdominal viscera, as pre- 
viously described, and apply a clamp from the 
left side of the aorta directly to the proximal 
superior mesenteric artery at its origin. In this 
instance the left kidney may be left in the 
retroperitoneum as the medial rotation is 
performed. 

Injuries to the superior mesenteric artery 
also occur beyond the pancreas at the base of 
the transverse mesocolon (Fullen zone II, 
between the pancreaticoduodenal and middle 
colic branches of the artery) . Although there 
is certainly more space in which to work in 
this area, the proximity of the pancreas and 
the potential for pancreatic leaks near the 
arterial repair make injuries in this location 
almost as difficult to handle as the more prox- 
imal injuries. If the superior mesenteric artery 
has to be ligated at its origin from the aorta 
or beyond the pancreas (Fullen zone I or II) , 
collateral flow from both the foregut and the 
hindgut should theoretically maintain the via- 
bility of the midgut in the distribution of this 



vessel. Exsanguinating hemorrhage from 
injuries in this area, however, often leads to 
profound shock with intense vasoconstriction 
of the distal superior mesenteric artery. For 
this reason, collateral flow is often inadequate 
to maintain viability of the distal midgut, 
especially the cecum and ascending colon. In 
the hemodynamically unstable patient with 
hypothermia, acidosis and a coagulopathy, the 
insertion of a temporary intraluminal shunt 
into the debrided ends of the superior mesen- 
teric artery is a better choice than ligation and 
fits the definition of damage control. If replace- 
ment of the proximal superior mesenteric 
artery is to be performed at a first operation 
or at a reoperation after damage control, it is 
safest to place a saphenous vein or prosthetic 
graft on the distal infrarenal aorta, away from 
the injury to the pancreas and other upper 
abdominal organs (Fig. 16-6) . A graft in this 
location should be tailored so that it will pass 
through the posterior aspect of the mesen- 
tery of the small bowel and then be sutured 
to the mid or distal superior mesenteric 
artery in an end-to-side or end-to-end fashion 
without significant tension. It is mandatory to 
cover the proximal suture line on the 
infrarenal aorta with retroperitoneal fat or a 
viable omental pedicle to avoid an aortoen- 
teric fistula at a later time. Injuries to the more 
distal superior mesenteric artery beyond the 
transverse mesocolon (Fullen zone III, beyond 
the middle colic branch) should be repaired 
if at all possible to avoid ischemia of the distal 
midgut. Injuries to the segmental branches 
(Fullen zone IV) are usually ligated and fol- 
lowed by resection of portions of the midgut 
as needed. 

The survival rate among patients with pen- 
etrating injuries to the superior mesenteric 
artery during the 1970s and 1980swas approx- 
imately 58%, and this is still true (Table 16- 
1). This decreases to 20% to 25% when any 
form of repair more complex than lateral arte- 
riorrhaphy is necessary. In three more recent 
series in which 84 patients with injuries to the 
superior mesenteric artery were described 
(Asensio and colleagues, 2000; Davis and col- 
leagues, 2001; Tyburski and colleagues, 2001 ) , 
survival was approximately 49% (Table 16-2). 

A large multi-institutional review of such 
injuries was reported in 2001 by Asensio and 



chl6.qxd 4/16/04 3:40 PM Page 306 



306 



IV • SPECIFIC VASCULAR INJURIES 



■ FIGURE 16-6 

It may be dangerous to place 
the proximal suture line of a 
graft in Fullen zone I or II of the 
superior mesenteric artery near 
an associated pancreatic 
injury. The proximal suture line 
should be on the lower aorta, 
away from the upper 
abdominal injuries, and 
covered with retroperitoneal 
tissue. (From Baylor College of 
Medicine, 1985.) ■ 




colleagues. There were 250 patients with 
injuries to the superior mesenteric artery 
(52% penetrating; 48% blunt) treated in 34 
trauma centers over a 10-year period. Data 
were available on operative management in 
244 patients including 175 (72%) with liga- 
tion's (22%) with suture repair, and 16 (6%) 



with insertion of an autogenous (no. 10) or 
PTFE (no. 6) graft. Overall survival was 61% 
and ranged from 23.5% for patients with 
Fullen zone I injuries to 76.9% for those with 
Fullen zone IV injuries. Finally, logistic regres- 
sion analysis was used to identify independent 
risk factors for mortality. These risk factors 



TABLE 16-2 

SURVIVAL WITH INJURIES TO THE SUPERIOR MESENTERIC ARTERY 



Reference 

J Trauma 12:656, 1972 
Surgery 84:835, 1978 
Ann Surg 193:30, 1981 
J Trauma 22:672,1982 
J Trauma 23:372, 1983 
J Trauma 26:313, 1986 
Am J Surg 180:528, 2000 
Am Surg 67:565, 2001* 
J Trauma 50:1020, 2001 
J Am Coll Surg 193:354, 2001 

(multi-institutional) 

Overall 



No. Patients 


No. Survivors 


% Survival 


8 


5 


62.5 


45 


27 


60.6 


15 


10 


66.7 


6 


4 


66.7 


20 


14 


70.0 


22 


7 


31.8 


28 


13 


46.4 


15 


8 


53.3 


41 


20 


48.8 


250 


153 


61.2 



450 



261 



58.0 



'Excludes patients with exsanguination before repair or ligation. 



chl6.qxd 4/16/04 3:40 PM Page 307 



16 • INJURY TO ABDOMINAL AORTA AND VISCERAL ARTERIES 



307 




©Baylor College of Medicine 1980 



■ FIGURE 16-7 

Vessel loops or umbilical tapes 
are placed around the proximal 
renal vessels before perirenal 
hematomas are entered. (From 
Baylor College of Medicine, 
1980.) ■ 



included the following: transfusion of more 
than 10 units of packed red blood cells; intra- 
operative acidosis; dysrhythmias; injury in 
Fullen zone I or II; or the development of 
multisystem organ failure. 



RENAL ARTERY 

Injuries to the proximal renal arteries may also 
present with a supramesocolic hematoma or 
with hemorrhage in this area. With an injury 
to the proximal renal artery, supraceliac 
control of the abdominal aorta by either of 
the methods previously described will be nec- 
essary. A tamponaded injury closer to the renal 
hilum allows for proximal control of the renal 
artery in the midline retroperitoneum (Fig. 
16-7) . The transverse mesocolon is retracted 
superiorly, and the small bowel is eviscerated 
to the right. The ligament of Treitz is then 
divided as the inferior mesenteric vein is 
retracted to the left. With extensive mobi- 
lization of the duodenojejunal junction, the 
left renal vein crossing over the juxtarenal 
abdominal aorta is exposed and mobilized, 
as needed, by ligation and division of the left 
adrenal, gonadal, and renal lumbar veins. 
Such extensive mobilization will allow this vein 



to be retracted 6 to 7cm in a superior direc- 
tion. The origin of the left renal artery at the 
4-o'clock position on the juxtarenal abdomi- 
nal aorta is readily identified by dissection of 
the surrounding retroperitoneal tissue of 
modest density. A vessel loop is then passed 
around the proximal left renal artery. To 
expose the origin of the right renal artery at 
the 7-o'clock position on the juxtarenal 
abdominal aorta, the surgeon may need to 
retract the adjacent infrarenal inferior vena 
cava to the rightwith a vein retractor. The right 
renal vein cannot be looped until an exten- 
sive Kocher maneuver is performed to expose 
the juxtarenal inferior vena cava. Options for 
repair of either the proximal or the distal renal 
artery are described later in this chapter. 



Inframesocolic Area of Zone 1 

INFRARENAL ABDOMINAL AORTA 

The second major area of hematoma or 
hemorrhage in the midline retroperitoneum 
is the inframesocolic area. Patients with 
injuries to the infrarenal (or suprarenal) abdom- 
inal aorta and signs of life upon arrival in the 
operating room always have a massive midline 



chl6.qxd 4/16/04 3:40 PM Page 308 



308 



IV • SPECIFIC VASCULAR INJURIES 



hematoma in the retroperitoneum. The size 
of this hematoma is often intimidating to the 
inexperienced trauma surgeon, but one 
simple rule should be kept in mind: The hole 
in the aorta is under the highest point of the 
hematoma. Therefore, an injury just below 
the base of the mesocolon is likely to involve 
thejuxtarenal abdominal aorta and demands 
proximal aortic control in the upper abdomen 
by using the previously described techniques. 
A midline hematoma over the lower lumbar 
area is likely to be over an injury to the 
infrarenal abdominal aorta, and exposure and 
control will be easier. With either a true 
inframesocolic hematoma or an area of hem- 
orrhage, proximal aortic control is obtained 
as described previously for exposure of the 
proximal renal arteries. The transverse meso- 
colon is elevated superiorly, the small bowel 
is eviscerated to the right, and the ligament 
of Treitz is divided to allow for application of 
an aortic cross clamp in the infrarenal posi- 
tion (Fig. 16-8). Exposure to allow for appli- 
cation of the distal vascular clamp is obtained 




■ FIGURE 16-8 

Gunshot wound of infrarenal abdominal aorta 
viewed through standard inframesocolic 
exposure (head of patient is toward the 
proximal clamp). (From Feliciano DV, Burch JM, 
Graham JM: Abdominal vascular injury. In 
Mattox KL, Moore EE, Feliciano DV [eds]: 
Trauma, 1st ed. Stamford, Conn, Appleton & 
Lange, 1988, pp 519-536.) ■ 



by dividing the midline retroperitoneum 
down to the aortic bifurcation, carefully avoid- 
ing the left-sided origin of the inferior mesen- 
teric artery; however, this vessel may be 
sacrificed whenever necessary for exposure 
in young trauma patients. 

As with injuries to the suprarenal aorta, 
injuries in the infrarenal abdominal aorta are 
repaired in a transverse fashion with 3-0 or 4- 
polypropylene suture or by patch aortoplasty, 
end-to-end anastomosis, or insertion of a 
woven Dacron graft, an albumin-coated 
Dacron graft, or a PTFE graft, none of which 
requires preclotting. Because of the small size 
of the aorta in young trauma patients, it is 
unusual to be able to place a tube graft larger 
than 12, 14, or 16 mm in diameter if one is 
required, as previously noted. The principles 
of completing the suture lines and flushing 
are exactly the same as those for aortic repairs 
in the suprarenal area. Because the retroperi- 
toneal tissue is often thin in young patients, 
it may be worthwhile to cover an extensive 
aortic repair or the suture lines of a prosthe- 
sis with mobilized omentum before closure 
of the retroperitoneum. One option is to 
divide the gastrocolic omentum, flip the 
omentum superiorly into the lesser sac, and 
make a window in the left side of the trans- 
verse mesocolon. The mobilized pedicle is 
passed through the window and placed over 
the aortic repair or graft. The other option is 
to mobilize the gastrocolic omentum away 
from the right side of the transverse colon. 
This mobilized pedicle is then placed lateral 
to the ligament of Treitz and over the aortic 
repair or graft. With either technique, 2-0 or 
3-0 absorbable sutures are used to attach the 
omental pedicle to the opened retroperitoneal 
edges around the area of repair in the 
infrarenal abdominal aorta. 

The survival rate among patients with pen- 
etrating injuries to the infrarenal abdominal 
aorta during the 1970s, 1980s, and early 1990s 
was approximately 46% (Table 16-3). In a 
more recent series with 35 patients, the sur- 
vival rate was 34.3% (Tyburski and colleagues, 
2001). In three other recent series (Coimbra 
and colleagues, 1996; Asensio and colleagues, 
2000; Davis and colleagues, 2001) in which 
140 patients with injuries to the abdominal 
"aorta" (not specified as to whether the loca- 



chl6.qxd 4/16/04 3:40 PM Page 309 



16 • INJURY TO ABDOMINAL AORTA AND VISCERAL ARTERIES 



309 



TABLE 16-3 

SURVIVAL WITH INJURIES TO THE INFRARENAL ABDOMINAL AORTA 



Reference 



Arch Surg 109:706, 1974 

Am Surg 41:755, 1975* 
J Trauma 22:672, 1982 
J Trauma 22:481, 1982 
Surg Gynecol Obstet 160:313, 1985 
Am Surg 58:622, 1992 
J Trauma 50:1020, 2001 
Overall 



No. Patients 


No. Survivors 


% Survival 


15 


7 


46.7 


40 (aortoiliac) 


17 


42.5 


9 


4 


44.4 


12 


7 


58.3 


10 


4 


40.0 


7 


4 


57.1 


35 


12 


34.3 


88 


38 


43.2 



*Not included in overall figures. 



tion was suprarenal or infrarenal) were 
treated, the survival was 42.8% (12/28), 
20.6% (13/63), and 39.1% (25/64, exclud- 
ing 13 patients who exsanguinated before 
repair) (Table 16-4). The overall survival of 
approximately 32% for all aortic injuries in 
these recent reviews, a decrease of approxi- 
mately 6% of all the aortic injuries before 1993 
(Tables 16-1 and 16-3) are included, is quite 
interesting. The recent survival figures may 
reflect shorter scene times in urban environ- 
ments, which would bring more exsan- 
guinated patients to the trauma center, or 
this change may be a manifestation of 
more patients with multiple penetrating 
wounds and/or injuries. 

There is one interesting report by Soldano 
and colleagues (1988) of the long-term follow- 
up of 11 survivors of penetrating wounds to 
the abdominal aorta (9 infrarenal injuries and 
5 suprarenal injuries in the 11 patients) from 



the Vietnam War. Ankle-to-brachial pressure 
ratios were decreased in five (one only with 
exercise) , and all had calcification of the area 
of repair on abdominal computed tomogra- 
phy(CT). 



Zone 2 or Upper 
Lateral Retroperitoneum 

RENAL ARTERY 

If a hematoma or hemorrhage is present in 
the lateral perirenal area, injury to either the 
distal renal artery, the renal vein, or both or the 
kidney should be suspected. In hemodynami- 
cally stable patients who have suffered blunt 
abdominal trauma and have normal preop- 
erative IVP, renal arteriogram, or CT of the 
kidneys, there is no justification for explor- 
ing the kidney through its perirenal 



TABLE 16-4 

RECENT SURVIVAL WITH INJURIES TO THE ABDOMINAL AORTA (NOT 
OTHERWISE SPECIFIED) 



Reference 


No. 


Patients 


No. 


Survivors 


% Survival 


Am J Surg 172:541, 1996 




28 




12 


42.8 


Am J Surg 180:528, 2000 












Isolated injury 




46 




10 


21.7 


With other arterial injury 




17 




3 


17.6 


Am Surg 67:565, 2001* 




64 




25 


39.1 


Overall 




155 




50 


32.3 



'Excludes patients with exsanguination before repair. 



chl6.qxd 4/16/04 3:40 PM Page 310 



310 



IV • SPECIFIC VASCULAR INJURIES 



hematoma at a laparotomy performed for 
other injuries. As previously noted, the perire- 
nal hematoma should be opened if it is pul- 
satile, expanding rapidly, or has already 
ruptured partially. 

In highly selected and hemodynamically 
stable patients withpenetratingwounds to the 
flank, CT has been used to document an iso- 
lated minor renal injury and operation has 
been avoided. All other patients found to have 
a perirenal hematoma at the time of explo- 
ration for a penetrating abdominal wound 
should have unroofing of the hematoma and 
exploration of the underlying kidney ("Huey 
Long rule"). 

If the hematoma is not rapidly expanding 
and there is no free intra-abdominal bleed- 
ing, most surgeons will loop the ipsilateral 
renal artery with avascular tape in the midline 
at the base of the mesocolon as previously 
described. It should be noted that there is little 
consensus on the value of preliminary arter- 
ial control at the midline in stable patients. 

If there is active bleeding from the kidney 
through Gerota's fascia or from the retroperi- 
toneum overlying the renal vessels, no central 
renovascular control is necessary. The surgeon 
should simply open the retroperitoneum 
lateral to the injured kidney, divide Gerota's 
fascia, and manually elevate the kidney directly 
into the wound. A large vascular clamp can 
be applied proximal to the hilum orjust lateral 
to the inferior vena cava on the right to control 
any further bleeding. 

Renovascular injuries from penetrating 
trauma are difficult to manage, especially 
when the renal artery is involved. It is an ex- 
traordinarily small vessel that is deeply em- 
bedded in the retroperitoneum. Occasionally, 
small perforations of the artery from pene- 
trating wounds can be repaired by lateral arte- 
riorrhaphy or resection with an end-to-end 
anastomosis. Interposition grafting using 
either a saphenous vein or a PTFE graft or 
use of borrowed arteries, such as the splenic 
artery to replace the left renal artery and the 
hepatic artery to replace the right renal 
artery, is indicated only when the renal artery 
to the patient's only kidney is injured. In other 
patients with multiple intra-abdominal injuries 
or a long preoperative period of ischemia, 
nephrectomy is a better choice, as long as 



intraoperative palpation has confirmed a 
normal contralateral kidney. The survival rate 
for patients with injuries to the renal arteries 
from penetrating trauma in two older series 
(1980; 1990) was approximately 87%, with 
renal salvage in only 30% to 40%. 

Controversy continues to surround the 
role of renal revascularization after the delayed 
diagnosis of thrombosis of the renal artery 
from blunt trauma. Intimal tears in the renal 
arteries may result from deceleration in motor 
vehicle crashes, automobile-pedestrian 
crashes, and falls from heights. These usually 
lead to secondary thrombosis of the vessel and 
complaints of upper abdominal and flank 
pain. One literature review in 1980 noted that 
only 30% of patients with intimal tears in the 
renal arteries had gross hematuria, 43% had 
microscopic hematuria, and 27% had no 
blood in the urine. A more recent report in 
1998 documented that seven of eight patients 
in whom a urinalysis was performed had hema- 
turia. Therefore, the diagnosis may be missed, 
because an IVP or CT may not be performed 
in stable patients with normal abdominal 
examinations and no hematuria or microhe- 
maturia, only, after blunt trauma. 

Lack of enhancement of a kidney with intra- 
venous contrast on an abdominal CT is 
pathognomonic of blunt thrombosis of the 
ipsilateral renal artery. As the intimal tear is 
always 2 to 4 cm from the abdominal aorta, 
the value of a follow-up renal arteriogram 
to confirm the diagnosis is questionable 
(Fig. 16-9). 

The operative technique when renal revas- 
cularization is attempted is straightforward. 
Resection of the area of the intimal tear and 
an end-to-end anastomosis, insertion of an 
aortorenal artery bypass graft, or ex vivo per- 
fusion of the ischemic kidney followed by 
autotransplantation into the pelvis can all be 
performed by an experienced vascular or 
transplantation surgery team. The value of 
external cooling of the ischemic kidney or 
infusing a cold renal perfusion solution 
before a revascularization procedure after 
the artery is opened is unclear. The same 
can be said for the value of decapsulation of 
the previously ischemic kidney to prevent a 
post-revascularization "kidney compartment 
syndrome." 



chl6.qxd 4/16/04 3:40 PM Page 311 



16 • INJURY TO ABDOMINAL AORTA AND VISCERAL ARTERIES 



311 




■ FIGURE 1 6-9 

Blunt occlusion of the right 
renal artery on an abdominal 
aortogram. ■ 



The controversy regarding revascularization 
is related to the poor results that have been 
reported. The time from injury to revascu- 
larization appears to be critical, as would be 
expected when dealing with one kidney that 
receives 12.5% of the cardiac output each 
minute. In one review in 1978, some renal 
function was restored in 80% of patients 
undergoing renal revascularization within 12 
hours of occlusion. This figure decreased to 
57% if revascularization did not occur for 18 
hours. A more recent report of 12 patients 
with blunt thrombosis of the renal artery (one 
bilateral) by Haas and colleagues in 1998 is 
even more discouraging. In the group of five 
patients who underwent attempted revascu- 
larization of the renal artery at a median warm 
ischemia time of 5 hours (4.5 to 36 hours), 
four were felt to be "technically successful." 
Immediate nephrectomy was performed in 
another, with an unsuccessful attempt at 
revascularization. The outcomes for the four 
patients with successful revascularization were 
as follows: nephrectomy at 1 day (no function 
on postoperative renal scan) in one; death on 
hemodialysis at 2 months in another; nephrec- 
tomy at 6 months because of delayed hyper- 
tension in a third; and minimal function (9% 
differential) at 1 month on a renal scan in the 
fourth. In the same series, seven patients with 
blunt thrombosis of the renal artery did not 
undergo revascularization. A delayed nephrec- 
tomy was required at a mean time of 5 months 
in three patients (43% ) who developed hyper- 
tension, and four were normotensive at a mean 



time of 11 months from injury. Based on the 
historical and recent data, it is difficult to rec- 
ommend revascularization of one renal artery 
in a patient with a functioning contralateral 
kidney after sustaining blunt trauma. This is 
especially true if the patient has other serious 
injuries and time to revascularization would 
exceed 6 hours from injury. This conclusion, 
of course, would not be acceptable to the 
authors of case reports or reviews document- 
ing successful late renal revascularization 
(one or both kidneys) after bilateral throm- 
bosis of the renal arteries. Such successful 
repairs have been performed at 12, 15, 18, 
and 19 hours, as reported by Greenholz and 
colleagues (1986). Patients not undergoing 
revascularization of one thrombosed renal 
artery need to be monitored for 6 to 12 months 
after injury to allow for early detection of 
delayed hypertension. 

There are isolated case reports in 
which patients with failure of bilateral renal 
revascularization and no revascularization 
because of biopsy-proven renal necrosis had 
return of renal function starting at 4 to 8 
weeks after injury. This phenomenon is pre- 
sumably related to maintenance of some 
renal viability via collateral flow to renal 
capsular vessels and to recanalization of 
the thromboses in the renal arteries. If 
patients with bilateral thrombosis with or 
without attempts at renal revascularization 
remain dependent on hemodialysis, they 
should be put on a waiting list for renal 
transplantation. 



chl6.qxd 4/16/04 3:40 PM Page 312 



312 



IV • SPECIFIC VASCULAR INJURIES 



■ FIGURE 16-10 

Blunt intimal tear in the left 
renal artery demonstrated on a 
"pullout" abdominal aortogram. 
(From Feliciano DV, Burch JM, 
Graham JM: Vascular injuries of 
the chest and abdomen. In 
Rutherford RB, et al [eds]: 
Vascular Surgery, 3rd ed. 
Philadelphia, WB Saunders, 
1989, pp 588-603.) ■ 




There are patients with blunt trauma who 
undergo "pullout" abdominal aortograms 
after evaluation of the thoracic aorta or pre- 
liminary abdominal aortography before pelvic 
arteriography who are found to have intimal 
tears in the renal artery without thrombosis 
(Fig. 16-10). When there is no extravasation 
of contrast at the site of injury, observation 
and follow-up arteriography within the first 
week after injury are appropriate. The role of 
anticoagulation is problematic because so 
many of these patients have associated injuries. 
In the absence of serious associated injuries, 
anticoagulation would seem appropriate, 
recognizing the absence of meaningful data. 
An intimal or wall defect that was the pre- 
sumed source of embolic infarctions in the 
ipsilateral kidney has been treated successfully 
by insertion of an endovascular stent in one 
recent report by Villas and colleagues (1999). 



Complications 

The complications of repairs of the abdom- 
inal aorta or visceral arteries include distal 
embolization, thrombosis, dehiscence of a 
suture line, and infection. Occlusion is not 



uncommon when small vasoconstricted 
vessels, such as the superior mesenteric artery 
or renal artery, undergo lateral arteriorrha- 
phy. In such patients, it may be valuable to 
perform a second-look operation within 12 
to 24 hours after the patient's blood pressure, 
temperature, and coagulation abnormalities 
have returned to normal. When this is done, 
correction of a vascular thrombosis may be 
successful. 

As previously noted, dehiscence of vascu- 
lar suture lines in the superior mesenteric 
artery near a pancreatic injury may occur if 
a small pancreatic leak occurs in the post- 
operative period. For this reason the proxi- 
mal anastomosis of such a graft should be on 
the infrarenal aorta far away from the pan- 
creas as described. 

In addition, the postoperative develop- 
ment of vascular enteric fistulas occurs most 
commonly in patients who have anterior 
aortic repairs, aortic grafts, or grafts to the 
superior mesenteric artery from the aorta. 
Again, this problem can be avoided by proper 
coverage of suture lines on the aorta with 
retroperitoneal tissue or a viable omental 
pedicle and on the recipient vessel with 
mesentery. 



chl6.qxd 4/16/04 3:40 PM Page 313 



16 • INJURY TO ABDOMINAL AORTA AND VISCERAL ARTERIES 



313 



REFERENCES 

Asensio JA, Britt LD, Borzotta A, et al: Multiinsti- 
tutional experience with the management of 
superior mesenteric artery injuries. J Am Coll 
Surg 2001;193:354-366. 

Asensio JA, Chahwan S, Hanpeter D, et al: 
Operative management and outcome of 302 
abdominal vascular injuries. Am J Surg 
2000;180:528-534. 

Asensio JA, Forno W, Roldan G, et al: Abdominal 
vascular injuries: Injuries to the aorta. Surg Clin 
North Am 2001 Dec;81(6):1395-1416, xiii-xiv. 
Review. 

Coimbra R, Hoyt D, Winchell R, et al: The ongoing 
challenge of retroperitoneal vascular injuries. Am 
J Surg 1996;172:541-545. 

Davis TP, Feliciano DV, Rozycki GS, et al: Results 
with abdominal vascular trauma in the modern 
era. Am Surg 2001;67:565-571. 

Feliciano DV: Management of traumatic retroperi- 
toneal hematoma. Ann Surg 1990;211:109-123. 



Feliciano DV, Burch JM, Graham JM: Abdominal 
vascular injury. In Mattox KL, Feliciano DV, 
Moore EE (eds) : Trauma, 4th ed. New York, 
McGraw-Hill, 2000, pp 783305. 

Fry WR, Fry RE, Fry WJ: Operative exposure of the 
abdominal arteries for trauma. Arch Surg 
1991;126:289-291. 

Haas CA, Dinchman KH, Nasrallah PF, SpirnakJP: 
Traumatic renal artery occlusion: A 15-year 
review. J Trauma 1998;45:557-561. 

Meghoo CA, Gonzalez EA, Tyroch AH, Wohltmann 
CD: Complete occlusion after blunt injury to the 
abdominal aorta. J Trauma 2003 Oct;55(4) :795- 
799. 

Roth SM, Wheeler JR, Gregory RT, etal: Bluntinjury 
of the abdominal aorta: A review. J Trauma 
1997;42:748-755. 

TyburskiJG, Wilson RF, Dente C, et al: Factors affect- 
ing mortality rates in patients with abdominal 
vascular injuries. J Trauma 2001;50:1020-1026. 



chl7.qxd 4/16/04 3:38PM Page 315 




Injuries of the Inferior 
Vena Cava and Portal 
Venous System 



ROBERT F. BUCKMAN, JR. 
ABHIJIT S. PATHAK 
KEVIN M. BRADLEY 



INJURIES OF THE INFERIOR VENA CAVA 

Surgical Anatomy 

Patterns of Injury 

Initial Assessment and Management 

Exposure and Control 

Control of hemorrhage: intrahepatic inferior vena cava 

Control of hemorrhage: retrohepatic inferior vena cava 
Inferior Vena Cava Repair 
Postoperative Management 
Summary 

PORTAL VEIN INJURIES 
Surgical Anatomy 

Portal vein 

Superior mesenteric and splenic vein 

Collaterals in portal obstruction 
Patterns of Injury 

Associated injuries 
Initial Assessment and Management 



315 



chl7.qxd 4/16/04 3:38PM Page 316 



316 



IV • SPECIFIC VASCULAR INJURIES 



Exposure and Initial Vascular Control 

Stable hematoma 

Suprapancreatic exposure 

Retropancreatic exposure 

Pancreatic division 

Control of multiple vascular injuries 
Definitive Repair 

Suprapancreatic wounds 

Retropancreatic wounds 

Portal vein ligation 
Postoperative Management 
Summary 



Venous injury in the upper abdomen 
most often involve the inferior vena 
cava (IVC) and the portal venous 
system. Often such injuries occur simultane- 
ously. The literature on these injuries is 
usually presented separately; therefore, these 
injuries are presented in two sections of this 
chapter. 



INJURIES OF THE INFERIOR 
VENA CAVA 

The IVC, though deeply protected against the 
accidents of nature, is by no means immune 
to wounding. It has been estimated that 10% 
to 15% of cases of abdominal penetration 
result in an injury to a major vein and that 1 
of every 50 gunshot wounds to the abdomen 
strikes the IVC (Starzl and colleagues, 1962; 
Wiencek and Wilson, 1986). Although pene- 
trating mechanisms cause most caval injuries 
and can involve any portion of the IVC, the 
retrohepatic and intrapericardial sections are 
the only portions of the vessel that are injured 
by blunt trauma. 

Whether caused by blunt or penetrating 
mechanisms, caval wounds are highly lethal. 
As many as 50% of patients with such injuries 



die before reaching the hospital and the 
mortality among patients who arrive at a 
trauma center with signs of life has ranged 
between 20% and 57% (Duke, Jones, and 
Shires, 1965; Quast and colleagues, 1965; 
Weichert and Hewitt, 1970; Burns and 
Sherman, 1972; Graham and colleagues, 
1978; Kudsk, Sheldon, and Lim, 1982; 
Feliciano and colleagues, 1984; Wiencek and 
Wilson, 1986; Klein, Baumgartner, and 
Bongard, 1994; Burch and colleagues, 1998; 
Asensio and colleagues, 2001). The three 
factors that are most important in the prog- 
nosis for survival are the hemodynamic con- 
dition of the patient on arrival, the occurrence 
of spontaneous tamponade of the caval injury, 
and to a lesser degree, the location of the caval 
laceration (Weichert and Hewitt, 1970; 
Graham and colleagues, 1978; Kashuk and 
colleagues, 1982; Kudsk, Sheldon, and Lim, 
1982; Wiencek and Wilson, 1986; Klein, 
Baumgartner, and Bongard, 1994; Burch and 
colleagues, 1998). Patients who arrive in 
shock and fail to respond to initial resuscita- 
tive measures, those who are still actively bleed- 
ing at the time of laparotomy, and those with 
wounds of the retrohepatic vena cava have a 
low probability of survival. Death is most com- 
monly due to intraoperative exsanguination 
(Nakamura and Tzuzuki, 1981; Wiencek and 
Wilson, 1986; Burch and colleagues, 1998). 



chl7.qxd 4/16/04 3:38PM Page 317 



17 • INJURIES OF THE INFERIOR VENA CAVA AND PORTAL VENOUS SYSTEM 



317 



Surgical Anatomy 

The IVC originates by the confluence of the 
common iliac veins just anterior to the body 
of the fifth lumbar vertebra and posterior to 
the right common iliac artery. As it ascends 
along the right side of the lumbar vertebral 
bodies, the cava receives numerous tributa- 
ries including four or five pairs of lumbar 
(sometimes called "segmental") veins, the 
right gonadal vein, the renal veins, the right 
adrenal vein, and finally the hepatic and 
phrenic veins. It then traverses the mid- 
diaphragm to reach the right atrium. The IVC 
is a relatively delicate and thin-walled vessel, 
1.5 inches in diameter, valveless throughout 
its length, with a high flow at an intraluminal 
pressure of about 5 cm H 2 0. 

The intra-abdominal vena cava may be 
divided into five sections, each of which has 
anatomic peculiarities that affect the exposure 
and control of injuries that section (Weichert 
and Hewitt, 1970). The lowest section is the 
bifurcation. Above this are the infrarenal, the 
perirenal, the suprarenal/subhepatic, and 
the retrohepatic sections. The management 
of injuries in each of these segments is dis- 
cussed separately. 

An important anatomic feature of both the 
bifurcation and the infrarenal sections of the 
IVC is an abundant collateral circulation, of 
which the lumbar veins constitute the prin- 
cipal elements. These paired veins are con- 
nected with one another, with the common 
iliac, hypogastric, iliolumbar, and renal veins 
and with the azygos and hemiazygos system 
through bilateral ascending lumbar veins. This 
extensive network is capable of bypassing 
any obstruction of the bifurcation or of the 
infrarenal segment of the vena cava, but the 
very richness of the collateral circulation also 
confounds efforts to achieve proximal and 
distal control of injuries in these zones 
(Fig. 17-1). 

The perirenal area extends about 1 inch 
above and below the renal veins and lies pos- 
terior to the pancreas and duodenum. The 
high flow from the renal veins requires that 
these vessels and the IVC itself be occluded 
to control bleeding from wounds in this 
section of the cava (Wiencek and Wilson, 1986; 
Burch and colleagues, 1998). 




■ FIGURE 1 7-1 

The abundant collaterals of the infrarenal vena 
cava. Lumbar veins communicate with 
ascending veins that drain into the azygos and 
hemiazygos systems. (From Buckman RF Jr, 
Pathak AS, Badellino MM, Bradley KM: Injuries 
of the inferior vena cava. Surg Clin North Am 
2001;81[6]:1433.) ■ 



Between the perirenal segment and the 
beginning of the retrohepatic segment is a 
short, suprarenal-subhepatic region of the 
IVC. Injuries in this segment are difficult to 
control because of the renal vessels below 
and the proximity of the liver above, with the 
passage of the cava underneath the liver into 
the retrohepatic zone. The portal vein lies 
immediately anterior to this short segment of 
the IVC. 

The segment of the IVC that has the most 
unique anatomic features is the retrohepatic 
section, lying above the right adrenal vein and 
below the phrenic veins. This portion of the 
cava, 7 to 10 cm in length, lies in a groove or 
tunnel on the posterior aspect of the liver 
within its "bare" area (Nakamura and Tzuzuki, 
1981). This area, completely circumscribed 
by the hepatic suspensory ligaments with the 



chl7.qxd 4/16/04 3:38PM Page 318 



318 



IV • SPECIFIC VASCULAR INJURIES 



diaphragm behind and the liver in front, has 
the ability to confine or contain bleeding asso- 
ciated with retrohepatic caval injuries, pro- 
vided that the diaphragm, the posterocentral 
liver, or hepatic ligaments are not themselves 
severely disrupted by the traumatic event 
or by surgical intervention (Buckman, 
Miraliakbari, and Badellino, 2000) . The retro- 
hepatic vena cava is joined by two or three 
major hepatic veins shortly before it traverses 
the diaphragm and below this by seven or more 
accessory hepaticveins of varying sizes. These 
numerous tributaries bind the cava to the liver, 
making its circumferential mobilization dan- 
gerous. The exposure of caval injuries in the 
retrohepatic zone is exceptionally difficult and 
is usually unnecessary if spontaneous con- 
tainment of hemorrhage has been achieved 
by the suspensory ligaments, the liver, and the 
diaphragm. 



Patterns of Injury 

With the exception of the retrohepatic and 
intrapericardial vena cava, which may be 
injured by blunt or penetrating trauma, vir- 
tually all other IVC injuries are caused by pen- 
etrating mechanisms. Gunshot wounds are 
much more likely than stab wounds to lacer- 
ate the IVC and generate far more destruc- 
tive wounding patterns. Although stab wounds 
cause linear lacerations of the vena cava, which 
often spontaneously tamponade, gunshot 
wounds, especially the high-energy wounds 
of the current era, produce large tangential 
avulsions involving varying amounts of the 
circumference or actual transection of the 
vessel. 

Almost every patient with a penetrating 
wound of the vena cava has injuries to other 
viscera, other major vessels, or both (Bricker 
and Wukasch, 1970; Bricker and colleagues, 
1971; Mattox and colleagues, 1974, 1975). 
Injuries of the liver, duodenum, pancreas, 
bowel, and colon are common. Approxi- 
mately 10% of the patients wounded in the 
IVC have a second major vascular injury, most 
commonly involving the aorta or portal vein 
(Mattox and colleagues, 1975; Linker and 
colleagues, 1982). In rare instances, the 



combined penetration of the vena cava with 
the aorta leads to the development of an aor- 
tocaval fistula. Acute traumatic fistulas may 
also occur between the cava and the renal 
arteries or the cava and the duodenum. 

Blunt injuries to the IVC are generally 
caused by shearing forces in violent deceler- 
ation accidents and may take the form of avul- 
sion of the atriocaval junction or tearing of 
hepaticveins from the retrohepatic vena cava. 
Intraparenchymal lacerations of the hepatic 
veins or the anterior surface of the retrohep- 
atic cava may occur in severe blunt fractures 
of the posterocentral liver caused by crush- 
ing injuries. 

For a caval or other venous injury to bleed 
freely, there must exist, in addition to the 
venous wound itself, a major breach of sur- 
rounding tissues normally capable of confin- 
ing or containing low-pressure hemorrhage. 
The capacity for self-tamponade is charac- 
teristic of caval injuries because it is of all 
venous injuries and has important clinical 
implications. Among patients with IVC 
injuries, more than half will spontaneously 
contain the site of injury with cessation of 
bleeding (Ochsner, Crawford, and DeBakey, 
1961; Starzl and colleagues, 1962; Duke, 
Jones, and Shires, 1965; Weichert and Hewitt, 
1970; Burch and colleagues, 1998) . This phe- 
nomenon is more likely to occur with oblique, 
crossing, low-velocity gunshot wounds and 
with stab wounds than with straight, front- 
to-back, high-powered gunshot wounds or 
massive hepatic fractures. A beveled or slit- 
like retroperitoneal track favors containment 
and tamponade. Spontaneous tamponade is 
also likely to occur in wounds of the cava that 
are behind the pancreas, duodenum, or liver, 
provided that the overlying viscera are not 
extensively disrupted. In such instances 
of a tamponaded injury, profuse iatrogenic 
rebleeding occurs at the time of surgical 
exposure. 

Numerous authors have observed that the 
retroperitoneal hematoma associated with a 
caval injury may not be large, and that there 
may be minimal free intraperitoneal blood if 
the tamponade occurs early. Although survival 
is far more likely in patients with spontaneous 
cessation of bleeding (Starzl and colleagues, 



chl7.qxd 4/16/04 3:38PM Page 319 



17 • INJURIES OF THE INFERIOR VENA CAVA AND PORTAL VENOUS SYSTEM 



319 



1962; Duke,Jones, and Shires, 1965; Weichert 
and Hewitt, 1970; Mattox and colleagues, 
1974), not all patients who have tamponaded 
will survive. Up to 40% of them may die of 
exsanguination after the tamponade is surgi- 
cally decompressed unless the hemorrhage 
from the cava and associated vascular injuries 
can be completely and quickly controlled 
(Duke, Jones, and Shires, 1965). 

Initial Assessment 
and Management 

Approximately half of patients with wounds 
of the IVC will present with some degree of 
hypotension, often with profound hemody- 
namic compromise (Ochsner, Crawford, and 
DeBakey, 1961; Wiencek and Wilson, 1986; 
Beal, 1990; Klein, Baumrartner, and Bongard, 
1994). Of these, most will show temporary 
improvement with the institution of ap- 
propriate intravenous fluid resuscitation 
(Feliciano and colleagues, 1984). The failure 
of the hypotensive patient to respond to initial 
volume repletion correlates with the presence 
of continued active bleeding, that is, a failure 
of spontaneous tamponade, and portends a 
poor prognosis. At the other extreme, patients 
with IVC injuries who have achieved early 
spontaneous containment of their bleeding 
are often normotensive on arrival. Rare pre- 
sentations of caval injury include acute caval- 
duodenal fistula with hypotension and copious 
emesis of dark blood or acute aortocaval fistula 
characterized by a wide pulse pressure, ab- 
dominal bruit, and hematuria (Linker and 
colleagues, 1982). 

Patientswhose wound trajectories or clinical 
presentations suggest the possibility of major 
intraabdominal vascular wounding should 
have supradiaphragmatic intravenous access 
and be taken directly the operating room. 
Those with the most extreme degrees of car- 
diovascular collapse, who fail to respond to 
initial appropriate resuscitative measures, 
may require resuscitative thoracotomy, per- 
formed in the emergency department. Most 
patients, however, will show a dramatic hemo- 
dynamic improvement with volume repletion 
and can be transported for operation. 



Exposure and Control 

Injuries of the IVC most often present at 
operation as stable hematomas of the central 
retroperitoneum (Ochsner, Crawford, and 
DeBakey, 1961; Starzl and colleagues, 1962; 
Duke, Jones, and Shires, 1965; Burch and 
colleagues, 1998) . Varying amounts of free 
intraperitoneal blood may be present, 
although active hemorrhage from the cava 
often has ceased. When active bleeding is 
occurring, the initial operative maneuver 
should be the manual tamponade of the bleed- 
ing point with a tightly rolled gauze pack. 
Aortic compression may be indicated in 
severely compromised patients until the hemo- 
dynamic condition improves. 

In most patients, tamponade having been 
achieved either spontaneously or by the 
assistance of the surgeon, some circumspec- 
tion is possible before exploration of the 
hematoma. It is often feasible, on the basis of 
the location of the points of retroperitoneal 
penetration to deduce the path of the wound- 
ing agent relative to major retroperitoneal 
structures. 

Recalling that the vast majority of pa- 
tients who die from IVC injuries succumb to 
intraoperative exsanguination (Ochsner, 
Crawford, and DeBakey, 1961; Duke, Jones, 
and Shires, 1965; Weichert and Hewitt, 1970) 
and that many of these patients have sponta- 
neously tamponaded the wound before explo- 
ration, the first question that the surgeon must 
askis whether the hematoma surrounding the 
suspected caval injury truly requires explo- 
ration. Early writers on the subject of caval 
injury and many experienced surgeons sub- 
sequently have urged restraint in the explo- 
ration of stable, nonpulsating retroperitoneal 
hematomas, especially those behind the liver, 
unless an injury to the pancreas, duodenum, 
colon, kidney, or ureter or an associated 
arterial injury is strongly suspected and 
demands exposure (Ochsner, Crawford, and 
DeBakey, 1961; Starzl and colleagues, 1962; 
Duke, Jones, and Shires, 1965; Graham and 
colleagues, 1978; Burch and colleagues, 1998; 
Buckman, Miraliakbari, and Badellino, 2000) . 

Nonpulsatile hematomas accompanying 
presumed injuries to the retrohepatic (or the 



chl7.qxd 4/16/04 3:38PM Page 320 



320 



IV • SPECIFIC VASCULAR INJURIES 



immediately subhepatic) vena cava that have 
spontaneously tamponaded or that can be 
induced to tamponade by manual compres- 
sion or gauze packing are not often associ- 
ated with other retroperitoneal injuries and 
are probably better left unexplored. Decom- 
pression of such hematomas by radical hepatic 
mobilization is often associated with massive, 
sometimes lethal, hemorrhage that cannot 
any longer be controlled by packing after the 
natural containment structures have been sur- 
gically destroyed. Although rebleeding fol- 
lowing spontaneous or assisted tamponade of 
presumed caval injuries is rare, an estimated 
10% to 40% of patients with originally stable 
hematomas bleed to death following op- 
erative exposure of a caval wound (Ochsner, 
Crawford, and DeBakey, 1961; Duke, Jones, 
and Shires, 1965; Weichert and Hewitt, 1970; 
Burch and colleagues, 1998).Mosthematomas 
below the level of the immediate subhepatic 
segment of vena cava require exploration not 
just to fix a possible caval wound, but because 
of the risk of injuries to other retroperitoneal 
visceral or vascular structures (Duke, Jones, 
and Shires, 1965). 

Having reached whatever conclusions are 
possible regarding the probable location 
and nature of the vascular injury in a central 
hematoma, and having determined that either 
active hemorrhage or the risk of associated 
injuries outweighs the dangers of caval explo- 
ration, preparations to enter the hematoma 
should include an adequate supply of blood, 
an autotransfuser, vascular instruments, skilled 
assistance, large-bore venous access above the 
diaphragm, rolled packs, stick sponges, ade- 
quate suction, intravascular balloon occlusion 
catheters, and 4-0 vascular suture on large 
needles. Preliminary aortic control is obtained 
if a major arterial injury is suspected and the 
patient should be placed in a slight, reverse 
Trendelenburg position to obviate venous air 
embolism (Bricker and colleagues, 1971). As 
the hematoma is opened, massive hemorrhage 
should be expected. When encountered, it 
must be immediately tamponaded with a 
tightly rolled pack held by an assistant, until 
more definitive control is established with 
clamps or occlusion catheters. In most wounds 
of the IVC, attempts to obtain remote proxi- 
mal and distal control, before entering the 



area of caval wounding, are not valuable 
because of the abundant collateral circulation. 

Wide exposure of the caval wound and any 
associated injury is of the utmost importance. 
Wounds of the vena cava from the immediate 
subhepatic segment to the bifurcation are best 
exposed by a mobilization of the duodenum, 
the head of the pancreas, the right colon, and 
the base of the mesentery from the cecum to 
the duodenojejunal flexure (Fig. 17-2) . This 
combined rotational maneuver provides expo- 
sure, not only of the entire vena cava below 
the liver but also of any associated aortic or 
renovascular injury below the origin of the 
superior mesenteric artery (Cattell and 
Braasch, 1960; Hunt and colleagues, 1971; 
Mattox and colleagues, 1975; Feliciano, 1988) . 
In the course of this wide exposure, if a major 
bleeding source is encountered, the mobi- 
lization maneuvers are interrupted while a 
tamponading pack is placed and held by an 
assistant over the source of hemorrhage. Full 
mobilization is then completed. It has been 
observed that in most cases of caval injury, 
shock is not maximal at the onset of the oper- 
ation but during that portion of the proce- 
dure when the hematoma is opened and caval 
hemorrhage and caval compression occur. 
The anesthesiologist should be forewarned of 
this possibility (Weichert and Hewitt, 1970; 
Burch and colleagues, 1998) . 

Three sections of the intra-abdominal IVC, 
the perirenal, the bifurcation, and the retro- 
hepatic areas, may require special exposure 
maneuvers beyond those listed earlier. The 
posterior elements of wounds of the perire- 
nal area may require medial rotation of one 
kidney or division of the renal vein between 
clamps to visualize the back of the cava at its 
junction with a renal vein. 

Full exposure of the caval bifurcation zone 
is best achieved by division of the right 
common iliac artery between clamps with 
subsequent arterial reanastomosis (Salam 
and Stewart, 1985) . This maneuver is more 
likely to be necessary if repair of the IVC is 
desired rather than simple caval ligation. 

Active hemorrhage from the retrohepatic 
cava, which cannot be controlled by any form of 
tamponade, may rarely require caval expo- 
sure to attempt hemostasis. Exposure of the 
retrohepatic or immediate subhepatic cava 



chl7.qxd 4/16/04 3:38PM Page 321 



17 • INJURIES OF THE INFERIOR VENA CAVA AND PORTAL VENOUS SYSTEM 



321 



Duodenum 

Portal 
vein 

Cecum 




■ FIGURE 1 7-2 

Exposure of the inferior vena cava from 
the liver to the bifurcation is best achieved 
by a medial rotation of the duodenum and 
pancreas together with the right colon 
and mesenteric base. (From Buckman RF 
Jr, Pathak AS, Badellino MM, Bradley KM: 
Injuries of the inferior vena cava. Surg Clin 
North Am 2001;81[6]:1438.) ■ 



necessitates extensive mobilization of the 
right triangular ligament, including its caval 
crossing point at the level of the right adrenal 
vein (Mattox and colleagues, 1974). For left- 
sided injuries, full incision of the left trian- 
gular ligament is necessary. Because the 
anterior surface of the retrohepatic cava is 
bound to the liver by numerous tributaries, it 
cannot be exposed except by dividing the liver 
along the interlobar plane. Although some 
have used resection of the left lateral segment 
of liver to gain access to the left anterior as- 
pect of the retrohepatic vena cava (Klein, 
Baumgartner, and Bongard, 1994) and 
Schrock, Blaisdell, and Mathewson (1968) 
have actually suggested division of the liver 
along the interlobar plane to expose the entire 
retrohepatic cava, such maneuvers cannot be 
recommended unless they constitute mere 
completions of massive traumatic fractures 
along these planes. A right thoracoabdomi- 
nal incision or preferably a median sternotomy 
often is required, in addition to the extensive 
division of hepatic suspensory ligaments 
to expose the retrohepatic cava (Klein, 
Baumgartner, and Bongard, 1994). It cannot 
be too strongly emphasized that radical hepatic 



mobilization and exposure of retrohepatic vena 
cava injuries is associated with an extremely 
high mortality and is not advisable unless 
active bleeding is present and cannot be 
contained by perihepatic packing (Beal and 
Ward, 1989; Beal, 1990; Cue and colleagues, 
1990; Buckman, Miraliakbari, and Badellino, 
2000). 



CONTROL OF HEMORRHAGE: 
INFRAHEPATIC INFERIOR VENA CAVA 

Mostwounds of the infrahepatic vena cava can 
be controlled by manual or pack pressure until 
proximal and distal dissection is carried out 
or a partial occlusion clamp applied. Care must 
be taken to avoid avulsion of lumbar veins 
during these attempts to gain clamp control 
of the caval wound (Graham and colleagues, 
1978). 

The abundant lumbar collateral circulation 
makes satisfactory proximal and distal control 
of the infrarenal cava or confluence difficult. 
In areas where there is a problem achieving 
control with clamps, intraluminal balloon 



chl7.qxd 4/16/04 3:38PM Page 322 



322 



IV • SPECIFIC VASCULAR INJURIES 



catheters may permit bleeding control with 
minimal dissection (Ravikumar and Stahl, 
1985; Buckman, Miraliakbari, and Badellino, 
2000). Both urinary catheters and Fogarty 
vascular catheters have been used for this 
purpose. The widely recommended proximal 
and distal compression of the IVC with stick 
sponges is rapid but cumbersome and diffi- 
cult to maintain during repair of the vein 
(Graham and colleagues, 1978; Feliciano, 
1988). 

The method for control of caval wounds 
used by the authors of this chapter consists of 



the immediate tamponade of the wound with 
a tightly rolled pack, followed by the slow 
rolling of the pack down the wound from one 
end, exposing small portions of the injury 
while the remainder of the injury is still com- 
pressed. As the opalescent venous intima is 
visualized, revealing the location of the wound 
edges, Babcock clamps are applied sequen- 
tially to each exposed portion until the entire 
wound has been coapted (Fig. 17-3). The 
clamps are applied vertically in most wounds 
but can be applied transversely to produce less 
caval narrowing in suitable injuries. 




■ FIGURE 1 7-3 

Method of control of the inferior vena cava wound using tightly-rolled pack and Babcock clamps. 
(From Buckman RF Jr, Pathak AS, Badellino MM, Bradley KM: Injuries of the inferior vena cava. 
Surg Clin North Am 2001;81[6]:1440.) ■ 



chl7.qxd 4/16/04 3:38PM Page 323 



17 • INJURIES OF THE INFERIOR VENA CAVA AND PORTAL VENOUS SYSTEM 



323 



Once the hemorrhage has been controlled 
by this method, the injury can be secondar- 
ily underclamped with a partial occlusion 
clamp or with Crafoord or "bulldog" clamps 
from each end, to permit exact suturing or 
patching of the defect (Fig. 17-4). Alterna- 
tively, the Babcock clamps themselves can be 
simply under-run with suture from end to end, 
with this suture line constituting the final 
repair. 

Once the anterior and lateral aspects of the 
wound have been controlled, lumbar veins can 
be divided between ligatures and, if necessary, 
the cava can be rotated to repair the poste- 
rior parts of the wounds. The best "control" 
for caval hemorrhage is with a rapid repair, 
which can then be revised if improvement of 
the initial closure is deemed necessary. 



CONTROL OF HEMORRHAGE: 
RETROHEPATIC INFERIOR VENA CAVA 

The extreme dangers associated with expo- 
sure of the retrohepatic vena cava have been 
previously described. The most dire problems 
with the initial control of caval hemorrhage 
are those that occur following wide hepatic 
mobilization and decompression of retro- 
hepatic caval wounds (Carmona, Peck, and 
Lim, 1984; Beal and Ward, 1989; Buechter 
and colleagues, 1989; Beal, 1990; Cue and 
colleagues, 1990; Burch and colleagues, 1998) . 



This disaster is best avoided by reinforcing the 
structures capable of tamponading a retro- 
hepatic bleeding site rather than destroying 
them by mobilizing the liver (Weichert and 
Hewitt, 1970; Sharp and Locicero, 1992). 

In cases of transparenchymal hepatic 
venous or caval hemorrhage, containment 
may be restored by omental packing, deep liver 
sutures, and perihepatic gauze packing 
(Weichert and Hewitt, 1970; Stone and Lamb, 
1975; Carmona, Peck, and Lim, 1984; Beal, 
1990; Cue and colleagues, 1990; Fabian and 
colleagues, 1991 ) . Intracaval shunts generally 
do not constitute a method of initial he- 
morrhage control (Shrock, Blaisdell, and 
Mathewson, 1968; Beal and Ward, 1989; Burch 
and colleagues, 1998). In order to place the 
shunt, if its use is deemed indispensable, bleed- 
ing control should be achieved by tampon- 
ade before the insertion of the shunt (Bricker 
and colleagues, 1971; Wiencek and Wilson, 
1986; Rovito, 1987; Beal and Ward, 1989; 
Burch and colleagues, 1998) . However, if 
bleeding can be stopped by packing, the use 
of the atriocaval shunt is required only to 
attempt a direct suture repair of the suspected 
caval or hepatic vein injury (Shrock, Blaisdell, 
and Mathewson, 1968) . There is no evidence 
supporting the need for venous repair in this 
area of the cava, and strategies that seek such 
repairs are associated with mortality rates of 
70% to 90%. (Kudsk, Sheldon, and Lim, 1982; 
Moore, Moore, and Seagraves, 1985; Beal and 




■ FIGURE 1 7-4 

Technique of controlling caval wound by partially underclamping the wound from each end to 
control lumbar inflow. (From Buckman RF Jr, Pathak AS, Badellino MM, Bradley KM: Injuries of the 
inferior vena cava. Surg Clin North Am 2001 ;81[6]:1441.) ■ 



chl7.qxd 4/16/04 3:38PM Page 324 



324 



IV • SPECIFIC VASCULAR INJURIES 



Ward, 1989; Burch and colleagues, 1998; 
Cogbill and colleagues, 1998) . 

Direct clamping of the suprahepatic and 
infrahepaticvena cava, together with the appli- 
cation of portal inflow occlusion by a Pringle 
maneuver, has also been advocated by some 
as a method to limit retrohepatic bleeding in 
pursuit of the strategy of direct repair of 
the retrohepatic vena cava (Yellin, Chaffee, 
and Donovan, 1971; Klein, Baumgartner, 
and Bongard, 1994) . Vascular isolation by this 
technique carries the danger of triggering a 
cardiac arrest in a severely hypovolemic 
patient (Weichert and Hewitt, 1970; Klein, 
Baumgartner, and Bongard, 1994). Like all 
methods used to approach and suture retro- 
hepatic vena caval injuries, its use is associ- 
ated with an extremely high likelihood of 
death and cannot be recommended in any 
case in which retrohepatic hemorrhage has 
ceased spontaneously or can be contained by 
perihepatic packing. 

Venovenous bypass (Baumgartner and col- 
leagues, 1995) and hypothermic circulatory 
arrest (Shrock, Blaisdell, and Mathewson, 
1968; Carmona, Peck, and Lim, 1984) have 
also been reported to permit vascular isola- 
tion and repair of the retrohepatic vena cava 
in rare cases. These are not hemorrhage 
control tactics but are practicable only when 
prior control of the caval injury by manual 
or pack tamponade has gained sufficient 
time to institute the bypass procedure. As is 
true for the techniques of atriocaval shunting 
and clamp vascular isolation of the liver, their 
sole value is in pursuing the highly dubious 
goal of direct suture repair of the retrohepatic 
cava. 

Nearly all successful repairs of retrohepatic 
vena caval injuries, in case reports describing 
actual clinical events, have occurred in patients 
who had stable hematomas at the time of oper- 
ation (Bricker and Wukash, 1970; Burns and 
Sherman, 1972; Fullen and colleagues, 1974; 
DePinto, Mucha, and Powers, 1976; Mullin, 
Lucas, and Ledgerwood, 1980; Misra, Wagner, 
and Boneval, 1983; Rovito, 1987; Hartman 
and colleagues, 1991; Baumgartner and 
colleagues, 1995; Feldman, 1996). These 
hematomas, having been disrupted by hepatic 
mobilization, released a massive hemorrhage 
that was then, in thefortunatepa.tients, stopped 



by manual tamponade. Control by tampon- 
ade allowed time for the insertion of an intra- 
caval shunt or for the institution of venovenous 
bypass in pursuit of direct suture repair of the 
caval injury. This sequence permitted, in a few 
lucky patients, successful repair of the injuries. 
Aside from case reports such as those 
described earlier, it is doubtful whether there 
are any successful applications of these tech- 
niques. Despite the occasional technical fea- 
sibility of carrying it out, there is no evidence 
that injuries of the retrohepatic or immedi- 
ate subhepatic vena cava, associated with 
spontaneously contained hematomas, require 
repair to prevent recurrent hemorrhage or 
thromboembolic complications. All strategies 
and techniques designed to effect such repairs, 
at the cost of disrupting a stable hematoma, 
are ill founded and are less likely to produce 
survival than methods that produce tampon- 
ade or reinforce the spontaneously occurring 
containment of retrohepatic hemorrhage 
(Buckman, Miraliakbari, and Badellino, 
2000). 



Inferior Vena Cava Repair 

At the outset of the discussion of definitive 
caval repair, it is necessary to emphasize three 
important facts: First, patients who do not die 
of uncontrolled intraoperative hemorrhage 
or the consequences of prolonged shock 
tend to be long-term survivors regardless of 
the method of managing the caval injury 
(Weichert and Hewitt, 1970; Bricker and col- 
leagues, 1971; Graham and colleagues, 
1978; Wiencek and Wilson, 1986; Burch and 
colleagues, 1998). Second, complications of 
caval repairs or of the expectant management 
of spontaneously tamponaded caval injuries 
are very uncommon (Beal, 1990). Third, 
the long-term outcome for ligation of the 
infrarenal IVC is about the same as that for 
repair (Duck, Jones, and Shires, 1965; Quast 
and colleagues, 1965; Weichert and Hewitt, 
1970; Graham and colleagues, 1978; Burch 
and colleagues, 1998). It follows that mini- 
mization of the shock period and rapid 
control of active caval hemorrhage are the 
principal goals to be pursued in the defini- 
tive operative management of wounds of the 



chl7.qxd 4/16/04 3:38PM Page 325 



17 • INJURIES OF THE INFERIOR VENA CAVA AND PORTAL VENOUS SYSTEM 



325 



intra-abdominal IVC. Sometimes, as indi- 
cated earlier in this chapter, these goals can 
be met without exposing or suturing the caval 
wound. 

Caval wounds that have demanded expo- 
sure can be rapidly repaired, in most cases, 
using a lateral suture technique with 4-0 
cardiovascular suture material (Fullen and 
colleagues, 1974; Burch and colleagues, 1998) . 
The posterior portion of a wound may be 
accessed by extension of the anterior wound 
or by rotation of the cava. This type of repair, 
in a patient with severe or multiple injuries 
and shock, is preferable to struggling with fine 
suture material on a tiny needle or attempt- 
ing elegant caval reconstruction in locations, 
such as the bifurcation or the infrarenal vena 
cava, where precise repair has no provable 
impact on outcome. After lateral repair, if the 
narrowing of the cava is deemed unaccept- 
able and the patient is stable, revision by patch 
angioplasty or graft replacement to restore 
luminal diameter can be considered. However, 
the need for revision is not common. 

Because retrohepatic caval injuries are best 
managed expectantly or by tamponade, and 
because there is no credible evidence that caval 
narrowing or even ligation (Mullins, Lucas, 
and Ledgerwood, 1980; Burch and colleagues, 
1998) below the renal veins affects long-term 
outcome, the issue regarding complex recon- 
struction of the vena cava really devolves down 
to injuries of the suprarenal and perirenal seg- 
ments. In these sections, it is not known how 
much narrowing of the vena cava can be tol- 
erated, although caval ligation above the renal 
veins is claimed to be incompatible with sur- 
vival. A reduction of up to 75% of the luminal 
cross section probably would be tolerated, but 
this cannot be stated with certainty (Burch 
and colleagues, 1998). In the absence of cer- 
tainty, it is advisable that unless the patient 
is exsanguinating, a lumen of at least 25% or 
more should be preserved during repair of 
the suprarenal or perirenal IVC. After the 
initial repair, if the lumen is believed to be 
less than this and the hemodynamic condi- 
tion of the patient permits, patch angioplasty, 
using vein or polytetrafluoroethylene (PTFE) 
can be done (Klein, Baumgartner, and 
Bongard, 1994) . Very rarely the replacement 
of a damaged segment of the perirenal or 



suprarenal IVC with panel grafts of vein or 
externally supported PTFE may be justified 
(Fig. 17-5). 

Revision of narrowed repairs of the 
infrarenal vena cava cannot be easily justified 
because there is no evidence that the long- 
term outcome is better with patent than with 
thrombosed repairs or with any repair rather 
than ligation. In severe caval wounds, espe- 
cially with profound shock and multiple vas- 
cular injuries, ligation of the infrarenal vena 
cava or bifurcation, with separate ligation or 
clipping of any lumbar veins entering the 
wounded segment, is an acceptable method 
of management (Duke, Jones, and Shires, 
1965; Agarwal and colleagues, 1982; Moore, 
Moore, and Seagraves, 1985; Burch and 
colleagues, 1998) . All evidence suggests that 
rapid enlargement of existing abundant 
lumbar collaterals and the ascending lumbar 
veins will allow the continuation of caval flow 
around the area of ligation. It has been 
reported that even suprarenal caval ligation 
can be safely carried out, if the pressure in 
the vena cava below the ligature does not rise 
above 30 cm of saline and if indigo carmine 
excretion by the kidney, following intravenous 
administration, is demonstrated (Caplan, 
Halasz, and Bloomer, 1964). The same 
network of collaterals to the azygous and hemi- 
azygous systems that permits infrarenal liga- 
tion may also prove adequate in the case of 
suprarenal ligation. The evidence on this 
subject is not sufficient for a definite conclu- 
sion to be drawn. 

For an exposed but irreparable injury of 
the suprarenal IVC in a patient unable to 
withstand complex reconstruction, an alter- 
native to ligation might be placement of a 
temporary heparin-bonded shunt. In an 
experimental model, caval shunts have 
maintained their patency for up to 24 hours 
(Aldridge, Buckman, and Badellino, 1997). 
Clinical experience with this desperate 
expedient is limited (Burch and colleagues, 
1998). 



Postoperative Management 

Regardless of whether the IVC is repaired or 
ligated or managed by tamponade, stagnation 



chl7.qxd 4/16/04 3:38PM Page 326 



326 IV • SPECIFIC VASCULAR INJURIES 



IVC 






Saphenous 
vein segment 




Completed 
graft 




IVC with graft 
in place 



■ FIGURE 1 7-5 

A-D, Reconstruction of a destroyed suprarenal segment of the inferior vena cava using a panel 
graft of saphenous vein. (From Buckman RF Jr, Pathak AS, Badellino MM, Bradley KM: Injuries of 
the inferior vena cava. Surg Clin North Am 2001;81[6]:1444.) ■ 



of blood in the lower extremities is undesir- 
able. Leg elevation, elastic bandage wrapping, 
and sequential compression devices promote 
venous flow and may reduce thromboembolic 
complications at and below the caval repair. 
Whether the use of anticoagulants improves 
the outcome of narrowed IVC repairs is not 
known, but a postoperative infusion of dextran 
for 24 hours is empirically used by many sur- 
geons. Edema of the lower extremities may 
occur in the early postoperative period 
following caval repair or ligation, but it is 
almost never a long-lasting or severe problem 



(Mullins, Lucas, and Ledgerwood, 1980; 
Burch and colleagues, 1998) . 

Sudden death due to pulmonary embolism 
has been reported to occasionally occur fol- 
lowing IVC repair, especially in patients older 
than 50 years (Burch and colleagues, 1998) . 
Use of vena cava filters, placed above the 
repair, may be considered in this subgroup. 
The actual incidence of subclinical thrombotic 
complications following caval repair or liga- 
tion has not been determined by systematic 



chl7.qxd 4/16/04 3:38PM Page 327 



17 • INJURIES OF THE INFERIOR VENA CAVA AND PORTAL VENOUS SYSTEM 



327 



Summary 

Injuries of the IVC, whether caused by blunt 
or penetrating mechanisms, are highly lethal. 
Patients who arrive in shock and fail to 
respond to initial resuscitative measures, those 
who are still actively bleeding at the time of 
laparotomy, and those with wounds of the 
retrohepatic vena cava have a low probability 
of survival. Death is most commonly due to 
intraoperative exsanguination. Knowledge 
of the anatomy and exposure techniques 
for the five segments of the intraabdominal 
vena cava is very important to the trauma 
surgeon. Although some wounds of the vena 
cava are best left unexplored, especially 
those of the retrohepatic vena cava, most 
injuries below this level can be exposed and 
repaired by lateral suture technique. Preser- 
vation of a lumen of at least 25% of normal 
is probably important in the suprarenal 
vena cava but is of no provable value below 
the renal veins. There is no evidence sup- 
porting the need to expose and repair wounds 
of the vena cava that have spontaneously 
stopped bleeding. Such wounds, especially in 
the retrohepatic area, maybe managed expec- 
tantly provided there is no strong suspicion 
of an associated injury to a major artery or 
hollow viscus. 



1975) . Portal vein injuries are caused in 90% 
of cases by penetrating trauma (Mattox, 
Espada, and Beall, 1974; Bostwick and 
Stone, 1975; Graham, Mattox, and Beall, 1978; 
Peterson, Sheldon, and Lim, 1979; Busuttil 
and colleagues, 1980), with gunshot wounds 
more commonly the cause of the injury and 
far more lethal than stab wounds. Not only 
has there been no decrease in mortality from 
this form of trauma over the last 20 years, but 
the case-fatality rate may actually be increas- 
ing, despite all the advances in prehospital and 
hospital care, because of the increased fre- 
quency of gunshot as the wounding mecha- 
nism and the greater destructive power of 
wounding weapons. 



Surgical Anatomy 

The portal system drains the splanchnic 
territories supplied by the celiac and mesen- 
teric arteries. Collecting effluent from the 
unpaired abdominal viscera, the portal vein 
delivers this blood, rich in oxygen and nutri- 
ents, to the liver, accounting for nearly 80% 
of total hepatic blood flow. Despite its high 
flow, portal pressure is normally less than 
6mm Hg. 



PORTAL VEIN INJURIES 



Wounds of the portal vein, though un- 
common, represent one of the most highly 
lethal of all vascular injuries. The reported 
case-fatality rate among patients with such 
wounds who reach the hospital alive has been 
39% to 71% in most series (Chisholm and 
Lenio, 1972; Mattox, Espada, and Beall, 1974; 
Bostwick and Stone, 1975; Graham, Mattox, 
and Beall, 1978; Peterson, Sheldon, and Lim, 
1979; Busuttil and colleagues, 1980; Stone, 
Fabian, and Turkelson, 1982; Sheldon and 
colleagues, 1985; Dawson, Johansen, and 
Jurkovich, 1991; Jurkovich and colleagues, 
1995). This high death rate is due mainly 
to intraoperative exsanguination during 
attempts to control the injured vessel (Mattox, 
Espada, and Beall, 1974; Bostwick and Stone, 



PORTAL VEIN 

The portal vein forms by the confluence of 
the superior mesenteric vein and the slightly 
smaller splenic vein behind the upper third 
of the neck of the pancreas (Fig. 17-6). This 
confluence is located just to the right of the 
body of the second lumbar vertebra and imme- 
diately anterior to the left border of the vena 
cava. The inferior mesenteric vein, the third 
major tributary contributing its flow to the 
portal veinjoins either the splenic or the supe- 
rior mesenteric vein in the immediate vicin- 
ity of the major confluence. In as many as 30% 
of patients, the inferior mesenteric vein enters 
at the angle of the major confluence itself 
(Ivatory and colleagues, 1987) . The retropan- 
creatic confluence zone is not intimately 
related to the superior mesenteric artery, the 
bile duct, or the hepatic artery. A sound knowl- 
edge of the anatomy of the portal confluence 



chl7.qxd 4/16/04 3:38PM Page 328 



328 



IV • SPECIFIC VASCULAR INJURIES 



Anterior and posterior superior %^ 
pancreaticoduodenal veins 




Superior 

mesenteric 

vein 



■ FIGURE 1 7-6 

Anterior (A) and posterior (B) views of the portal vein and its major tributaries in relation to the 
pancreas. Wounds of the retropancreatic confluence zone are the most difficult portal injuries to 
control and repair. (From Buckman RF Jr, Pathak AS, Badellino MM, Bradley KM: Portal vein injuries. 
Surg Clin North Am 2001;81[6]:1450.) ■ 



(and potential anomalies) is of utmost impor- 
tance to the trauma surgeon attempting to 
manage injuries in this dangerous and unfa- 
miliar area. 

From its origin, the valveless portal vein 
passes cephalad, inclining slightly rightward 
over its course of 3 to 4 inches, to reach the 
hilum of the liver, where it divides extrahep- 
atically into right and left branches. During 
its course, it passes, in succession, behind the 
upper pancreatic neck, and the first portion 
of the duodenum. Then, upon entering the 
hepatoduodenal ligament, it comes into rela- 
tionship with the hepatic artery and bile duct, 
lying behind these structures and forming the 
anterior border of the foramen of Winslow. 
Throughout its length, the portal vein lies 



immediately anterior to the suprarenal 
segment of the IVC. 

In addition to its main tributaries, the portal 
vein receives the pyloric vein from the pan- 
creas and duodenum, the coronary (left 
gastric) vein, and the superior pancreatico- 
duodenal vein (see Fig. 17-6). A cystic vein, 
if present, also drains into the portal vein. 
These veins represent major potential collat- 
erals in cases of portal vein obstruction. 



SUPERIOR MESENTERIC AND 
SPLENIC VEIN 

In addition to the anatomy of the portal vein 
itself, certain features of its major tributaries 



chl7.qxd 4/16/04 3:38PM Page 329 



17 • INJURIES OF THE INFERIOR VENA CAVA AND PORTAL VENOUS SYSTEM 



329 



are important to the trauma surgeon. The 
superior mesenteric vein, representing the 
confluence of all the tributaries that corre- 
spond to the branches of the superior mesen- 
teric artery, is formed in the mesentery as 
numerous intestinal veins. The ileocolic, 
right colic, and middle colic veins join the 
main venous trunk. The mesenteric vein 
also receives the right gastroepiploic vein, the 
inferior pancreaticoduodenal vein, and in 
some cases, the inferior mesenteric vein. In 
addition, a number of small unnamed veins 
drain into the right lateral aspect of the 
superior mesenteric vein from the head of 
the pancreas. 

The main trunk of the superior mesenteric 
vein passes anterior to the third portion of 
the duodenum and in front of the uncinate 
process. Then, it courses behind the neck of 
the pancreas to enter confluence zone where 
it converges with the splenic vein to form 
the main portal vein. The proximal portion 
of the superior mesenteric vein is located in 
a groove of the pancreas behind the neck and 
may be completely encircled by the pancre- 
atic tissue. The numerous tributaries enter- 
ing the superior mesenteric vein all along its 
course provide abundant collateral pathways 
in the event of obstruction of this vein. Of par- 
ticular importance are the gastroepiploic 
vein and the inferior pancreaticoduodenal 
vein, which join the superior mesentericjust 
before its confluence with the splenic vein, 
and communicate with the portal vein above 
the confluence. 

The most important distinctions regarding 
the superior mesenteric vein, from the trauma 
surgical standpoint, are first that a part of the 
superior mesenteric vein is retropancreatic 
and difficult to expose, while most of it is infra- 
pancreatic and easily accessible; and second, 
that it has abundant collaterals. 

The splenic vein, as it courses along the 
dorsum of the pancreas, receives many small 
pancreatic branches and often receives the 
inferior mesenteric vein just prior to the con- 
fluence of the splenic vein with the superior 
mesentericvein. In addition to the splenic and 
pancreaticoduodenal veins, a large accessory 
pancreatic vein may be present and may enter 
the portal vein directly. 



COLLATERALS IN PORTAL 
OBSTRUCTION 

It is obvious from the regional vascular 
anatomy that in the event of portal obstruc- 
tion in the hepatic hilum, there would be vir- 
tually no way for antegrade portal flow to be 
reconstituted. In such a case, the portosystemic 
collaterals would expand to drain the efflu- 
ent of the portal circulation. However, it is 
equally evident that the closer any of the major 
veins (i.e., portal, mesenteric, or splenic) is 
obstructed to the confluence of these veins, 
the more abundant are the potential major 
collateral veins that could reconstitute portal 
flow. The regional vascular anatomy makes it 
apparent that any retropancreatic injury of the 
portal vein and its main tributaries could be ligated, 
with probable preservation, not only of adequate 
splanchnic drainage, but also, with the expectation 
of collateral antegrade portal flow. 



Patterns of Injury 

ASSOCIATED INJURIES 

Because of the dense crowding of major vessels 
and viscera in the upper midabdomen, pen- 
etrating portal venous injuries are almost 
always associated with injury to the liver, biliary 
tract, pancreas, duodenum, or bowel. Of far 
greater concern, however, than these visceral 
injuries, are major vascular wounds of the JVC, 
aorta, superior mesenteric artery, or renal 
vessels, which accompany portal vein wounds 
in 70% to 90% of patients (Mattox, Espada, 
and Beall, 1974; Bostwick and Stone, 1975; 
Graham, Mattox, and Beall, 1978; Perterson, 
Sheldon, and Lim, 1979; Stone, Fabian, and 
Turkelson, 1982; Jurkovich and colleagues, 
1995). The suprarenal vena cava, which lies 
just behind the entire course of the portal vein, 
is the most common vessel to be injured in 
association with the portal vein, as might be 
expected. In rare cases, the simultaneous 
wounding of a major artery and the portal 
vein may lead to an arteriovenous fistula 
(Smith and Northrop, 1976; Dingledin, 
Proctor, and Jaques, 1977; Robb and 
Costa, 1984; Epstein and colleagues, 1987; 



chl7.qxd 4/16/04 3:38PM Page 330 



330 



IV • SPECIFIC VASCULAR INJURIES 



Deitrick and colleagues, 1990; Lumsden and 
colleagues, 1993). 

Associated major vascular injuries are nearly 
always posterior to the plane of the portal vein 
itself and may be multiple, involving both 
arteries and veins and tending, together with 
the portal venous wound, to produce massive 
and chaotic retropancreatic hemorrhage, 
which is extremely difficult to control. 

Portal venous wounds have been reported 
to involve the supraduodenal and retropan- 
creatic zones of the vein with similar frequency 
(Peterson, Sheldon, and Lim, 1979; Sheldon 
and colleagues, 1985; Dawson,Johansen, and 
Jurkovich, 1991; Jurkovich and colleagues, 
1995) . Hilar wounds are more rare. Whereas 
stab wounds tend to produce limited portal 
lacerations or clean transections, high-energy 
gunshot wounds striking the portal vein 
produce extensive avulsions or transection of 
the impacted vessel and may disrupt impor- 
tant potential collateral pathways, as well as 
the main trunk of the portal vein. Damage to 
other viscera and vessels is also more severe 
with gunshot wounds. Wounds of this type, 
involving the retropancreatic portal conflu- 
ence zone, are almost uniformly fatal due to 
rapid transpancreatic exsanguination (Stone, 
Fabian, and Turkelson, 1982). 



Initial Assessment 
and Management 

Most patients with portal venous injuries 
arrive at the hospital in hemorrhagic shock, 
with many in advanced circulatory collapse 
(Mattox, Espada, and Beall, 1974; Bostwick 
and Stone, 1975). Approximately one half 
of such patients will respond to initial fluid 
resuscitation, and these will usually be found 
at operation to have achieved some degree of 
spontaneous tamponade of their vascular 
wounds. The remaining patients have active 
hemorrhage and require immediate opera- 
tion as an indispensable element of their 
resuscitation. In patients with the most dire 
degrees of hemodynamic collapse, resuscita- 
tive thoracotomy and aortic cross clamping 
may be required in the emergency depart- 
ment. Few such patients will survive. More 
commonly, immediate transport to the 



operating room for emergency laparotomy is 
required. 

Exposure and Initial 
Vascular Control 

Even when accompanied by other vascular 
wounds, especially to the IVC, portal injuries 
may present at operation as stable hematomas 
of the upper central retroperitoneum, hepa- 
toduodenal ligament, or mesenteric root. In 
perhaps one half of patients, active intraperi- 
toneal bleeding is continuing at the time of 
laparotomy. This bleeding may have its origin 
from the portal vein injury itself or from an 
associated vascular wound. 



STABLE HEMATOMA 

If there is a stable hematoma, determination 
of the location of the points of retroperitoneal 
penetration and consideration of the proba- 
ble wound tract may allow a deduction to be 
reached concerning the vessels most likely to 
be wounded. Before opening the hematoma, 
it is prudent to prepare the equipment that 
may be necessary to control multiple vascu- 
lar injuries. Vascular instruments, balloon 
occlusion catheters, stick sponges, tightly 
rolled laparotomy pads, and an adequate 
supply of blood for transfusion must be at 
hand. If an autotransfusion apparatus is avail- 
able, it should be prepared for use. When a 
major arterial injury is suspected, preliminary 
control of the aorta is desirable. It is also wise 
to place the patient in a mild reverse Tren- 
delenburg position to obviate the possibility 
of venous air embolism if a caval injury is 
disclosed. 

In penetration of the region of the portal 
vein, multiplevasculdj: injuries are the rule, and 
an apparently minor hematoma with modest 
free hemoperitoneum may harbor major 
wounds of both the portal vein and the vena 
cava, which will bleed most impressively once 
unroofed. The vast majority of patients who 
die from portal vein injuries exsanguinate 
intraoperatively, after exposure of their vas- 
cular wounds. This misfortune may befall even 
those who were stable preoperatively. Because 



chl7.qxd 4/16/04 3:38PM Page 331 



17 • INJURIES OF THE INFERIOR VENA CAVA AND PORTAL VENOUS SYSTEM 



331 



the prevention of this type of death is the major 
problem in the management of injuries of 
the portal vein, the issue of vascular control 
assumes the highest importance. 

The initial procedure upon entry into the 
abdomen of a severely shocked patient with 
active transpancreatic hemorrhage is to man- 
ually compress the aorta at its hiatus and then 
to locate and manually compress the site of 
bleeding. As these things are done, volume 
repletion and the transfusion of blood may 
be performed, to remove the patient from the 
immediate danger of cardiac arrest. 

Some reduction in flow in the portal vein 
and other vessels in the region may be 
obtained by double clamping the aorta, both 
above the celiac axis and below the renal 



vessels. This maneuver will reduce, but not 
abolish, flow through the celiac and superior 
mesenteric arteries, aorta, vena cava, and renal 
vessels and will indirectly decrease portal vein 
flow. 



SUPRAPANCREATIC EXPOSURE 

In patients with suspected injury to the portal 
vein or its major tributaries, wide exposure to 
locate and immediately control the sources 
of hemorrhage is crucial. Wounds of the supra- 
pancreatic portal vein can be exposed by a 
wide Kocher maneuver with rotation of the 
hepatic flexure of the colon as needed (Fig. 
17-7). If a major source of hemorrhage is 



Bile duct 



Hepatic artery 

Portal vein 




■ FIGURE 1 7-7 

Exposure of the retropancreatic portal vein and vena cava by a combined medial rotation of the 
pancreas, duodenum, and hepatic flexure. (From Buckman RF Jr, Pathak AS, Badellino MM, 
Bradley KM: Portal vein injuries. Surg Clin North Am 2001;81[6]:1455.) ■ 



chl7.qxd 4/16/04 3:38PM Page 332 



332 IV • SPECIFIC VASCULAR INJURIES 




■ FIGURE 1 7-8 

Control of a suprapancreatic portal vein injury 
using intraluminal catheters for control of the 
bifurcation and a clamp proximally. A wound of 
this type may require an interposition vein graft. 
(From Buckman RF Jr, PathakAS, Badellino 
MM, Bradley KM: Portal vein injuries. Surg Clin 
North Am 2001 ;81[6]:1456.) ■ 



encountered, it must be immediately con- 
trolled with a pack held by an assistant. Fol- 
lowing preliminary hepatic inflow occlusion 
and the division of the cystic duct to facilitate 
exposure, the suprapancreatic portal vein may 
be dissected to obtain distal control with avas- 
cular clamp or occlusion catheter (Peterson, 
Sheldon, and Lim, 1979) (Fig. 17-8). 



RETROPANCREATIC EXPOSURE 

Retropancreatic wounds involving the portal 
confluence or its major tributaries and supra- 
pancreatic wounds with suspected additional 
injury of the IVC or other vessels are exposed 
by a combination of the Kocher maneuver and 
mobilization of the entire right colon and 
mesenteric base, from the cecum to the duo- 
denojejunal flexure (Cattell and Braasch, 
1960; Peterson, Sheldon, and Lim, 1979). 
When combined with leftward mobilization 
of the hepatic flexure, this maneuver provides 
access to the entire portal vein and the prox- 
imal portions of its major tributaries. It also 
exposes the entire infrahepatic vena cava and 
the aorta up to the origin of the superior 



mesenteric artery. Rarely, left medial rotation 
of the spleen and the tail of the pancreas may 
be necessary to expose the left lateral aspect 
of the portal vein confluence (Fish, 1966). 



PANCREATIC DIVISION 

Surgical transection of the neck of the pan- 
creas has been occasionally used as a method 
of exposing portal injuries (Stone, Fabian, and 
Turkelson, 1982) . This maneuver takes time 
and (in the opinion of the authors) is rarely 
of value in controlling retropancreatic hem- 
orrhage. Visualization of the anterior aspect 
of a portal or superior mesenteric vein injury 
is the only advantage gained by dividing the 
pancreas, and the maneuver has generally 
been performed in pursuit of precise lateral 
repair of a portal or superior mesenteric 
vein injury. It is not clear that this maneuver 
is justified unless it represents the mere com- 
pletion of a traumatic pancreatic fracture, 
which would itself have necessitated proximal 
or distal pancreatectomy. It is not a good 
emergency maneuver for hemorrhage 
control. 



CONTROL OF MULTIPLE 
VASCULAR INJURIES 

Once the retroperitoneal hematoma has been 
entered, the surgeon must be prepared to 
immediately control two or more major vascu- 
lar injuries. Clamp control of the portal injury 
is often of secondary concern. Great vessel 
lacerations deep to the plane of the portal vein 
must usually be managed first while the 
fingers of an assistant compress the portal 
venous injury, behind or above the mobilized 
pancreas. Rotation of the duodenum and 
pancreatic head during a Kocher maneuver 
provides an opportunity for broad manual 
compression of the retropancreatic portal vein 
and its major tributaries in a plane anterior 
to the great vessels of the retroperitoneum. 
This must provide immediate portal hemor- 
rhage control while deeper and more des- 
perate associated vascular injuries are 
addressed. 



chl7.qxd 4/16/04 3:38PM Page 333 



17 • INJURIES OF THE INFERIOR VENA CAVA AND PORTAL VENOUS SYSTEM 



333 



Definitive Repair 

SUPRAPANCREATIC WOUNDS 

In the suprapancreatic or hilar portions of 
the portal vein, precise lateral repair, with or 
without vein patching, or even vein graft inter- 
position, may be used after proximal and distal 
control has been obtained. In cases of com- 
bined hepatic artery and portal vein wound- 
ing, repair of the portal vein, following ligation 
of the hepatic artery, is generally recom- 
mended (Fuller and Anderson, 1978). Recon- 
struction of a divided bile duct may also be 
necessary in this location (Sheldon and col- 
leagues, 1985) . End-to-end anastomosis of the 
portal vein in the suprapancreatic zone is 
generally not feasible because, as has been 
described by Stone, Fabian, and Turkelson 
(1982), there is a loss of exposure as the two 
ends of the vein are brought together. For this 
reason, interposition saphenous vein grafting 
may be a wiser choice for the management 
of a divided suprapancreatic portal in which 
reconstruction is necessary. Ligation of the 
portal vein in this location is compatible with 
survival, provided that the hepatic artery is 
intact (Fish, 1966; Pachter and colleagues, 
1979). 



RETROPANCREATIC WOUNDS 

Wounds of the retropancreatic confluence 
zone of the portal vein offer fewer and more 
difficult options for repair. Although the 
suprapancreatic portal vein can be fully mobi- 
lized and exposed for precise repair, except 
perhaps in the hilum itself, the situation in 
the retropancreatic zone is far more chal- 
lenging. Because of the relatively medial 
location of the vein, and its fixation to the 
pancreas by its numerous tributaries, only the 
posterior aspect of the vein can be visualized 
by the standard rotation maneuvers. Visual- 
ization of the anterior portion of the vein 
requires transection of the pancreas or full 
mobilization of the confluence from the 
numerous, laterally inserting tributaries. 
Because of these hardships, as well as the near 
inevitability of major hemorrhage, and the 



difficulties of obtaining proximal and distal 
control in a vessel with so many tributaries, 
the opportunities for repair are severely 
limited in wounds of the retropancreatic zone 
(Stone, Fabian, and Turkelson, 1982) . Many 
authors who have reported lateral "repairs" 
in this zone have likely often oversewn the 
vein in a way that amounted to complete or 
near-complete obliteration of the lumen, 
not only of the portal vein itself but also of its 
major tributaries. No major complications 
have been reported from the use of this 
approach. 



PORTAL VEIN LIGATION 

A second approach to devastating wounds in 
the retropancreatic zone, and an approach 
that has been found to be life saving by expe- 
rienced surgeons, is the deliberate and 
immediate ligation of any portal injury that 
cannot be easily repaired by lateral suture. 
Despite the reports from some experienced 
surgeons that ligation has, in their hands, been 
associated with a higher mortality than lateral 
repair (Mattox, Espada, and Beall, 1974; 
Graham, Mattox, and Beall, 1978; Peterson, 
Sheldon, and Lim, 1979; Busuttil and col- 
leagues, 1980; Sheldon and colleagues, 1985; 
Jurkovich and colleagues, 1995), the reported 
experience does not permit a conclusion that 
all or any of their purported repairs remained 
patent, or that any form of repair offered a 
survival advantage compared to ligation. The 
best evidence on this subject comes from a 
large series reported by Stone, Fabian, and 
Turkelson (1982), in which survival was 
achieved in 1 7 of 20 patients in whom imme- 
diate portal ligation was carried out "when- 
ever lateral repair was impossible or 
impractical." 

Because the cause of most deaths in por- 
tal injury is uncontrolled hemorrhage, the 
method that provides the quickest definitive 
control should be preferred. Portal vein 
repair is desirable, but prolonged efforts to 
carry out complex venous repair, in the face 
of continuing blood loss and shock, to avoid 
ligation, cannot be justified by the existing 
evidence. In fact, prolonged reconstructive 



chl7.qxd 4/16/04 3:38PM Page 334 



334 



IV • SPECIFIC VASCULAR INJURIES 



efforts, followed by ligation as a desperation 
maneuver, may explain why ligation has had 
a high mortality in some series in which it 
was rarely employed (Mattox, Espada, and 
Beall, 1974; Graham, Mattox, and Beall, 1978; 
Busuttil and colleagues, 1980; Sheldon and 
colleagues, 1985; Jurkovich and colleagues, 
1995), but not in other series in which liga- 
tion was done quickly (Stone, Fabian, and 
Turkelson, 1982). 

Although a preponderance of evidence 
casts doubt on the wisdom of undertaking 
technically difficult and time-consuming 
reconstruction of most portal venous injuries, 
special circumstances make repair of the 
portal vein necessary. The first is the destruc- 
tion of the hepatic artery, as alluded to earlier 
in this chapter. When both of the hepatic 
inflow vessels are divided, one of them must 
be repaired to permit survival (Fuller and 
Anderson, 1978; Sheldon and colleagues, 
1985; Jurkovich and colleagues, 1995). Most 
authors have recommended that the portal 
vein be reconstructed in this situation. The 
second circumstance in which portal vein 
reconstruction might be unavoidable would 
be an extensive destruction of the potential 
collateral pathways, in association with tran- 
section of the portal vein itself. Under these 
rare conditions, regardless of difficulty, the 
portal vein may require reconstruction. 
Interposition grafting using saphenous 
vein (Symbas, Foster, and Scott, 1961; Stone, 
Fabian, and Turkelson, 1982), a segment 
of transposed splenic vein (Busuttil and 
colleagues, 1980), or externally supported 
PTFE to bridge a gap in the portal vein may 
be technically feasible. Alternatively, the distal 
end of the splenic vein may be anastomosed 
to the proximal stump of the superior mesen- 
teric vein (Busuttil and colleagues, 1980) (Fig. 
17-9). 

Portocaval or mesocaval shunting has 
been used to provide effluent flow from the 
intestines following portal vein ligation. 
Experience with this method has been uni- 
formly unfavorable, with nearly all patients 
becoming encephalopathic. It should not be 
considered an acceptable approach to the 
management of portal or mesenteric vein 
injuries (Fish, 1966; Stone, Fabian, and 
Turkelson, 1982). 



Postoperative Management 

The postoperative care of patients who have 
undergone portal vein reconstruction or lig- 
ation is similar to that of any patient who has 
suffered abdominal wounding with majorvas- 
cular injury and hemorrhagic shock. However, 
certain additional considerations apply to 
patients who have undergone either portal lig- 
ation or a repair of the portal vein in which 
venous narrowing has led to the threat of 
repair site thrombosis. 

Acute occlusion of the portal vein causes 
certain predictable effects, which were 
described by Child and colleagues (1952) in 
a classic series of experiments carried out a 
half-century ago and by Milnes and Child 
( 1 949) . There is a maj or but transient decrease 
in systemic blood pressure caused by pooling 
of blood in the splanchnic viscera, accompa- 
nied by a marked elevation of the portal 
pressure below the ligature or thrombo- 
sis. Massive bowel edema is common. The 
hypotensive effect of portal occlusion can be 
ameliorated by intravenous volume repletion 
in an amount sufficient to compensate for 
blood trapped in the splanchnic veins. The 
effects of splanchnic venous hypertension 
resolve as collateral pathways enlarge over 
a period of days to weeks. Chronic portal 
hypertension appears to be rare (Pachter and 
colleagues, 1979). 

Any patient who has undergone portal vein 
ligation or a venous repair that has markedly 
constricted the portal vein may be expected 
to have extraordinary fluid requirements in 
the immediate postoperative period. Replace- 
ment of this volume may be best guided with 
a pulmonary artery catheter. Most will also 
develop marked bowel edema with some small 
risk of venous intestinal infarction (Bostwick 
and Stone, 1975; Pachter and colleagues, 1979; 
Peterson, Sheldon, and Lim, 1979; Sheldon 
and colleagues, 1985). In the anticipation of 
these consequences of portal vein ligation or 
the acute thrombosis of a repair, considera- 
tion may given to skin-only suture of the 
abdominal wall at the conclusion of the pro- 
cedure or even the placement of a temporary 
abdominal wall prosthesis to avoid abdomi- 
nal compartment syndrome when the bowel 
edema becomes maximal over the first 24 to 



chl7.qxd 4/16/04 3:38PM Page 335 



17 • INJURIES OF THE INFERIOR VENA CAVA AND PORTAL VENOUS SYSTEM 335 



A 








D 

■ FIGURE 1 7-9 

Four methods of managing portal vein injuries. Of these, only lateral repair (A) and ligation (B) have 
been commonly used. End-to-end anastomosis (C)and graft (D). (From Buckman RF Jr, Pathak AS, 
Badellino MM, Bradley KM: Portal vein injuries. Surg Clin North Am 2001;81[6]:1460.) ■ 



48 hours. The use of a "second-look" proce- 
dure to inspect the bowels for viability has 
been recommended by some (Pachter and 
colleagues, 1979; Peterson, Sheldon, and 
Lim, 1979). 

Administration of anticoagulants to prevent 
mesenteric thrombosis is no trustified by exist- 
ing evidence. Follow-up of the portal vein 
repair using abdominal ultrasound has been 
recommended (Milnes and Child, 1949). 



Summary 

Wounds of the portal vein are caused most 
commonly by penetrating trauma and have 
a very high mortality. Most deaths are due to 
exsanguination, occurring intraoperatively 
as the surgeon struggles to control the hem- 
orrhage from the portal vein and associated 
vascular injuries. A thorough knowledge of 
the anatomy of the area and the likely pat- 



chl7.qxd 4/16/04 3:38PM Page 336 



336 



IV • SPECIFIC VASCULAR INJURIES 



terns of wounding is important. At operation, 
the surgeon must be prepared to deal with 
multiple vessel wounding. Although most 
authors have advocated lateral repair of the 
portal vein when it can be accomplished, 
portal ligation appears to be a safe alterna- 
tive. Complex repairs are justified only when 
a contraindication to ligation exists. Postop- 
erative care must recognize the need for extra- 
ordinary fluid replacement and the small risk 
of postoperative bowel infarction following 
repair or ligation of the portal vein. 

REFERENCES 

Agarwal N, Shah PM, Clauss RH, et al: Experience 
with 115 civilian venous injuries. J Trauma 
1982;22:827-832. 

Aldridge SA, Buckman RF, Badellino MM: 
Extended patency of intravascular shunts in an 
experimental model of vascular injury. J 
Cardiovasc Surg 1997;38:183-186. 

AsensioJA, Chahwan S, HanpeterD, etal: Operative 
management and outcome of 302 abdominal vas- 
cular injuries. Am J Surg 2001;180:528-534. 

Baumgartner F, Scudamore C, Nair C, et al: Veno- 
venous bypass for major hepatic and caval 
trauma. J Trauma 1995;39:671-673. 

Beal SL: Fatal hepatic hemorrhage: an unresolved 
problem in the management of complex liver 
injuries. J Trauma 1990;30:163-169. 

Beal SL, Ward RE: Successful atrial caval shunting 
in the management of retro-hepatic venous 
injuries. Am J Surg 1989;158:409-413. 

Bostwick J, Stone HH: Trauma to the portal venous 
system. South Med J 1975;68(11):1369-1372. 

Bricker DL, Morton JR, Okies JE, et al: Surgical 
management of injuries to the vena cava: Chang- 
ing patterns of injury and newer techniques of 
repair. J Trauma 1971 ;1 1 :725-735. 

Bricker DL, Wukasch DC: Successful management 
of an injury to the supra-renal inferior vena cava 
using an internal vena caval shunt. Surg Clin 
North Am 1970;50:999-1002. 

Buckman RF, Miraliakbari R, Badellino MM: 
Juxtahepatic venous injuries: A critical review of 
reported management strategies. J Trauma 
2000;48:978-983. 

Buckman RF, Pathak AS, Badellino MM, Bradley 
KM: Injuries of the inferior vena cava. Surg Clin 
North Am 2001;81(6) :1431-1447. 

Buechter KJ, Sereda D, Gomez G, Zeppa R: Retro- 
hepatic vein injuries: Experience with 20 cases. 
J Trauma 1989;29:1698-1704. 



Burch JM, Feliciano DV, Mattox KL, Edelman M: 
Injuries of the inferior vena cava. Am J Surg 
1998;156:548-552. 

Burns GR, Sherman RT: Trauma of the abdomi- 
nal aorta and inferior vena cava. Am Surg 1972; 
38:303-306. 

Busuttil RW, Kitahama A, Cerise E, et al: Manage- 
ment of blunt and penetrating injuries to the 
porta hepatis. Ann Surg 1980;191 (3):641- 
648. 

Caplan BB, Halasz NA, Bloomer WE: Resection and 
ligation of the supra renal inferior vena cava. 
J Urol 1964;92:25-29. 

Carmona RH, Peck DZ, Lim RC Jr: The role of 
packing and planned re-operation in severe 
hepatic trauma. J Trauma 1984;24:779-784. 

Cattell RB, Braasch JW: A technique for exposure 
of the third and fourth portions of the duode- 
num. Surg Gynecol Obstet 1960;113:379-380. 

Child CG III, Holswade GR, McClure RD Jr, et al: 
Pancreaticoduodenectomy with resection of the 
portal vein in the Macaca mulatta monkey and 
in man. Surg Gynecol Obstet 1952;94:31-45. 

Chisholm TP, Lenio PT: Traumatic injuries of the 
portal vein. Am J Surg 1972;124:770-773. 

Cogbill TH, Moore EE, Jurkovich GL, et al: Severe 
hepatic trauma: A multicenter experience with 
1335 liver injuries.J Trauma 1998;28:1433-1438. 

Cue JL, Cryer HG, Miller FB, et al: Packing and 
Planned re-exploration for hepatic and retroperi- 
toneal hemorrhage: Critical refinements of a 
useful technique. J Trauma 1990;30:1007-1013. 

Dawson DL, Johansen KH, Jurkovich GJ. Injuries 
to the portal triad. Am J Surg 1 991 ;1 61:545- 
551. 

Deitrick J, McNeill P, Posner MP, et al: Traumatic 
superior mesenteric artery — portal vein fistula. 
Ann Vase Surg 1990;4:72-76. 

DePinto DJ, Mucha SJ, Powers PC: Major hepatic 
vein ligation necessitated by blunt abdominal 
trauma. Ann Surg 1976;183:243-246. 

Dingledin GP, Proctor HJ, Jaques PF: Trau- 
matic aorto-caval— portal-duodenal fistula: Case 
Report. J Trauma 1977;17:474-476. 

Douglas BE, Baggenstoss AH, HollinsheadWH:The 
anatomy of the portal vein and its tributaries. 
Surg Gynecol Obstet 1950;91:562-576. 

Duke JH Jr, Jones RC, Shires GT: Management 
of injuries to the inferior vena cava. Am J Surg 
1965;110:759-763. 

Epstein BM, Bocchiola FC, Andrews JC, Bester L: 
Case report: Traumatic arterio-venous fistula 



chl7.qxd 4/16/04 3:38PM Page 337 



17 • INJURIES OF THE INFERIOR VENA CAVA AND PORTAL VENOUS SYSTEM 



337 



involving the portal venous system. Clin Radiol 
1987;38:91-93. 

Fabian TC, Croce MA, Stanford GG, et al: Factors 
affecting morbidity following hepatic trauma: A 
prospective analysis of 482 injuries. Ann Surg 
1991;21:540-548. 

Feldman EA: Injury to the hepatic vein. Am J Surg 
1996;111:244-246. 

Feliciano DV: Approach to major abdominal vas- 
cular injury. J Vase Surg 1988;7:730-735. 

Feliciano DV, Bitondo CG, Mattox KL, et al: Civil- 
ian trauma in the 1980s. A one-year experience 
with 456 vascular and cardiac injuries. Ann Surg 
1984;199:717-724. 

Fish JC: Reconstruction of the portal vein: Case 
reports and literature review. Am Surgeon 
1966;32(7):472-478. 

Fullen WD, McDonough JJ, Popp MJ, Altemeier 
WA: Sternal splitting approach for major hepatic 
or retrohepatic vena cava injury. J Trauma 
1974;14:903-911. 

Fuller JW, Anderson PH: Operative management 
of combined injuries to the portal vein and 
hepatic artery. South Med J 1978;71:423-424. 

Graham JM, Mattox KL, Beall AC: Portal venous 
system injuries. J Trauma 1978;18:419-422. 

Graham JM, Mattox KL, Beall AC Jr, Debakey MD: 
Traumatic injuries of the inferior vena cava. Arch 
Surg 1978;113:413-418. 

Hartman AR, Yunis J, Frei L, et al: Profound 
hypothermic circulatory arrest for the manage- 
ment of penetrating retrohepatic venous injury: 
Case report. J Trauma 1991;31:1310-1311. 

Hunt TX, Leeds FH, Wanebo HJ, Blaisdell FW: 
Arteriovenous fistulas of major vessels in the 
abdomen. J Trauma 1971;11:483-493. 

Ivatury RR, Nallathambi M, Lankin DH, et al: Portal 
vein injuries. Ann Surg 1987;206(6):733-737. 

Jurkovich GJ, Hoyt DB, Moore FA, et al: Portal triad 
injuries. J Trauma 1995;39(3) :426-433. 

Kashuk JL, Moore EE, Millikan JS, Moore JB: 
Major abdominal vascular trauma — A unified 
approach. J Trauma 1982;22:672-679. 

Klein SR, Baumgartner FJ, Bongard FA: Contem- 
porary management strategy for major inferior 
vena caval injuries. J Trauma 1994;37:35-42. 

Kudsk KA, Sheldon GF, Lim RC Jr: Arterial-caval 
shunting (ACS) after trauma. J Trauma 1982; 
22:81-85. 

Linker RW, Crawford FAJr, Rittenbury MS, Barton 
M: Traumatic aorto-cava fistula. J Trauma 1982; 
22:81-85. 



Lumsden AB, Allen RC, Sreeram S, et al: Hepatic 
arterioportal fistula. Am Surg 1993;59:722-726. 

Mattox KL, Espada R, Beall AC: Traumatic injury 
to the portal vein. Ann Surg 1974;181(5):519- 
522. 

Mattox KL, McCollum WM, Jordon GL, et al: Man- 
agement of upper abdominal vascular trauma. 
Am J Surg 1974;128:823-828. 

Mattox KL, Whisennand HH, Espada R, Beall AC 
Jr: Management of acute combined injuries to 
the aorta and inferior vena cava. Am J Surg 
1975;130:720-725. 

Milnes RF, Child CG III: Acute occlusion by liga- 
ture of the portal vein in the macacus rhesus 
monkey. Proc Soc Exp Biol Med 1947;70:332- 
333. 

Misra B, Wagner R, Boneval H: Injuries of hepatic 
veins and retrohepatic vena cava. Am Surg 
1983;49:55-60. 

Moore FA, Moore EE, Seagraves A: Non-resectional 
managementofmajor hepatic trauma: An evolv- 
ing concept. Am J Surg 1985;150:725-729. 

Mullins RJ, Lucas CE, Ledgerwood AM: The 
natural history following venous ligation for 
civilian injuries. J Trauma 1980;20:737-743. 

Nakamura S, Tzuzuki T: Surgical anatomy of 
the hepatic veins and inferior vena cava. Surg 
Gynecol Obstet 1981;152:43-50. 

Ochsner JL, Crawford ES, Debakey ME: Injuries of 
the vena cava caused by external trauma. Surgery 
1961;49:397-405. 

Pachter HL, Drager S, Godfrey N, et al: Traumatic 
injuries of the portal vein: The role of acute lig- 
ation. Ann Surg 1979;189(4):383-385. 

Pachter HL, Spencer FC, Hofstetter SR, et al: The 
management of juxtahepatic venous injuries 
without an atriocaval shunt: Preliminary clinical 
observation. Surgery 1986;99:569-575. 

Peterson SR, Sheldon GF, Lim RC: Management 
of portal vein injuries. J Trauma 1979;19(8) :616- 
620. 

Quast DC, Shirkey AL, Fitzgerald JB, et al: Surgi- 
cal correction of injuries of the vena cava: An 
analysis of sixty-one cases. JTrauma 1965;5:1-10. 

Ravikumar S, Stahl WM: Intra-luminal balloon 
catheter occlusion for major vena cava injuries. 
J Trauma 1985;25:458-460. 

Robb JV, Costa M: Injuries to the great veins of 
the abdomen. South Afr J Surg 1984;22(4) :223- 
228. 

Rovito PF: Atrial caval shunting in blunt hepatic 
vascular injury. Ann Surg 1987;205:318-321. 



chl7.qxd 4/16/04 3:38PM Page 338 



338 



IV • SPECIFIC VASCULAR INJURIES 



Salam A, Stewart MT: New approach to wounds of 
the aortic bifurcation and inferior vena cava. 
Surgery 1985;98:105-108. 

Sharp KW, Locicero RJ: Abdominal packing for 
surgically uncontrollable hemorrhage. Arm Surg 
1992;215:467-474. 

Sheldon GF, Lim RC, Yee ES, et al: Management 
of injuries to the porta hepatis. Ann Surg 
1985;202(5):539-545. 

Shrock T, Blaisdell W, Mathewson CJr: Manage- 
ment of blunt trauma to the liver and hepatic 
veins. Arch Surg 1968;96:698-704. 

Smith CR, Northrop CH: Stab wound causing 
mesenteric-portal arteriovenous fistula: An 
unusual case with a spontaneous closure. J 
Trauma 1976;16:408-410. 

Starzl TH, Kaupp HA, Beheler EM, et al: The treat- 
ment of penetrating wounds of the inferior vena 
cava. Surgery 1962;51:195-204. 



Stone HH, Fabian TC, Turkelson ML: Wounds 
of the portal venous system. World J Surg 
1982;6:335-341. 

Stone HH, LambJM: Use of pedicled omentum as 
an autogenous pack for control hemorrhage in 
major injuries to the liver. Surg Gynecol Obstet 
1975;141:92-94. 

SymbasPN, Foster JH, Scott HW: Experimental vein 
grafting in the portal venous system. Surgery 
1961;50:97-106. 

WeichertRF, Hewitt RL: Injuries to the inferiorvena 
cava: report of 35 cases. J Trauma 1970;10:649- 
657. 

Wiencek RG, Wilson RF: Abdominal venous 
injuries. J Trauma 1986;26:771-778. 

Yellin AE, Chaffee CB, Donovan AJ: Vascular iso- 
lation in the treatment of juxtahepatic venous 
injuries. Arch Surg 1971;102:566-573. 



chl8.qxd 4/16/04 3:37PM Page 339 




Iliac Vessel Injuries 



DEMETRIOS DEMETRIADES 


JAM 


ES A. MUf 


3RAY 


JUA 


N A. ASEN 


SIO 






O 


INTRODUCTION 






o 


ANATOMY 






o 


INCIDENCE AND EPIDEMIOLOGY 






o 


CLINICAL PRESENTATION 






o 


DIAGNOSTIC INVESTIGATIONS 

Radiographic Studies 
Computed Tomography 
Angiography 






o 


OPERATIVE MANAGEMENT 

Arterial Injuries 

Venous Injuries 

Adjunct Measures for Bleeding Control 

Fasciotomy 






o 


PERIOPERATIVE MANAGEMENT 



Role of Interventional Radiology 

Complications 

Mortality 



INTRODUCTION 

Iliac vessel injuries are among some of the 
most lethal injuries sustained by trauma 



patients. Their complex anatomy and the 
often associated injuries, particularly to the 
gastrointestinal and genitourinary structures, 
may challenge the skills of even the most expe- 
rienced trauma surgeons. Rapid transport to 

339 



chl8.qxd 4/16/04 3:37PM Page 340 



340 



IV • SPECIFIC VASCULAR INJURIES 



a trauma center, prompt recognition of the 
injury, good knowledge of the local anatomy, 
and sound surgical judgment remain the 
cornerstone for survival. 



ANATOMY 

The abdominal aorta bifurcates at approx- 
imately the level of the fourth and fifth 
lumbar vertebra into two common iliac arter- 
ies. The level of the bifurcation corresponds 
roughly to the level of the umbilicus. The 
common iliac arteries course inferiorly and 
laterally through the pelvis and divide at the 
level of the sacroiliac joint into the internal 
and external iliac arteries. The ureter crosses 
over the bifurcation of the common iliac 
artery. The external iliac artery courses along 
the pelvis, exiting anteriorly beneath the 
inguinal ligament to become the common 



femoral artery. The internal iliac artery 
provides blood supply to the pelvic viscera. It 
divides at the level of the sciatic notch into 
anterior and posterior divisions. The anterior 
division includes the vesicular, obturator, 
pudendal, and inferior gluteal branches. The 
main posterior division includes the iliolum- 
bar, superior gluteal, and lateral sacral 
branches. 

The common iliac veinsjoin at the level of 
the fifth lumbar vertebra to form the inferior 
vena cava. The confluence of the two veins 
occurs below the level of the aortic bifurca- 
tion and behind the right common iliac artery 
(Fig. 1 8-1 ) . Theleft common iliac vein courses 
behind and medial to the left common iliac 
artery. The right common iliac vein passes infe- 
riorly behind the junction of the right exter- 
nal iliac and the right internal iliac artery. This 
anatomic arrangement makes combined arte- 
rial venous injuries common and complicates 
the exposure of the right common iliac vein. 



Phrenic artery 



Celiac artery 



SMA 



■ FIGURE 18-1 

Anatomy of the iliac vessels. 
Note the confluence of the two 
common iliac veins behind the 
right iliac artery and the 
relationship of the ureter with 
the bifurcation of the common 
iliac artery. ■ 




Left 
Renal vein 

Testicular 
(Ovarian) vein 



Testicular (Ovarian) artery 

IMA 

Ureter 



chl8.qxd 4/16/04 3:37PM Page 341 



18 • ILIAC VESSEL INJURIES 



341 



TABLE 18-1 

INCIDENCE OF ILIAC VESSEL INJURIES IN PATIENTS UNDERGOING LAPAROTOMY 
FOR TRAUMA 



Mech- 
anism of 
Injury 


No. of 
Laparotomies 


Patients with 
Iliac Vessel 
Injuries (%) 


Patients with 
Iliac Artery 
Injuries (%) 


Patients with 

Iliac Vein 

Injuries (%) 


Combined 
Artery-Vein 
Injuries (%) 


Gunshot wound 
Stab wound 
Blunt trauma 


1310 
638 
633 


131 (10) 

12(2) 

33 (5.5) 


64(5) 

6(1) 

30(5) 


67(5) 

6(1) 

3 (0.5) 


35(2.7) 
2 (0.3) 
1 (0.2) 



From USC Trauma Center, 1993-2000. Unpublished trauma registry data. 



Each common iliac vein is formed by thejunc- 
tion of an internal and external iliac vein. The 
external iliac vein accompanies the external 
iliac artery, and the internal iliac vein is formed 
by numerous small and delicate tributaries. 



INCIDENCE AND 
EPIDEMIOLOGY 

Though uncommon, iliac vessel injuries are 
not necessarily rare in busy urban trauma 
centers. The incidence of iliac artery injury 
varies depending on the setting. During 
World War II DeBakey and Simeone (1946) 
reported 43 iliac arterial injuries in 2471 
patients for an incidence of 1.7%. Both 
Hughes (1958) during the Korean conflict and 
Rich, Baugh, and Hughes (1970) during the 
Vietnam conflict reported incidences of 2. 3% 
and 2.6%, respectively. 

Iliac vessel injuries are reported with greater 
frequency from the civilian arena. The inci- 
dence of iliac vascular injuries in patients 
undergoing laparotomy for trauma at the Uni- 
versity of Southern California trauma center 
is shown in Table 18—1. Overall, the incidence 
of iliac vessel injuries is 10% for gunshot 
wounds, 2% for stab wounds, and 5.5% in 
blunt trauma. In a recent survey at an urban 
level I trauma center, Bongard (1990) reported 
that iliac arterial injuries represented only 10% 
of abdominal vascular injuries and less than 
2% of all vascular trauma. Mattox and col- 



leagues (1989), in a series of 5760 cardiovas- 
cular injuries in 4459 patients, reported 232 
iliac artery and 289 iliac venous injuries, for 
an overall incidence of 12% of patients or 9% 
of cardiovascular injuries. In a series of 504 
abdominal vascular injuries from the Los 
Angeles County and University of Southern 
California trauma center, there were 112 iliac 
vessel injuries (22% of all abdominal vascu- 
lar injuries) . 

Burch and colleagues (1990) in a series of 
233 patients sustaining iliac vessel injuries 
reported that the common iliac artery was the 
most frequently injured vessel, with an inci- 
dence of 40%, and both external and inter- 
nal iliac arteries accounted for 30% of these 
injuries. In the venous system, the common 
iliac vein was the most frequently injured vessel 
(48%), the external iliac vein was injured in 
32% of the patients, and the internal iliac vein 
accounted for 20% of the venous injuries. In 
this series, Burch and colleagues (1990) 
reported a 70% incidence of combined arte- 
riovenous injuries. 

In contrast to penetrating injuries, blunt 
trauma usually involves the internal iliac 
vessels and their branches. Injury to the 
common or external iliac artery following 
blunt trauma is not common, although there 
are several case reports. The usual mecha- 
nism is stretching of the vessel over the pelvic 
wall, resulting in intimal tear and possibly 
thrombosis (Fig. 18-2). In addition, direct 
laceration of a vessel may occur from a bone 
fragment. 



chl8.qxd 4/16/04 3:37PM Page 342 



342 



IV • SPECIFIC VASCULAR INJURIES 



■ FIGURE 18-2 

Blunt trauma with pelvic 
fracture. The patient had an 
absent femoral pulse. 
Angiography shows occlusion 
of the right common iliac 
artery. ■ 




CLINICAL PRESENTATION 

Penetrating injuries to the lower abdomen, 
hips, or buttock, especially in the presence of 
shock, should prompt suspicion of an associ- 
ated iliac vessel injury. Some of the signs and 
symptoms associated with these injuries 
include hemorrhagic shock, abdominal dis- 
tention, and absent or diminished femoral 
pulse. Additional evidence of injury to the 
pelvic viscera such as gross hematuria or evi- 
dence of rectal injuries should heighten con- 
cerns for the presence of iliac vascular injury. 
In most cases the diagnosis of vascular injury 
is made intraoperatively. 

Blunt injuries to the iliac arteries are typi- 
cally but not always associated with pelvic frac- 
tures. Absent or diminished femoral pulse is 
highly suggestive of injury to the common or 
external iliac arteries. Hemorrhage is typically 
due to injury to the branches of the internal 
iliac vessels. These patients may demonstrate 
signs and symptoms of severe hemorrhagic 
shock upon initial presentation requiring 
aggressive resuscitation. Other patients may 
present with gradual and persistent bleeding. 
Both these scenarios require exclusion of 
intraperitoneal hemorrhage. In the absence 
of intraperitoneal hemorrhage or peritonitis, 
these patients should undergo immediate 
angiographic evaluation and possibly 
embolization. In rare occasions with blunt 



trauma, an arterial intimal tear may remain 
undetected during the initial hospitalization, 
only to manifest at a later stage with signs of 
leg ischemia due to secondary thrombosis. 

In patients with suspected pelvic fractures, 
a thorough physical examination can deter- 
mine the stability of the pelvis by examining 
the anterior, lateral, and posterior compo- 
nents of the pelvic ring. Injudicious and 
repeated examinations can exacerbate bleed- 
ing and lead to life-threatening hemorrhage. 
Therefore once a patient has been noted to 
have an unstable pelvis by one examiner, 
further examinations by other physicians are 
contraindicated. Radiographic examination 
of the pelvis to confirm the clinical diagnosis 
should follow expeditiously and measures to 
stabilize the pelvis should be promptly insti- 
tuted. Early application of a pelvic binder or 
external pelvic fixation may contain the 
expansion of the pelvic hematoma and reduce 
bleeding (Fig. 18-3). 



DIAGNOSTIC INVESTIGATIONS 
Radiographic Studies 

Radiographic evaluation of penetrating 
injuries to the abdomen should be performed 
only if the patient is hemodynamically fairly 
stable. These investigations may include a 



chl8.qxd 4/16/04 3:37PM Page 343 



18 • ILIAC VESSEL INJURIES 



343 




■ FIGURE 18-3 

Pelvic binder may contain 
the expansion of a pelvic 
hematoma and reduce 
bleeding from a pelvic 
fracture. ■ 



plain radiograph of the abdomen and pelvis. 
The presence of missiles or fragments in the 
pelvis, especially in the presence of hypoten- 
sion, should prompt the physician to suspect 
injury to the iliac vessels (Fig. 18-4). In blunt 
trauma, radiographic findings from the pelvis 
known to be associated with increased risk of 
bleeding from the internal iliac vessels include 
the presence of symphysis pubis diastasis of 
greater than 2.5 cm, sacroiliacjoint disruption, 
and the presence of superior and inferior rami 
fractures bilaterally ("butterfly fracture"). In 
our center these patients undergo early angio- 
graphic embolization before hemodynamic 



decompensation and massive transfusions 
are required. 



COMPUTED TOMOGRAPHY 

Computed tomography continues to play a 
major role in the evaluation of hemodynam- 
ically stable blunt trauma patients. The pres- 
ence of a significant pelvic hematoma is 
suggestive of an injury to the internal iliac 
vessels or their branches. Extravasation of 
intravenous contrast is diagnostic of arterial 
bleeding or false aneurysm and requires 




■ FIGURE 18-4 

Missiles in the pelvis on 
radiographs, especially in the 
presence of shock, are 
suggestive of iliac vessel 
injury. ■ 



chl8.qxd 4/16/04 3:37PM Page 344 



344 



IV • SPECIFIC VASCULAR INJURIES 



prompt angiographic evaluation and possibly 
embolization. Lack of contrast opacifying the 
lumen of the major iliac vessels is consistent 
with thrombosis and should be investigated 
further with angiography to confirm the diag- 
nosis, provided the extremity does not appear 
threatened by ischemia. If the extremity 
appears compromised, prompt surgical explo- 
ration should be performed. 



ANGIOGRAPHY 

Angiography has dramatically improved the 
management of patients with hemorrhage 
from pelvic fractures secondary to blunt 
trauma. It should be considered early in 
patients with clinical evidence of severe bleed- 
ing from the pelvis (low hematocrit or 
hypotension) as soon as intraperitoneal 
bleeding or peritonitis has been ruled out. 
Similarly, the presence of extravasation of 
intravenous contrast on computed tomo- 
graphic scan or certain radiographic findings 
on plain pelvic films (pubis diastasis >2.5 cm, 
major sacroiliac joint disruption, and "but- 
terfly" fracture) should prompt the surgeon 
to seek an early angiogram. It is critical that 
during angiography the patient is closely mon- 
itored and resuscitated continuously under 
the supervision of a senior member of the 
trauma team. 

Angiography is able to identify the site and 
severity of bleeding and control bleeding with 



embolization. Additionally, it may identify 
occlusions or major intimal tears of the 
common or external iliac arteries that require 
operative intervention. If massive hemor- 
rhage from a major artery is identified at the 
time of angiographic evaluation, temporary 
control of the bleeding may be achieved with 
an intraluminal balloon while the patient is 
transported to the operating room (Fig. 
18-5). 



OPERATIVE MANAGEMENT 



The operative findings depend on the 
mechanism of injury, associated injuries, and 
the nature of vascular injury. In blunt trauma 
the usual finding is a zone 3 retroperitoneal 
hematoma, which may or may not be pulsatile 
or expanding. However, in some cases with 
intimal tear and thrombosis, often there is a 
small or even no local hematoma and the 
injury may be missed. Azone 3 retroperitoneal 
hematoma resulting from blunt trauma 
should not be explored routinely. Exploration 
is indicated only if there is a suspicion of iliac 
artery injury — that is, absent or diminished 
femoral pulse. 

In penetrating iliac vessel injuries upon 
entering the abdominal cavity, the surgeon 
may encounter free intraperitoneal bleeding, 
azone 3 hematoma, or a combination of both. 
It is an important surgical principle that all 



■ FIGURE 18-5 

Motor vehicle accident with 
severe pelvic fracture. 
Angiography showed massive 
bleeding from the left common 
iliac artery. Balloon occlusion of 
the injured artery achieved 
temporary control of the 
bleeding and the patient was 
taken to the operating room. ■ 




chl8.qxd 4/16/04 3:37PM Page 345 



18 • ILIAC VESSEL INJURIES 



345 



zone 3 hematomas caused by penetrating 
trauma be explored. 



Arterial Injuries 

Any active bleeding is controlled initially by 
direct compression and subsequently by prox- 
imal and distal vascular control. The iliac 
vessels may be exposed by direct dissection of 
the peritoneum over the vessels or by dissec- 
tion of the paracolic peritoneum and medial 
rotation of the right or left colon. Care should 
be taken to avoid injury to the ureter, which 
crosses over the bifurcation of the common 
iliac artery. 

The anatomic level of proximal control 
depends on the site of bleeding and the site 
and size of the hematoma. For suspected 
iliac artery injuries near the bifurcation of 
the aorta, proximal arterial control may be 
achieved with aortic cross clamping above 
the bifurcation. In more distal external iliac 
artery injuries, proximal control can be 
achieved by applying a vessel tape around 
the common iliac artery. During the dissec- 
tion, care should be taken to avoid acciden- 
tal injury to the underlying vein, especially 
the right common iliac vein. Isolation and 
control of the internal iliac artery is essential 
because the bleeding may continue despite 
proximal and distal control. The identifica- 
tion and isolation of the internal iliac artery 
can be facilitated by retracting the vascular 
tapes proximally and distally and dissecting 
toward the middle until the vessel is identi- 
fied. A similar technique of gradual dissec- 
tion of the vessel and progressive movement 
of the vascular clamps toward the injury 
can be used in cases with active bleeding or 
large hematoma, in which a direct approach 
to the injured site may be difficult. If 
the exposure of the distal iliac vessels is 
difficult, especially in men with a narrow 
pelvis, extending the midline incision by 
adding a transverse lower abdominal inci- 
sion may be necessary. In some cases with 
bleeding from the vessels near the groin or 
in the presence of a large hematoma, distal 
control can be facilitated by a longitudinal 
incision over the groin and division of the 



inguinal ligament. 



Small common or external iliac artery 
injuries can be managed by primary repair, 
using a 4-0 or 5-0 vascular suture and taking 
care to avoid significant stenosis. In the appro- 
priate cases a venous or PTFE patch may be 
necessary to avoid stenosis at the repair site. 
This patch should not be excessive in size in 
order to avoid aneurysmal dilation. In more 
extensive injuries, especially in gunshot 
wounds or blunt trauma in which debridement 
is always necessary, an end-to-end anastomo- 
sis with or without a prosthetic graft (size 6 or 
8 PTFE) may be required. It is strongly 
recommended that all vascular repairs are 
performed under loupe magnification. Local 
heparin (20 to 30 mL of solution of 100 units 
of heparin per 100 mL) should be adminis- 
tered to prevent thrombosis during the vas- 
cular repair. A balloon-tipped catheter should 
always be passed in the distal arterial tree to 
remove any clots. 

More complex procedures, such as extra- 
anatomic femorofemoral bypass or mobiliza- 
tion and use of the internal iliac artery to 
replace the external iliac artery, are time 
consuming and often technically difficult. 
Extra-anatomic bypasses may be necessary in 
patients with severe purulent peritonitis. 
Burch performed six extra-anatomic bypasses 
with poor results, including three deaths, three 
amputations, three compartment syndrome, 
and two graft thrombosis. The existing evi- 
dence suggests that the presence of enteric 
contamination is not a contraindication for 
an end-to-end repair or a PTFE interposition 
graft. In a study of 358 penetrating iliac vas- 
cular injuries, Burch reported that in general 
associated gastrointestinal or urologic injuries 
did not influence the management of vascu- 
lar injuries. However, many surgeons still 
suggest that in the presence of significant 
enteric contamination, an extra-anatomic 
bypass should be performed. Any enteric 
spillage should be controlled first and the peri- 
toneum cleaned meticulously before any 
vascular repair is performed. The peritoneum 
should be closed over the graft whenever 
possible. 

Ligation of the common or external iliac 
artery should never be considered, even in 
the most critically injured patients. Ligation 
is poorly tolerated and in most cases results 



chl8.qxd 4/16/04 3:37PM Page 346 



346 



IV • SPECIFIC VASCULAR INJURIES 



■ FIGURE 18-6 

Injuries to the right iliac artery 
and vein. Because of the 
critical condition of the patient, 
the vein was ligated (white and 
black arrows) and the artery 
was shunted with a catheter 
(white arrow). Definitive arterial 
reconstruction was performed 
24 hours later. ■ 




in ischemia of the leg and in about 50% of 
patients limb loss. The ischemia may aggra- 
vate the general condition of the patient by 
release of toxic metabolites into the systemic 
circulation. Subsequent attempts to re- 
establish blood flow may be even more dan- 
gerous because of reperfusion injury. In the 
critically ill, hypothermic, and coagulopathy 
patient, a temporary intraluminal shunt with 
semielective reconstruction of the artery at a 
later stage should be considered (Fig. 18-6). 
The fastest and cheapest way to construct a 
shunt is from a sterile intravenous or naso- 
gastric tube. The shunt is secured in place with 
proximal and distal ligatures. The incidence 
of thrombosis of the shunt is high, and the 
peripheral pulses and perfusion should be 
monitored closely. Systemic anticoagulation 
prophylaxis is usually contraindicated because 
of associated coagulopathy. 



Venous Injuries 

Iliac venous injuries may be technically 
more challenging than arterial injuries 
because of the more difficult surgical expo- 
sure and the risk of air embolism. The 
anatomic location of the right common iliac 
vein and the confluence of the two common 
iliac veins behind the right common iliac 



artery may make exposure a challenging task, 
especially in elderly patients with atheroscle- 
rosis and adhesions between the artery and 
vein. These difficulties have led some authors 
to recommend transection of the overlying 
iliac artery. We believe that such a drastic 
approach is excessive and should rarely be con- 
sidered. As a rule, satisfactory surgical expo- 
sure of the vein can be achieved by meticulous 
and adequate mobilization and retraction 
of the artery with vessel tapes. Ligation and 
division of the internal iliac artery may also 
facilitate exposure. 

Repair of iliac venous injuries should always 
be considered in cases in which it can be per- 
formed by lateral venorrhaphy without pro- 
ducing major stenosis. This is usually possible 
in more than 50% of cases. The management 
of complex venous injuries, which cannot be 
repaired by lateral venorrhaphy, is a contro- 
versial issue. Burch, in a study that included 
192 iliac venous injuries, performed only one 
PTFE reconstruction on a patient with exter- 
nal iliac vein injury. Repair associated with 
severe stenosis may result in thrombosis and 
possibly pulmonary embolism. In these cases, 
ligation of the vein maybe preferable to repair. 
Burch reported two cases with fatal embolism 
in a group of 82 patients with common or 
external iliac vein injuries treated with repair 
but none in 43 patients treated with ligation. 



chl8.qxd 4/16/04 3:37PM Page 347 



18 • ILIAC VESSEL INJURIES 



347 



Although theoretically a caval filter may 
prevent pulmonary embolism, there is no pub- 
lished clinical experience. The proponents of 
ligation argue that complex repair with spiral 
graft or other methods is time consuming and 
increases blood loss, and that there is no evi- 
dence of any improved outcome. Ligation is 
usually tolerated very well by almost all 
patients. Most patients develop transient leg 
edema, which responds well to elevation and 
elastic bandage wrapping. However, in some 
cases ligation results in massive edema and 
extremity compartment syndrome. In some 
extreme cases with massive leg and scrotal 
edema, we had to re-operate and re-establish 
the continuity of the vein with a prosthetic 
graft. 

The management of complex iliac vein 
injuries becomes even more controversial in 
patients with associated iliac artery injuries. 
Some surgeons have suggested that venous 
repair may protect the arterial repair by 
avoiding venous hypertension. However, 
many others challenge this concept and 
advocate ligation. These major injuries are 
usually associated with severe blood loss, and 
any complex procedures prolonging the 
operation may increase mortality. We believe 
that the decision to repair or ligate the vein 
should be individualized according to the 
condition of the patient and the nature of 
the venous injury. 



Adjunct Measures for 
Bleeding Control 



In some cases bleeding may persist even 
after repair or ligation of the iliac vessels. The 
source of bleeding is usually from deep vas- 
cular branches to the pelvic wall or the 
sacrum. Opening of the presacral fascia is ill- 
advised and often aggravates the bleeding. 
Damage control by packing, followed by post- 
operative angiographic embolization, should 
be considered at an early stage. Carillo and 
colleagues (1998) reported significantly 
reduced mortality in patients with iliac vas- 
cular injuries undergoing abbreviated 
laparotomy and damage control. 

Bleeding from a gunshot wound involving 
the bony pelvis can be troublesome and 
difficult to control. In these cases we have 
successfully used Foley catheter balloon 
tamponade. The balloon is inflated in the 
bone defect and the distal end of the catheter 
is brought outside the abdominal cavity 
through a small skin incision (Fig. 18-7). A 
gentle traction on the catheter is maintained 
by a clamp applied to the catheter just 
above the skin. The clamp is removed and 
the balloon is deflated 2 or 3 days postoper- 
atively, and if no bleeding occurs through 
the catheter, the Foley catheter is pulled 
out. 




■ FIGURE 18-7 

Foley catheter balloon 
tamponade of persistent 
bleeding from the posterior 
wall of the pelvis following a 
gunshot wound. The catheter 
is exiting through the left 
buttock. The balloon was 
removed 4 days later without 
any recurrent bleeding. ■ 



chl8.qxd 4/16/04 3:37PM Page 348 



348 



IV • SPECIFIC VASCULAR INJURIES 



Fasciotomy 

Therapeutic leg fasciotomy in patients with 
extremity compartment syndrome should be 
performed without any delay, even before 
reconstruction or shunting of any arterial 
injury. However, the role of liberal prophy- 
lactic fasciotomy is controversial. Although 
some authors advocate prophylactic fas- 
ciotomies in all patients with delayed arterial 
reconstruction or venous ligation, especially 
in combined arteriovenous injuries, many 
others practice a policy of "fasciotomy on 
demand." These authors suggest that fas- 
ciotomy is a procedure associated with sig- 
nificant complications and inferior cosmetic 
results and should be performed only for ther- 
apeutic purposes. In a study of 94 patients with 
fasciotomies for trauma, Velmahos and col- 
leagues (1997) reported local complications 
in 42% of patients with prophylactic fas- 
ciotomies. In 57% of these patients, primary 
closure of the wounds was not possible and 
there was a need of skin grafting. If an expec- 
tant policy is selected, the patient should be 
monitored very closely with frequent clinical 
examinations and measurements of com- 
partment pressures in the appropriate cases. 
Fasciotomy should be performed with the first 
signs of compartment syndrome. 



PERIOPERATIVE 
MANAGEMENT 

To avoid reperfusion injury following revas- 
cularization of the extremity, the surgeon must 
maintain good hydration and diuresis during 
the operation and the first few hours postop- 
eratively. In hemodynamically stable patients, 
administration of mannitol (0.5g/kg of body 
weight over 20 minutes) has many beneficial 
effects because of its oxygen free radical scav- 
enger, rheologic, and osmotic properties. 
There is evidence that early administration of 
mannitol blunts the effects of reperfusion 
injury, reduces the risk of extremity com- 
partment syndrome and the need for fas- 
ciotomy, and improves the microcirculation 
of tissues. It is our practice to administer a 
second dose 4 to 6 hours after injury. 



Mannitol is contraindicated in hypotensive 
patients because its diuretic effect may aggra- 
vate the hypotension. There is also concern 
that the vasodilating and rheologic properties 
of mannitol may increase bleeding in patients 
with active uncontrolled hemorrhage. 

The role of postoperative anticoagulation 
in uncomplicated vascular repairs or in venous 
ligations is not clear. Some authors use low- 
molecular-weight heparin prophylaxis for the 
first few days, followed by aspirin for the next 
few weeks. In patients with venous thrombo- 
sis after repair, oral anticoagulation should be 
given for at least 3 months. 

In venous injuries treated by ligation, it is 
important to elevate the leg, apply early com- 
pression elastic bandages, and monitor closely 
for extremity compartment syndrome. 



Role of Interventional 
Radiology 

Diagnostic angiography has a limited role in 
the preoperative evaluation of penetrating 
abdominal trauma. However, it may play a 
useful therapeutic role postoperatively in 
patients with incomplete hemostasis from 
deep iliac artery branches. In blunt trauma 
angiography may play a major diagnostic and 
therapeutic role. It remains the most useful 
investigation for the diagnosis of iliac artery 
thrombosis or bleeding following blunt 
trauma to the abdomen or pelvis. Bleeding 
from peripheral branches may be effectively 
controlled by embolization in most patients 
(Fig. 1 8-8) . In cases with major bleeding from 
the iliac artery, the interventional radiologist 
may be able to achieve temporary control by 
inflating an intraluminal balloon at the site 
of injury until surgical control is achieved in 
the operating room (Fig. 18-5). 

Angiographically placed stents have an 
important role in selected cases with iliac 
artery injuries. Patients with false aneurysms, 
arteriovenous fistulas, or significant intimal 
tears may benefit from this procedure. Stent- 
ing should not be attempted during the acute 
stage in patients with a thrombosed iliac artery, 
because of the risk of clot dislodgement and 
major hemorrhage in cases with a transected 
vessel. However, angiographic stenting may 



chl8.qxd 4/16/04 3:37PM Page 349 



18 • ILIAC VESSEL INJURIES 



349 




■ FIGURE 18-8 

Patient with severe pelvic 
fracture and major blood 
loss. Angiography shows two 
areas of significant bleeding 
(arrows), which were 
successfully controlled by 
embolization (right frame), m 



be a good option in patients with late iliac 
artery thrombosis. In most cases a skillful 
interventional radiologist may be able to pass 
a guidewire through the clot and deploy a stent 
(Fig. 18-9). 



Complications 

Complications directly related to the vascular 
injuries may appear early during the initial 
hospitalization or late. The overall incidence 
of early vascular complications in patients sur- 
viving for more than 24 hours is about 15% 
for arterial injuries and 12% for venous 
injuries. 



Thrombosis of the repaired artery remains 
the most common early arterial complication. 
The most important factors for early throm- 
bosis are the technique and the use of pros- 
thetic grafts. Burch reported no thrombosis 
in 25 patients with lateral suturing of the iliac 
artery. On the other hand, 25% of 16 PTFE 
grafts and 33% of 6 extra-anatomic bypasses 
thrombosed. Good surgical techniques, 
Fogarty balloon exploration and extraction 
of any clots from the peripheral arteries, intra- 
operative local heparinization, and liberal use 
of on-table angiography may reduce the inci- 
dence of early failure of the arterial repair. 
Postoperatively, the peripheral pulses and per- 
fusion should always be monitored closely and 




■ FIGURE 1 8-9 

An 18-year-old patient presenting with intermittent claudication many months after a major car 
accident for which he required a splenectomy and small bowel resection. Angiography revealed 
occlusion of the right common iliac artery (left frame). The occlusion was successfully stented by 
interventional radiology (right frame), m 



chl8.qxd 4/16/04 3:37PM Page 350 



350 



IV • SPECIFIC VASCULAR INJURIES 



emergency reoperation should be considered 
in patients with evidence of arterial thrombosis. 

Early postoperative bleeding is another 
fairly common problem. Bleeding during the 
first few hours after the operation may be due 
to a technical problem with the suture line, 
missed bleeding from a small vessel, or medical 
bleeding resulting from coagulopathy. 
Depending on the rate of bleeding and the 
coagulation status of the patient, reoperation 
may be necessary. Delayed bleeding a few days 
after the initial procedure, especially in the 
presence of other signs of infection, such as 
fever or leucocytosis, may be due to local sepsis 
and is an ominous sign. Infection of the 
repaired vessel, especially in the presence of 
a prosthetic graft, is a life-threatening com- 
plication. The controversies regarding the role 
of prosthetic grafts in a grossly contaminated 
field have already been discussed. Early recog- 
nition of the infection and aggressive anti- 
biotic treatment may salvage the graft. 
However, in advanced sepsis, especially in the 
presence of bleeding, reoperation with 
removal of the graft and ligation of the vessel 
combined with an extra-anatomic bypass 
remain the only option. 

The overall incidence and nature of early 
venous complications following iliac vein 
injury depend on the extent of venous injury 
and method of management. Generally, 
lateral repairs are associated with a lower inci- 
dence of venous complications than venous 
ligations (5% vs. 25% in the series by Burch) . 
Transient leg edema following ligation of the 
common or external iliac vein is by far the 
most common complication. The edema can 
be avoided or minimized by elevation and 
elastic wraps of the leg. Occasionally, the 
swelling is so severe that it results in extremity 
compartment syndrome, requiring fasciotomy. 

Deep venous thrombosis may occur in 
patients treated by ligation of the iliac vein or 
in cases with thrombosis of the repaired vein. 
The real incidence of early deep venous 
thrombosis is not known because no study has 
ever evaluated systematically all patients with 
iliacvenousinjuries. Ithasbeen suggested that 
anticoagulation prophylaxis and elastic wrap 
on the leg should be used in all patients with 
venous injuries. 



The most dangerous complication follow- 
ing iliac venous injury is pulmonary embolism . 
Patients with repair producing major venous 
stenosis are at risk of pulmonary embolism. 
Earlier military experience suggested that 
venous repairs may be associated with a high 
incidence of pulmonary embolism, especially 
if the lumen is narrowed more than 50 % . More 
recent civilian experience reported an inci- 
dence of about 2% of fatal pulmonary 
embolism in patients treated with venous 
repair. The role of prophylactic inferior vena 
cava filters and long-term anticoagulation has 
not been studied. It might be appropriate to 
use these modalities in cases with major steno- 
sis of the vein. 

The incidence of late complications fol- 
lowing iliac vascular trauma is not known. All 
existing studies are retrospective and lack 
systematic late follow-up. Late iliac artery 
complications include false aneurysm and 
arterial stenosis associated with intermittent 
claudication or a threatened limb. The 
method of treatment of these complications, 
such as open surgery or angiographically 
placed stents, should be individualized 
according to the age of the victim, the nature 
of the arterial pathology, and the experience 
of the trauma center. 

Late venous complications may include 
chronic venous insufficiency with leg edema 
and skin ulcers. The incidence of this com- 
plication is not known, and the reported 
figures from existing retrospective studies 
may be misleading, because those patients 
returning for late follow-up are usually the 
symptomatic ones. There is evidence that 
late venous complications are more likely to 
occur in patients treated with iliac vein 
ligation than in patients with lateral repair. 
Mullins suggested that iliac vein ligation 
does not often result in chronic venous com- 
plications, especially if elevation of the leg is 
instituted immediately after surgery. The 
rationale for this practice is that elevation 
may interrupt the cascade of events, which 
lead the vascular damage during the critical 
postinjury period. It certainly makes sense to 
elevate the leg and apply elastic wraps in 
these cases, but there is no proven evidence 
of any benefit. 



chl8.qxd 4/16/04 3:37PM Page 351 



18 • ILIAC VESSEL INJURIES 



351 



Mortality 



The mortality of iliac vascular injuries is high 
and depends on the type of vascular trauma 
(contained or free bleeding) , the presence of 
other associated injuries, the clinical condi- 
tion of the patient on admission, and the expe- 
rience of the trauma team. 

The mortality of patients undergoing emer- 
gency department thoracotomy is almost 
100%, with only very few survivors reported. 
The reported overall mortality varies from 
30% to 50% in arterial injuries and 25% to 
40% in venous injuries. In isolated vascular 
injuries, the mortality is about 20% for arter- 
ial injuries and about 10% for venous injuries. 

REFERENCES 

Asensio JA, Chahwan S, Hanpeter D, et al: Oper- 
ative management and outcome of 302 abdom- 
inal vascular injuries. Am J Surg 2000;180: 
528-534. 

Asensio JA, Lejarraga M: Abdominal vascular 
injuries. In Demetriades D, Asensio JA (eds): 
Trauma Management. Georgetown, Tex, Landes 
BioScience, 2000, pp 356-362. 

Asensio JA, Petrone P, Roldan G, et al: Analysis 
of 185 iliac vessel injuries: Risk factors and pre- 
dictors of outcome. Arch Surg 2003;138(11): 
1187-1193. 

Bongard FS, Dubrow T, Klein SR: Vascular injuries 
in the urban battleground: Experience at a 
metropolitan trauma center. Ann Vase Surg 
1990;4:415-418. 

Burch JM, Richardson RJ, Martin RR, Mattox KL: 
Penetrating iliac vascular injuries: Recent 
experience with 233 consenting patients. J 
Trauma 1990;30:1450. 



Carillo EH, WohPtmann CD, Spain DA, et al: 
Common and external iliac artery injuries asso- 
ciated with pelvic fractures. J Orthop Trauma 
1999;13:351-355. 

Carillo EH, Spain DA, Wilson MA, et al: Alterna- 
tives in the management of penetrating injuries 
to the iliac vessels. J Trauma 1998;44:1024-1030. 

DeBakey ME, Simeone FA: Battle injuries of the 
arteries in World War II: An analysis of 2,471 cases. 
Ann Surg 1946;123:534-579. 

Degiannis E, Velmahos G, Levy R, et al: Penetrat- 
ing injuries of the iliac arteries: A South Africa 
experience. Surgery 1996;119:146-150. 

Feliciano DV, Mattox KL, Graham JM, Bitondo 
CA: Five-year experience with PTFE grafts in 
vascular wounds. J Trauma 1985;25:75. 

Haan J, Rodriguez A, Chiu W: Operative manage- 
ment and outcome of iliac vessel injury: A ten- 
year experience. Am Surg 2003;69(7):581-586. 

Hughes CW: Arterial repair during the Korean War. 
Ann Surg 1958;147:555-561. 

Rich NM, Baugh JH, Hughes CW. Acute arterial 
injuries in Vietnam: 1,000 cases. J Trauma, 
1970:359-369. 

Mattox KL: Penetrating injuries to the iliac 
arteries. Am J Surg 1978;135:663. 

Mattox KL, Feliciano DV, Burch J, et al: Five 
thousand seven hundred and sixty cardio- 
vascular injuries in 4,459 patients. Epidemiologic 
evolution. Ann Surg 1989;209(6):698-707. 

Rogers FB, Cipolle MD, Velmahos G, et al: Prac- 
tice management guidelines for the prevention 
of venous thromboembolism in trauma patients: 
The EAST practice management guidelines 
work group. J Trauma 2002;53(1):142-164. 

Velmahos GC, Theodorou D, Demetriades D, 
et al: Complications and nonclosure rates of 
fasciotomy for trauma and related risk factors. 
World J Surg 1997;21:247-253. 



chl9.qxd 4/16/04 3:33PM Page 353 





Extremity Vascular Trauma 



MICHAEL J. SISE 
STEVEN R. SHACKFORD 



OVERVIEW OF EXTREMITY VASCULAR TRAUMA 

Clinical Presentation 

Diagnosis 

Nonoperative Management 

Operative Management 

SUBCLAVIAN ARTERY INJURIES 

Surgical Anatomy 

Epidemiology and Etiology 

Clinical Features and Diagnosis 

Surgical Treatment 

Results 

Management of Scapulothoracic Dissociation 

AXILLARY ARTERY INJURIES 

Surgical Anatomy 

Epidemiology and Etiology 

Clinical Features and Diagnosis 

Surgical Treatment 

Results 

BRACHIAL ARTERY INJURIES 

Surgical Anatomy 

Epidemiology and Etiology 

Clinical Features and Diagnosis 



353 



chl9.qxd 4/16/04 3:33PM Page 354 



354 IV • SPECIFIC VASCULAR INJURIES 



Surgical Treatment 
Results 
O RADIAL AND ULNAR ARTERY INJURY 
Surgical Anatomy 
Epidemiology and Etiology 
Clinical Features and Diagnosis 
Surgical Treatment 
Results 
O VENOUS INJURIES OF THE UPPER EXTREMITY 
O COMPARTMENT SYNDROME OF THE UPPER EXTREMITY 
O POST-TRAUMATIC CAUSALGIA 
O LOWER EXTREMITY VASCULAR INJURIES 

Common Femoral and Profunda Femoral Arteries 

Surgical anatomy 

Epidemiology and etiology 

Clinical features and diagnosis 

Surgical treatment 

Results 
Superficial Femoral Artery 

Surgical anatomy 

Epidemiology and etiology 

Clinical features and diagnosis 

Surgical treatment 

Results 
Popliteal and Tibial Arteries 

Surgical anatomy 

Epidemiology and etiology 

Clinical features and diagnosis 

Surgical treatment 

Results 
Lower Extremity Compartment Syndrome 
Mangled Lower Extremity 
Venous Injuries of the Lower Extremity 

Surgical anatomy 

Epidemiology and etiology 

Clinical features and diagnosis 

Surgical treatment 

Results 



chl9.qxd 4/16/04 3:33PM Page 355 



19 • EXTREMITY VASCULAR TRAUMA 



355 



OVERVIEW OF EXTREMITY 
VASCULAR TRAUMA 



Vascular trauma of the extremities is a highly 
morbid injury that is becoming more 
common. Improved prehospital management 
and regionalization of trauma care with rapid 
transport have increased the number of these 
injuries seen at trauma centers in the last 3 
decades. Patients who previously died in the 
field or in transit because of severe isolated 
peripheral vascular injuries or multiple 
injuries with associated vascular trauma are 
now presenting alive. 

Successful management of extremity 
vascular trauma is based on early diagnosis 
and prompt treatment. The severity of 
injury and the length of time until restora- 
tion of perfusion are the major determi- 
nants of outcome. The management strategy 
must focus on minimizing the duration of 
ischemia to maximize the chance of success- 
ful recovery and rehabilitation. The often 
insidious nature of extremity vascular trauma 
significantly increases the opportunity for 
errors in management. Clinically relevant 
and practical protocols for both diagnosis 
and treatment are the best tools for avoiding 
these errors and ensuring the best possible 
outcome. 



Clinical Presentation 

Extremity vascular trauma may be immediately 
apparent on presentation because of exter- 
nal hemorrhage, hematoma, or obvious limb 
ischemia. A history of penetrating trauma asso- 
ciated with hypotension, pulsatile bleeding, 
or a large quantity of blood at the scene sug- 
gests potential vascular injury. Blunt trauma 
is also capable of causing significant vascular 
injury that can be overlooked when serious 
head, chest, or abdominal injuries are present 
(Fig. 19-1). 

Peripheral neurologic deficit should alert 
the examining physician to a possible vascu- 
lar injury. The deficit may be due to direct 
injury of a nerve in close anatomic proximity 
to an artery, or it may be the result of advanced 
ischemia. 




■ FIGURE 19-1 

Brachial artery occlusion secondary to distal 
shaft of humerus fracture in patient with 
multiple injuries. Shown are (arrows,) collateral 
flow filling distal brachial artery. Absent pulse 
and low forearm systolic pressure prompted 
arteriography. A saphenous vein interposition 
graft was required to repair this lacerated and 
contused artery. ■ 



Diagnosis 

A thorough history and careful physical exam- 
ination of the extremities for signs of vascu- 
lar injury are the first and most important steps 
in making the diagnosis of extremity vascular 
trauma. Careful inspection of the injured sites, 
examination of wounds, sensory and motor 
assessment, and pulse examination must be 
part of the extremity physical examination. 
The presence of a hematoma, bruit, or thrill 
must be noted. If distal pulses are diminished 
or absent, ankle or wrist systolic blood pres- 
sure should be determined with a continu- 
ous-wave Doppler device and compared with 
the normal side. A significant difference in 
systolic blood pressure (>10mmHg) between 
extremities may be an indication of vascular 
injury. Duplex scanning of the extremities has 
no role in the acute evaluation of extremity 
vascular trauma. 

Patients with "hard signs" of vascular injury 
(Table 19-1) should be taken directly to the 
operating room . In less straightforward cases, 
arteriography may be indicated to rule out 
the need for operation. Arteriography is 
limited to suspected extremity vascular trauma 
when no clear indication for immediate 
operative therapy is present or when evidence 
of peripheral ischemia and multiple sites of 



chl9.qxd 4/16/04 3:33PM Page 356 



356 



IV • SPECIFIC VASCULAR INJURIES 



TABLE 19-1 

"HARD" AND "SOFT" 
VASCULAR INJURY 



SIGNS OF 



Hard Indicate need for operative intervention 

Pulsatile bleeding 
Expanding hematoma 
Palpable thrill 
Audible bruit 
Evidence of regional ischemia 

Pallor 

Paresthesia 

Paralysis 

Pain 

Pulselessness 

Poikilothermia 

Soft Suggest need for further evaluation 

History of moderate hemorrhage 
Injury (fracture, dislocation, or 

penetrating wound) 
Diminished but palpable pulse 
Peripheral nerve deficit 



injury in an extremity exist (e.g., a shotgun 
injury with multiple pellet wounds) (Fig. 
19-2) .Arteriography is both sensitive and spe- 
cific in the diagnosis of extremity vascular 
injuries. However, arteriography is time con- 
suming, and successful management of these 
injuries requires prompt control of hemor- 
rhage and a timely restoration of adequate 
blood flow. 

Spiral computed tomographic angiography 
with the latest generation scanners might 
prove an acceptable alternative to formal 



arteriography. Although this imaging tech- 
nique requires contrast infusion, it does not 
require arterial catheterization, is easily per- 
formed, and is extremely rapid. Its use in the 
diagnosis of peripheral vascular injury has not 
yet been systematically evaluated. 



Nonoperative Management 

The widespread application of arteriography 
in the evaluation of injured extremities results 
in the detection of clinically insignificant 
lesions. Intimal irregularity, focal spasm with 
minimal narrowing, and small pseudo- 
aneurysms are often asymptomatic and do not 
progress. Considerable evidence suggests that 
nonoperative therapy of many asymptomatic 
lesions is safe and effective. However, suc- 
cessful nonoperative therapy requires con- 
tinuous surveillance for subsequent occlusion 
or hemorrhage. Operative therapy is required 
for thrombosis, symptoms of chronic ischemia, 
and failure of small pseudoaneurysms to 
resolve. 

A limited role exists for interventional 
radiologic techniques in the management of 
extremity vascular injuries. This modality 
requires special training, expertise, and an 
established interventional radiology program . 
Only an experienced interventionalist can suc- 
cessfully manage an extremity vascular injury. 
Amultidisciplinary approach in anticipation 
of injuries amenable to endovascular therapy 
is best led by a trauma surgeon skilled in the 




■ FIGURE 19-2 

Close-range shotgun injury to right medial knee. Pedal pulses were palpable but diminished. 
Formal arteriograpy demonstrated patent popliteal artery and peroneal and posterior tibial arteries. 
This patient was successfully treated nonoperatively. ■ 



chl9.qxd 4/16/04 3:33PM Page 357 



19 • EXTREMITY VASCULAR TRAUMA 



357 



management of extremity vascular trauma. 
The indications for endovascular techniques 
in the extremities are limited to hemorrhage 
from branch vessels that may be occluded 
without producing ischemia, acute pseudo- 
aneurysms with a small lateral wall arterial 
injury, intimal flap without significant under- 
lying thrombosis, and acute arteriovenous 
fistulas. Endovascular techniques are not 
effective in acute arterial occlusion from 
trauma. Endovascular placement of stents and 
stent grafts for noniatrogenic vascular trauma 
remains experimental and should be per- 
formed only in the most carefully selected 
cases (Fig. 19-3). Long-term results are not 
yet available and their application remains 
limited to specialized centers. 



Operative Management 

The operative management of extremity vas- 
cular injuries must be carefully orchestrated 



with the overall care of the patient. Intra- 
venous broad-spectrum antibiotics should be 
administered preoperatively. Systemic heparin 
may be given preoperatively to patients with 
isolated extremity injury (e.g., in whom 
cavitary hemorrhage has been excluded) to 
prevent propagation of thrombus. However, 
heparin should be avoided in multi-injured 
patients, especially those with central nervous 
system trauma. 

A generous sterile field should be prepared 
to allow for adequate exposure of vessels, 
to obtain proximal and distal control. This 
includes the chest and abdomen in proximal 
injuries of the upper and lower extremities. 
An uninjured leg should be prepared for har- 
vesting of autologous venous conduit. 

An orthopedic surgeon has an essential role 
in the surgical management of extremity vas- 
cular trauma associated with skeletal injury 
and should be involved before the surgical pro- 
cedure begins. Restoration of blood flow is 
imperative and can be achieved by an initial 





■ FIGURE 19-3 

A, Axillary artery laceration with 
pseudoaneurysm (arrow) in a 36-year-old 
man stabbed in the left upper chest. 

B, Endovascular treatment was 
accomplished with a covered stent seen in 
place with exclusion of pseudoaneurysm 
(arrows). C, Stent position in the axillary 
artery (arrows). At 9 months follow-up, the 
stent graft was patent and the patient had 
resumed normal work activity. ■ 



chl9.qxd 4/16/04 3:33PM Page 358 



358 



IV • SPECIFIC VASCULAR INJURIES 



vascular repair or insertion of a vascular shunt. 
The vascular surgeon's role does not end after 
perfusion is restored. Careful surveillance 
must be maintained to ensure that orthope- 
dic appliances do not obstruct the shunt or 
disrupt the arterial repair. The early involve- 
ment of a plastic and reconstructive surgeon 
is essential for the successful management 
of vascular injuries associated with large soft 
tissue defects. 

The appropriate treatment of extremity 
arterial and venous injuries consists of debride- 
ment of the damaged vessel, a tension-free 
repair, use of saphenous interposition graft- 
ing when primary repair is not possible, and 
adequate coverage with healthy vascularized 
tissue. 

Alimited role for primary amputation exists 
in the management of complex extremity vas- 
cular injuries. Patients with extensive soft tissue 
loss, neurologic deficit, extensive fractures, 
and vascular injuries should be evaluated 
collaboratively with orthopedic and plastic 
surgery colleagues to determine whether 
primary amputation is the best initial man- 
agement. These mangled extremities can be 
objectively evaluated using a rating system that 
accounts for the age of the patient, the type 



of injury and the severity of the injury (Fig. 
19-4; see also Table 19-1). However, the use 
of this scoring system is for general assessment 
and should never be a substitute for thought- 
ful clinical judgment using the skills of ortho- 
pedic and plastic surgery consultants. 

Fasciotomy, particularly in the setting of 
prolonged ischemia, remains an important 
adjunct in the management of extremity vas- 
cular injury. Elevated compartment pressure 
is a sufficient indication to proceed with fas- 
ciotomy, even before arterial repair. If normal 
pressures are obtained, eventual reperfusion 
edema and subsequent swelling may occur 
with delayed compartment syndrome. Thus, 
continuous or intermittent compartment 
pressure monitoring may be necessary in the 
postoperative period. Lack of a timely fas- 
ciotomy remains the most common error 
leading to preventable limb loss following 
vascular trauma. 

Frequent postoperative physical examina- 
tions of the extremity with vascular repair 
are essential. Any deterioration in the exam- 
ination must be investigated. Loss of a pal- 
pable pulse is an absolute indication for 
re-exploration. Early and prompt return to 
the operating room when thrombosis is 




■ FIGURE 19-4 

Extensive tissue destruction of the lower extremity in a man struck and rolled over by a bus. A, Right 
leg neurovascular disruption was complete and there were fractures at multiple levels. B, The left 
leg was similarly fractured but neurovascular function was normal. The right leg was amputated 
right below the knee and external fixation device was placed immediately. Recovery was rapid and 
functional outcome was acceptable. ■ 



chl9.qxd 4/16/04 3:33PM Page 359 



19 • EXTREMITY VASCULAR TRAUMA 



359 



suspected is the best way to ensure successful 



limb salvage. 



SUBCLAVIAN ARTERY INJURIES 



The initial evaluation and management of 
peripheral vascular trauma is summarized in 
Table 19-2. 



TABLE 19-2 

SUMMARY OF THE MANAGEMENT OF 
PERIPHERAL VASCULAR TRAUMA 

1. Perform thorough clinical evaluation; formal 
arteriography (uncommon). 

2. Administer preoperative broad-spectrum 
antibiotics. 

3. Consider systemic heparinization for an isolated 
vascular injury without any possibility of cavitary 
hemorrhage. 

4. Prepare and drape to allow harvesting of 
autologous conduit. 

5. Achieve proximal and distal control before direct 
investigation of the injury. 

6. Perform proximal and distal catheter 
thrombectomy; proximal and distal infusion of 
heparin. 

7. Achieve complete debridement of damaged 
vessel. 

8. Cover vascular anastomoses with viable tissue. 

9. Consider fasciotomy for elevated compartment 
pressures or prolonged ischemia. 

10. Monitor frequently during the postoperative 
period. 



Surgical Anatomy 

The right subclavian artery originates from the 
innominate artery and passes through the base 
of the neck behind the sternoclavicular joint. 
The left arises from the aortic arch and follows 
a similar course (Fig. 19-5). Anomalies of 
the subclavian arteries are rare. The most 
common anomaly is a right subclavian origi- 
nating from the descending aorta as the 
most distal branch of the aortic arch and 
passing posterior to the esophagus. This is 
thought to occur in approximately 1% of the 
population. 

The subclavian artery has three parts based 
on its relationship to the anterior scalene 
muscle (Fig. 19-6); first or proximal (proxi- 
mal to the muscle), second or middle (pos- 
terior to the muscle) , and third or distal (from 
the lateral border of the muscle to the lateral 
border of the first rib) . 

The first or proximal part gives off three 
branches (vertebral, internal mammary, and 
thyrocervical trunk) close to its termination 
near the anterior scalene muscle. The proxi- 
mal part of the first portion is free of branches 
for 1 to 3 cm. Several important structures are 



Inf. thyroid a 
Trans, cervical a 
Suprascapular a. 

Dorsal 
scapular a 



Thyrocervic 
Int. thorac 
Vertebral 
Right common carot 

Brachiocephalii 
(innominate a. 



Scalenus medius m. 
Scalenus anticus m. 




Costocervical trunk 
Left subclavian a. 



Left common carotid a. 



■ FIGURE 19-5 

Anterior view of subclavian artery with branches of the subclavian artery arising from the right 
side. ■ 



chl9.qxd 4/16/04 3:33PM Page 360 



360 



IV • SPECIFIC VASCULAR INJURIES 



■ FIGURE 19-6 

The right lateral view of the 
subclavian artery illustrating the 
branches of the subclavian 
artery with potential collateral 
anastomoses. ■ 



Scalenus 
anticus m 

Scalenus 
medius m 



Costocervical 
trunk 



Dorsal 
scapular a 




Vertebral a. 



Thyrocervical 
trunk 

Int. thoracic a. 



related to the first portion: The phrenic and 
vagus nerves cross anteriorly, the internal 
jugular-subclavian vein confluence passes 
anteriorly, and the cervical dome of the 
pleura is located inferiorly. On both sides, the 
venous confluence will contain the termina- 
tion of lymphatic channels, which are often 
multiple. On the left, the thoracic duct is easily 
injured during retraction. 

The second portion usually contains one 
or two branches (costocervical trunk and 
dorsal scapular) and is related closely to the 
brachial plexus. The third portion contains 
no branches and is closely related to the 
plexus. 

The branches of the subclavian artery 
provide such a rich collateral network that 
interruption of flow at any of the three parts 
rarely produces limb-threatening ischemia 
(Fig. 19-7). However, adjacent soft tissue 
destruction can disrupt these collaterals and 
threaten limb viability in the presence of sub- 
clavian artery thrombosis. 



Epidemiology and Etiology 

Subclavian artery injuries are uncommon 
and represent fewer than 5% of all arterial 
injuries noted in most civilian and military 
series (Rich and Spencer, 1978). This is 



because the subclavian artery is relatively short, 
is well protected by the sternum, clavicle, and 
first rib, and when partially lacerated, can 
produce rapid exsanguination and death in 
the field before patients receive medical 
attention. 

A blunt mechanism of injury can produce 
several types of subclavian artery injury: 
avulsion of branches (producing significant 
hemorrhage); contusion with intimal dis- 
ruption and prolapse (producing thrombo- 
sis); puncture or laceration from shards of 
bone from either the clavicle or first rib (pro- 
ducing hemorrhage); or severe stretching 
producing complete separation of intima 
and media with adventitia intact or disrupted 
(producing hemorrhage, thrombosis, or 
pseudoaneurysm). Recent civilian series 
document a blunt mechanism of injury as high 
as 45%. 



Clinical Features and Diagnosis 

Approximately 50% of patients with subcla- 
vian artery injuries present to the hospital in 
shock. The classic signs of advanced ischemia 
(pulselessness, pallor, paresthesias, poikilo- 
thermia, and paralysis) may be present but 
are not common because of the substantial 
collateral circulation available. Associated 



chl9.qxd 4/16/04 3:33PM Page 361 



19 • EXTREMITY VASCULAR TRAUMA 361 



Transverse cervical 



Descending branch 
of transverse cervical 

Transverse scapular 



Thoracoacromial 




■4 Subclavian 



Scapular 
circumflex 

Lateral thoracic 



Subscapular 

L Ascending branch of 
profunda brachii 
"-Brachial 



mammary 



■ FIGURE 19-7 

Collateral circulation in the shoulder 
region. Important collateral vessels 
are the thoracoacromial, lateral 
thoracic, subscapular, and anterior 
and posterior humeral circumflex 
arteries. (From Levin PM, Rich NM, 
Hutton JE Jr: Collateral circulation in 
arterial injuries. Arch Surg 
1971;102:392-399.) ■ 



injuries to the chest wall, lung, and brachial 
plexus are common. 

The diagnosis may be obvious when a 
patient presents with a penetrating wound at 
the base of the neck or in the supraclavicular 
fossa combined with loss of the ipsilateral 
pulse. If these patients are in shock that is unre- 
sponsive to volume resuscitation, they must 
be taken to the operating room for control of 
hemorrhage. However, if patients are stable 
or stabilize with resuscitation, arteriography 
is extremely useful to plan the operative 
approach. The arteriogram can elucidate the 
portion of the artery that has been injured 
and allow the surgeon to determine the safest 
exposure for obtaining proximal control. A 
chest roentgenogram is also useful. In pene- 
trating injuries, the entrance and exit wounds 
should be marked with radiopaque material 
before obtaining an x-ray film. In blunt 
injuries, the chestx-ray film can provide infor- 
mation about the mediastinum and unsus- 
pected fractures of the ribs or clavicle. 



Surgical Treatment 

Surgical exposure of a subclavian artery injury 
can be quite difficult because the clavicle and 



sternum obstruct a direct route to the artery. 
In addition, the area of the subclavian con- 
tains many important anatomic structures 
that can be injured in the haste to obtain 
control. Surgical management of the injury 
is difficult because the subclavian is not a 
muscular or thick-walled artery and is intol- 
erant of heavy-handed traction or imprecise 
suturing. 

The second and third portions of the 
subclavian artery can be exposed through a 
supraclavicular incision. On the right, the first 
portion is best exposed through a median ster- 
notomy. On the left, the first portion can be 
exposed through either a median sternotomy 
or a left anterolateral thoracotomy. For the 
first portion on the left, we recommend the 
thoracotomy approach because it is much 
easier. 

The location of the injury, the condition of 
the wound, and the condition of the patient 
determine the best approach. For patients in 
shock or those with a massive hematoma of 
the neck or chest wall, proximal control of 
the first portion of the subclavian is the safest 
approach (even if the injury is in the third 
portion) . Thoracotomy or sternotomy in an 
uninjured field allows rapid proximal control 
and can be lifesaving. For patients with an 



chl9.qxd 4/16/04 3:33PM Page 362 



362 



IV • SPECIFIC VASCULAR INJURIES 



injury to the third portion who are stable, the 
artery can be approached through a supra- 
clavicular incision. If uncontrollable bleeding 
is encountered, the wound should be packed 
and control should be obtained as discussed 
earlier. If it appears that the bleeding or the 
wound is directly behind the clavicle, the clav- 
icle can be resected (completely or in part) 
without significant long-term morbidity. For 
all of these reasons, we recommend prepar- 
ing and draping a wide field to include the 
neck to the mastoid process superiorly, along 
the trapezius to the deltoid, the entire ipsi- 
lateral arm (extended on a board) , and the 
entire chest. The arm should be supported 
on a board but be mobile. The draping should 
allow space for one operator to be positioned 
cephalad to the arm support and one opera- 
tor to be positioned caudad to the arm 
support. 

The operation follows general guidelines 
specified previously and elsewhere (Shackford 
and Rich, 2001). Following debridement, 
catheter thrombectomy, and regional 
heparinization, the subclavian should be 
carefully inspected. Mobilization of the ends 
to attempt an end-to-end anastomosis is rea- 
sonable if no major branches are divided to 
achieve the mobilization. We recommend 
interposition grafting rather than attempting 
an end-to-end anastomosis with any tension. 
If tension exists, the anastomosis may tear 



because the artery, by nature, is thin and 
nonmuscular. Although experience with pros- 
thetic material for interposition grafting of the 
subclavian is extensive, the first choice for a 
conduit should be autologous proximal saphe- 
nous vein. We have found the size match 
of the proximal to be reasonable for short 
segment bypasses in both males and females. 
Prosthetic material is certainly acceptable 
when either no available suitable vein exists 
or the patient's condition is such that pro- 
longation of the operation to harvest a conduit 
may jeopardize outcome. 

Recent series have documented that most 
injuries are treated with primary repair (42% ) , 
followed by interposition grafting with autol- 
ogousvein (Table 19-3) .Another alternative, 
when the patient is unstable, is ligation. As 
previously described, the collateral circulation 
around the shoulder and neck is extensive and 
ligation of the subclavian artery is rarely asso- 
ciated with limb loss. 



Results 

Many reports document experience with sub- 
clavian artery injuries, but only a precious 
few document immediate, short-term, or 
long-term outcome. When outcome is 
reported, it is usually immediate or short term 
and focuses either on survival, on the patency 



TABLE 19-3 

SURGICAL TECHNIQUES USED FOR MANAGEMENT OF UPPER EXTREMITY 
VASCULAR INJURY: SELECTED REVIEW OF THE RECENT LITERATURE* 



Artery 



Series* 



Years* 



None 

(%)" 



Primary 
(%)' 



ASV 

(%r 



Prosthetic 



Ligate 

(%)** 



Subclavian 


6 


1988-2000 


378 


32 (8.5) 


160(42.3) 


128(33.8) 


47 (12.4) 


11(3) 


Axillary 


7 


1982-1998 


126 





59 (46.8) 


42 (33.3) 


20(15.8) 


5(4.1) 


Brachial 


5 


1984-1994 


223 


5(2.2) 


121 (54.3) 


87 (39) 





3(4.5) 


Radial/ulnar 


5 


1984-1994 


251 


3(1.1) 


161 (64.2) 


30(11.9) 





57(22.8) 



'References available on request. 

+ Number of published articles reviewed. 

*Years covered by the aggregated publications. 

§ l\lumber of patients. 

"No repair or exploration undertaken. 

"Either vein patch, end to end anastomosis or arteriography. 

"Saphenous vein interposition. 

^Prosthetic graft interposition. 

**Ligation, no repair. 



chl9.qxd 4/16/04 3:33PM Page 363 



19 • EXTREMITY VASCULAR TRAUMA 



363 



of the repair, or on limb salvage. Unfortu- 
nately, these outcome measures lack relevance 
because survival is rarely dependent solely on 
repair of the subclavian artery injury and 
thrombosis of the arterial repair rarely results 
in amputation of the upper extremity because 
of its abundant collateral circulation. Rather, 
long-term outcome is determined primarily 
by the neurologic function and secondarily 
by the orthopedic outcome (Hardin and col- 
leagues, 1985). 

Mortality rates are highly variable after 
subclavian artery injury because when death 
occurs, it is usually because of associated 
injuries. However, rare cases of death result- 
ing from uncontrolled hemorrhage from 
subclavian arterial lacerations that either go 
unnoticed or undergo attempted repair 
without proximal control have been reported. 

Amputation is rare following subclavian 
injuries and is usually a result of devastating 
soft tissue loss, multiple arterial injuries, infec- 
tion (primarily intractable osteomyelitis), or 
severe neurologic injury with "flail arm" (see 
"Scapulothoracic Dissociation," later in this 
chapter) . Graft infection (either of prosthetic 
material or vein) is uncommon and can be 
treated by ligation and extra-anatomic bypass 
(either carotid-subclavian or axilloaxillary) 
if either claudication or limb-threatening 
ischemia develops. 

Complete resolution of infection, swelling, 
pain, and neurologic deficit and complete 
healing of all wounds define a good outcome 



following upper extremity vascular injury 
(Hardin and colleagues, 1985). When pa- 
tients with scapulothoracic dissociation are 
included, the long-term outcome following 
subclavian artery injury is dismal, and only 
30% of patients have a good outcome (Table 
19-4) . Excluding those with scapulothoracic 
dissociation improves the good outcome 
to about 40%, with the balance of patients 
having persistent disability due to nerve 
injury, osteomyelitis, or causalgia. 

Management of 
Scapulothoracic Dissociation 

Scapulothoracic dissociation is a devastating 
injury of the upper extremity and shoulder 
girdle caused by blunt injury. The mechanism 
is stretch and avulsion of the vascular and neu- 
rologic elements of the arm from their more 
proximal origins in the shoulder and neck 
regions. Substantial separation and/or frac- 
ture of the musculoskeletal attachments of the 
shoulder girdle can occur. Scapulothoracic dis- 
sociation is a rare injury, with only 52 patients 
reported in the literature (Sampson and 
colleagues, 1993). On physical examination, 
there is absence of the radial pulse associated 
with a significant shoulder or chest wall 
hematoma and absence of sensory or motor 
function below the shoulder. Chest radiogra- 
phy will demonstrate a laterally displaced 
scapula (with acromioclavicular disruption 



TABLE 19-4 

OUTCOME FOLLOWING REPAIR OF UPPER EXTREMITY VASCULAR INJURY: 
SELECTED REVIEW OF THE RECENT LITERATURE* 



Artery 



Series 1 



Years* 



N § 



Die (%)" 



AMP (%)' ABNML (%)* 



NML (%)* 



Subclavian 


6 


1984-1993 


103 


17 (16.5) 


6 (5.8) 


45 (43.6) 


35(34.1) 


Axillary 


4 


1982-1990 


92 


2(2.2) 


1(1.1) 


66(71.7) 


23 (25) 


Braohia 


4 


1984-1994 


146 


1 (0.6) 


5(3.4) 


42 (28.7) 


98 (67.3) 


Radial/ulnar 


4 


1984-1994 


211 


1 (0.5) 


7 (3.5) 


74 (35) 


129(61) 



'References available on request. 

+ Number of published articles reviewed. 

*Years covered by the aggregated publications. 

§ Number of patients. 

"Death. 

"Amputation. 

"Abnormal function (see text) does not include amputations. 

^Normal function (see text). 



chl9.qxd 4/16/04 3:33PM Page 364 



364 



IV • SPECIFIC VASCULAR INJURIES 



and increased distance between the distal 
end of the clavicle and the acromion). An 
associated displaced clavicle fracture or a 
sternoclavicular disruption is often present 
(Sampson and colleagues, 1993). Unfortu- 
nately, the outcome is uniformly poor because 
of the neurologic disruption, not the arterial 
injury. Our experience and that of Sampson 
and colleagues (1993) suggest that delayed 
hemorrhage or limb-threatening ischemia 
is very rare and there are no benefits to 
revascularization. In the rare patient who is 
actively bleeding, we recommend ligation. In 
the infrequent patient with limb-threatening 
ischemia, primary amputation should be 
considered. 



AXILLARY ARTERY INJURIES 



Surgical Anatomy 

The axillary artery begins at the lateral margin 
of the first rib and ends at the lateral margin 



of the teres major muscle. Three parts are 
dependent on their relationship to the pec- 
toralis minor muscle (Fig. 19-8) : proximal to 
the muscle (first) and beneath (second) and 
distal to the muscle (third) . The first part has 
one branch (superior thoracic), the second 
has two (thoracoacromial, lateral thoracic), 
and the third has three (anterior and poste- 
rior circumflex, subscapular) . These branches 
provide a rich collateral circulation to this 
region. 

The axillary vein lies anterior and slightly 
inferior to the axillary artery. Close proxim- 
ity to the artery provides the anatomic basis 
for the development of an arteriovenous 
fistula following relatively minor trauma (e.g., 
arterial cannulation) . A similar close rela- 
tionship exists to branches of the brachial 
plexus (Fig. 19-9). Proximally, the plexus is 
posterior/lateral to the artery. Distally, the 
three cords of the plexus surround the second 
and third parts of the artery. This intimate 
relationship explains the high incidence of 
concomitant nerve injuries in axillary arter- 
ial trauma. 



Superior thoracic a. 




Ant. circumflex 
humeral a 

Post, circumflex 
humeral a. 



Subscapular a 



Thoracoacromial a. 



Lat. thoracic a. 



■ FIGURE 19-8 

Surgical anatomy of the axillary artery with the usual configuration of six branches coming from the 
three parts of the artery. ■ 



chl9.qxd 4/16/04 3:33PM Page 365 



19 • EXTREMITY VASCULAR TRAUMA 



365 



AXILLARY ARTERIAL INJURIES 

Upper trunk 
Middle trun 
Lower trunk 
Medial cord 
Post, cord 




Musculocutaneous n 

/ 

Circumflex n 

Radial n 

"Median n. 
^Ulnar n. 
"Axillary v. 

■ FIGURE 19-9 

The close proximity of the brachial plexus to the cord adjacent to the second part of the axillary 
artery and the branches surrounding the third part demonstrate why there is a high incidence of 
concomitant nerve injuries with axillary arterial trauma, as shown above. ■ 



Epidemiology and Etiology 

Axillary artery injuries are only slightly more 
common than subclavian artery injuries and 
represent 5% to 10% of all arterial injuries in 
most military and civilian injuries (Rich and 
Spencer, 1978). More than 95% of axillary 
artery injuries are from penetrating trauma. 
Included in this group are patients with iatro- 
genic injury following cannulation of the 
artery for arterial pressure monitoring or con- 
trast studies. Although blunt injuries are rare, 
two types merit consideration. The first is 
rupture, contusion, or stretching of the artery 
following fracture of the proximal humerus 
or anterior dislocation of the shoulder. 
Approximately 1 % of shoulder dislocations 
are associated with axillary artery injury 
(Sparks and colleagues, 2000) . The second is 
thrombosis following chronic repetitive 
impingement by crutch use. 



Clinical Features and Diagnosis 

Patients with axillary artery injury commonly 
present with regional signs and symptoms, 



such as a pulse deficit, advanced ischemia, pul- 
satile bleeding, or an expanding hematoma. 
Shock solely due to an axillary artery injury 
is rare. The most common associated injury 
is vascular (axillary vein) followed very closely 
by nerve (cords or branches of the brachial 
plexus) . 

The diagnosis of an axillary artery injury 
should be suspected in a patient with a 
penetrating wound of the axilla, a palpable 
subclavian pulse (detected by palpation in the 
supraclavicular fossa) , but no distal pulses. If 
evidence of advanced ischemia is present, the 
patient should be taken immediately to the 
operating room. 

Occasionally, a patient with a penetrating 
injury near the axilla will present with a pal- 
pable radial pulse and a thrill or bruit in the 
region of the injury. An arteriovenous fistula 
should be the primary consideration and 
formal arteriography should be performed. 
Formal arteriography can provide needed 
diagnostic information and, if performed 
by qualified and experienced angiographers, 
can afford a potential opportunity for 
endoluminal treatment (see Chapters 9 
and 10). 



chl9.qxd 4/16/04 3:33PM Page 366 



366 



IV • SPECIFIC VASCULAR INJURIES 



Surgical Treatment 

Anticipate proximal control before entering 
the site of the injury by preparing the site to 
include all of the shoulder, ipsilateral neck 
and supraclavicular fossa (to allow for expo- 
sure of the subclavian artery) , the arm and 
hand to the fingertips (to allow intraopera- 
tive palpation of the radial pulse) , and the 
contralateral leg (to allow a separate team to 
harvest) for a conduit (Fig. 19-10). The arm 
should be supported on a board but be mobile. 
The draping should allow space for one oper- 
ator to be positioned cephalad to the arm 




■ FIGURE 19-10 

Preparing for surgery. Note that the entire arm 
is being prepared as well as the ipsilateral neck 
and chest. We prefer to drape the contralateral 
proximal thigh for a conduit as this allows a 
second team to harvest the proximal 
saphenous vein, while the primary team obtains 
proximal control. The drapes should be placed 
in such a way as to allow an assistant to stand 
in the area cephalad to the shoulder and the 
arm. ■ 



support and one operator to be positioned 
caudad to the arm support. 

Exposure of the proximal axillary artery is 
best obtained by an infraclavicular incision, 
made approximately one fingerbreadth below 
and parallel to the clavicle. This proximal 
exposure is recommended for all cases of axil- 
lary artery trauma (Graham and colleagues, 
1982) because injuries that are more distal are 
often associated with significant hematoma 
(Fig. 19-11). Obtaining control of an axillary 




■ FIGURE 19-11 

A, Close range shotgun wound to the anterior 
axilla. Notice the large hematoma causing 
significant swelling of the anterior chest 
wall, axilla, and deltoid region of the arm. 

B, Intraoperative photo from same perspective 
(patient's head is located toward the right side 
of this picture) demonstrating the infraclavicular 
incision and exposure of the injury through a 
second incision. After proximal control was first 
obtained through the infraclavicular incision 
(note the vascular clamp in the wound), the 
hematoma was opened and evacuated, 
allowing better visualization of the injury and 
avoidance of iatrogenic trauma to the brachial 
plexus. ■ 



chl9.qxd 4/16/04 3:33PM Page 367 



19 • EXTREMITY VASCULAR TRAUMA 



367 



arterial injury in the midst of a large 
hematoma is fraught with problems that 
inevitably lead to increased blood loss and pos- 
sibly to an iatrogenic brachial plexus injury. 
The infraclavicular incision can be extended 
into the axilla and both the pectoralis minor 
and the major tendons can be divided, if nec- 
essary, to obtain distal control. 

Conduct of the operation follows general 
guidelines specified previously and elsewhere 
(Shackford and Rich, 2001). Clamps should 
never be blindly placed near the axillary artery 
because its intimate relationship to the axil- 
lary vein and to the brachial plexus mandate 
precise clamp placement. In the event that 
uncontrolled bleeding exists, tamponade can 
be obtained by gentle finger compression or, 
if the lumen can be visualized, insertion of a 
balloon tipped catheter followed by careful 
balloon expansion. 

Following debridement, catheter thrombec- 
tomy, and regional heparinization, the axil- 
lary artery should be carefully inspected. 
Mobilization of the ends to attempt an end- 
to-end anastomosis is reasonable if no major 
branches are divided to achieve the mobi- 
lization. Recent series have documented that 
most injuries are treated with primary repair 
(47%) followed by interposition grafting with 
autologous conduit (33%, see Table 19-3). 
We recommend interposition grafting with 
autologous proximal saphenous vein rather 
than attempting an end-to-end anastomosis 
with any tension. Ligation of the axillary artery 
is acceptable (there is a rich collateral circu- 
lation) in patients who are moribund and phys- 
iologically unstable, but this is not encouraged. 
Rather, if both ends of the artery can be 
visualized, a temporary intravascular shunt 
can be placed and the wound packed, towel 
clipped or stapled until the patient has stabi- 
lized. These shunts can be left in place several 
days without systemic anticoagulation 
(Granchi and colleagues, 2000). 



Results 

Similar to the literature describing treatment 
results of subclavian artery injuries, only a few 
reports document the outcome of axillary 
artery repair. When outcome is reported, it is 



usually immediate or short-term and focused 
on survival, the patency of the repair or limb 
salvage. Unfortunately, these outcome mea- 
sures lack relevance because survival is rarely 
dependent solely on repair of the axillary 
artery injury and thrombosis of the arterial 
repair rarely results in amputation of the upper 
extremity because of the abundant collateral 
circulation in the shoulder and arm. Rather, 
long-term outcome is determined primarily 
by the neurologic function and secondarily 
by the orthopedic outcome (Hardin and 
colleagues, 1985). 

Mortality and amputation are rare fol- 
lowing axillary artery injury (see Table 19-4) . 
A good outcome following axillary repair 
(as determined by complete resolution of 
swelling, pain and neurologic deficit) is rare 
because axillary artery injury is often accom- 
panied by nerve injury, which ultimately 
leads to long-term neuralgia or causalgia. In 
four recently published series of axillary 
artery injuries in which follow-up was docu- 
mented for 92 patients, only 23 (25%) had 
a good outcome (see Table 19-4) . The other 
66 patients had neurologic dysfunction, 
post-traumatic neuralgia producing dis- 
ability, or diminished use because of chronic 
pain associated with osseus or soft tissue 
injury. 



BRACHIAL ARTERY INJURIES 



Surgical Anatomy 

The brachial artery is a continuation of the 
axillary artery and begins at the lower border 
of the teres major muscle. Exiting the axilla, 
the brachial artery is a relatively superficial 
structure covered only by skin, subcutaneous 
tissue, and deep fascia. Proximally, it lies 
medial to the humerus and is accompanied 
by the median nerve (superiorly and laterally) 
and the ulnar and radial nerves (medially) . 
Distally, it lies anterior to the elbow and is 
crossed by the median nerve, which then lies 
medial to the artery. Just proximal to the 
elbow, the ulnar nerve is posterior to the artery 
as it goes behind the medial epicondyle of the 
ulna. The brachial artery terminates 1 inch 



chl9.qxd 4/16/04 3:33PM Page 368 



368 



IV • SPECIFIC VASCULAR INJURIES 



below the elbow skin crease where it divides 
into the radial and ulnar arteries. 

The brachial artery has three main branches 
(Fig. 19-12). The first (most proximal) is the 
profunda brachii, which accompanied by 
the radial nerve passes posteriorly between 
the medial and long head of the triceps 
muscle. The profunda brachii provides an 
important collateral anastomosis with the axil- 
lary artery through its posterior circumflex 
humeral branch. The profunda also has a col- 
lateral anastomosis with the radial recurrent 
artery. The second main branch of the brachial 
artery is the superior ulnar collateral, which 
accompanied by the ulnar nerve passes behind 
the medial epicondyle to provide a collateral 
anastomosis with the posterior ulnar 



recurrent. The third (most distal) main 
branch is the inferior ulnar collateral, which 
provides a rich anastomotic collateral network 
around the elbow with the ulnar artery 
through its anterior recurrent branch. 



Epidemiology and Etiology 

Brachial artery injury is the most commonly 
reported arterial injury of the upper extrem- 
ity. In large military and civilian series, brachial 
artery injury constitutes 15% to 30% of all 
peripheral arterial injuries. The reason for this 
relatively high frequency is that the brachial 
artery is relatively long, superficial, and 
exposed as compared to other peripheral 



Radial n 

Median n 

Ulnar n. 




■ FIGURE 19-12 

The brachial artery is a continuation of the axillary artery at the lower border of the teres major 
muscle. It terminates approximately 1 inch below the transverse skin crease in the antecubital 
fossa, where it divides into two branches. Important anatomic relationships include three associated 
nerves, three associated veins, and three main branches with the brachial artery lying successively 
on three muscles. ■ 



chl9.qxd 4/16/04 3:33PM Page 369 



19 • EXTREMITY VASCULAR TRAUMA 



369 



arteries. Furthermore, the upper extremity is 
often used as a lever, hammer, and weapon, 
as well as a protective or restraining device for 
the torso, all of which put the brachial artery 
in harm's way. 

Penetrating trauma is the most common 
cause of brachial artery injury. Recently, the 
increase in the number of diagnostic cardiac 
catheterizations has resulted in an increase 
in the number of brachial artery injuries 
seen at most tertiary medical centers. Blunt 
injury of the brachial artery is much less 
common but deserves emphasis because it can 
easily be overlooked unless there is a high 
index of suspicion. Supracondylar fracture 
of the humerus, particularly with anterior 
displacement or elbow dislocation (Endean 
and colleagues, 1992), should alert the 
clinician to the possibility of a brachial artery 
injury. 

Clinical Features and Diagnosis 

Patients with brachial artery injuries classically 
present with a cool, painful hand, no radial 
pulse, and diminished sensory and motor 
function of the forearm and hand. The classic 
findings, however, are not always present. 
Patients may have a complete thrombosis of 
the brachial artery and loss of a palpable radial 
pulse but have a warm hand without neuro- 
logic dysfunction. Conversely, the patient 
may have a laceration of the brachial artery 
and have a palpable radial pulse. If symptoms 
of ischemia associated with "hard" signs 
are present, the diagnosis is not certain. In 
patients with a supracondylar fracture or an 
elbow dislocation where doubt about the diag- 
nosis exists (diminished or absent pulse, but 
a warm, pink hand) , arteriography is indi- 
cated. In patients with closed blunt trauma 
and primarily neurologic signs and symptoms 
who have a warm pink hand and a palpable 
radial pulse, plethysmography and segmen- 
tal pressure determination can avoid a need- 
less arteriogram. 

Careful physical examination and com- 
prehensive documentation of the pulses and 
neurologic findings are essential, particularly 
in patients who are to undergo operative 
exploration. This point cannot be overem- 



phasized in patients with brachial artery 
injuries who have peripheral neurologic 
deficits before operation. Lack of documen- 
tation of the preoperative neurologic status 
leads to the assumption that the deficits arose 
out of some operative misadventure. 



Surgical Treatment 

Bleeding can be controlled by proximal com- 
pression against the humerus or by direct pres- 
sure over an open wound. Blindly attempting 
to clamp a bleeding vessel in the arm is never 
necessary and is fraught with the hazard of 
significant injury to the median, radial, or 
ulnar nerve. 

For suspected proximal injury, prepare and 
drape the patient similar to that used to 
manage an axillary artery injury (see previ- 
ous discussion). For injuries that are more 
distal, prepare the arm and hand to the fin- 
gertips (to allow intraoperative palpation of 
the radial pulse) and a leg (to allow a sepa- 
rate team to harvest) for a conduit. The arm 
should be supported on a board but be mobile. 
The draping should allow space for one oper- 
ator to be positioned cephalad to the arm 
support and one operator to be positioned 
caudad to the arm support. 

Exposure of the brachial artery is best 
obtained by a longitudinal incision in the pal- 
pable groove between the triceps and biceps 
muscle along the medial aspect of the arm. 
This incision can be extended distally across 
the antecubital fossa or proximally across 
the axilla with an 5-shaped curve (Fig. 19-13) . 
No matter where the exposure is obtained 
(proximally or distally), precise dissection 
and careful handing of all structures are 
mandatory. Careless dissection or heavy- 
handed retraction may result in injury to the 
associated nerve (particularly the median 
nerve) . 

Conduct of the operation follows general 
guidelines specified previously and elsewhere 
(Shackford and Rich, 2001). Following 
debridement, catheter thrombectomy, and 
regional heparinization, the brachial artery 
should be carefully inspected. Small lateral 
injuries, particularly those associated with 
iatrogenic injuries, may be reapproximated 



chl9.qxd 4/16/04 3:33PM Page 370 



370 



IV • SPECIFIC VASCULAR INJURIES 




Retracted biceps m 

L 



Cut bicipital 
aponeuros 



Cephalic v.- 
Radial a 




Cut median 
cubital v. 



F / 

I Li — Ulnar 



Median n. 



Retracted pronator 
teres m. 



■ FIGURE 19-13 

Surgical exposure of the brachial artery is rapidly obtained by a longitudinal incision along the 
course of the artery with an extension as an S curve either across the axilla proximally or across the 
antecubital fossa distally as needed. The median nerve and basilic veins are in close proximity to 
the artery. ■ 



with simple interrupted sutures placed in the 
same axis as the direction of the artery so no 
luminal compromise occurs. If more than two 
or three sutures are required, a vein patch is 
a better alternative because the artery is easily 
narrowed. Our preference for most injuries 
is excision of the area of injury and either an 
end-to-end anastomosis (performed with 
interrupted sutures and with the ends of the 
vessel distracted for placement of all sutures) 
or an interposition autologous saphenous vein 
graft. Recent civilian series support this 



approach, with the majority of the repairs 
being either primary end-to-end or interpo- 
sition vein grafting (see Table 19-3). If asso- 
ciated orthopedic injuries are present, our 
preference is to place an indwelling tempo- 
rary shunt in the artery and let the orthope- 
dic surgeons achieve length and stability of 
the arm before attempting definitive vascular 
repair. Ligation should never be a considera- 
tion because it carries a significant risk of 
amputation. If the patient is in extremis from 
other associated injuries, an indwelling 



chl9.qxd 4/16/04 3:33PM Page 371 



19 • EXTREMITY VASCULAR TRAUMA 



371 



temporary shunt can be placed quickly and 
left in place for several days without systemic 
heparinization. 



Results 

Amputation or death is rare following brachial 
artery repair. Long-term outcome following 
brachial artery repair is decidedly better than 
either subclavian or axillary artery injuries 
because the incidence of associated nerve 
injury is much less. In recent civilian series 
(see Table 19-4), a good outcome (as deter- 
mined by complete resolution of swelling, 
pain, and neurologic deficit) was achieved in 
almost 70% of patients. If patients develop 
symptoms of arm claudication on follow-up, 
they should undergo noninvasive vascular 
testing to include plethysmography, segmen- 
tal pressure determination, and duplex of the 
area of injury. If stenosis or occlusion is 
evident, the patients should have diagnostic 
arteriography for possible endoluminal or 
open revision. 



RADIAL AND ULNAR 
ARTERY INJURY 

Surgical Anatomy 

After the brachial artery crosses through the 
cubital fossa, it bifurcates into the radial and 
ulnar artery. The ulnar artery is the larger of 
the two, but this size discrepancy exists only 
in the proximal portion of the artery. Two 
branches immediately arise from the proxi- 
mal ulnar artery: the anterior and posterior 
ulnar recurrent arteries that form collateral 
anastomoses with the brachial artery around 
the anterior and posterior aspects of the elbow, 
respectively. The common interosseus also 
arises from the proximal ulnar artery (Fig. 
19-14) and passes laterally and posteriorly 
toward the interosseus membrane where, at 
the superior edge of the membrane, it divides 
into the volar (anterior) and dorsal (poste- 
rior) interosseus arteries. The dorsal 
interosseus gives rise in its proximal portion 
to the interosseus recurrent, which forms a 



collateral anastomosis with branches of the 
brachial artery. The ulnar artery terminates 
in the superficial or volar palmar arch. In its 
oblique proximal portion, it is crossed by the 
pronator teres and by the median nerve. The 
ulnar nerve joins the artery in its distal third 
(Fig. 19-15). 

The radial artery is unique in that no muscle 
or nerve crosses it in its relatively direct course 
to the wrist. The only major branch of the 
radial artery is the radial recurrent, which 
passes under the brachioradialis muscle to pass 
proximally and form a collateral anastomosis 
with branches of the profunda brachii. The 
radial artery gives a small branch to the super- 
ficial arch but terminates in the deep palmar 
arch. 



Epidemiology and Etiology 

Arterial injuries of the forearm are often 
reported in recent series describing vascular 
injuries and now make up between 5% and 
30% of the total peripheral vascular injuries. 
Approximately 95% are due to penetrating 
trauma. A relatively rare form of ulnar artery 
injury occurs in individuals with a history of 
repeatedly using their hypothenar eminence 
as a hammer. It is thought that the repeated 
trauma can produce aneurysmal dilatation, 
distal embolization, or thrombosis. 



Clinical Features and Diagnosis 

Complete interruption of either the radial or 
the ulnar artery will often have no adverse 
effect on the circulation of the forearm or 
hand because of the rich collateral circula- 
tion. Signs of advanced ischemia warrant 
surgical exploration or operating room 
arteriography. If doubt exists about the 
integrity of the circulation and the condition 
of the patient and the condition of the arm 
permit, formal arteriography with magnifi- 
cation views of the hand are helpful for 
diagnosis and the planning of operative 
management. 

Puncture wounds of the forearm can be 
quite insidious because the small skin wound 
will not allow sufficient egress of venous or 



chl9.qxd 4/16/04 3:33PM Page 372 



372 



IV • SPECIFIC VASCULAR INJURIES 



Brachial a. 
Radial collateral a. 



Mid. collateral a.— V 

Trans, branch inf. 
ulnar collateral a. 



Radial recurrent a 



Interosseous 
recurrent a. 



Radial a. 



Volar interosseous a 



Superf. volar 
branch radial a. 




Superior ulnar 
collateral a. 

Inf. ulnar collateral a. 



Ant. branch inf. 
ulnar collateral a. 

Post, ulnar recurrent a. 
Ant. ulnar recurrent a. 



Interosseous a. 



Ulnar a. 



Dorsal interosseous a. 



Superf. & deep 
branches ulnar a. 



■ FIGURE 19-14 

As the brachial artery divides into the radial artery (its more direct continuation) and the ulnar artery 
(the larger of the two branches) in the forearm, there are important collateral branches, which help 
form the rich anastomosis around the elbow. The common interosseus is also an important branch 
of the ulnar artery. ■ 



arterial blood, which can accumulate in 
significant quantity in the subcutaneous and 
subfascial planes to produce a forearm com- 
partment syndrome. Physical signs that should 
alert the examiner to the possibility of an 
expanding hematoma include marked tension 
in the dorsal or volar forearm, superficial 
venous engorgement, paresthesias in the 
hand, or diminished sensation to light touch 
in the fingers. Without fasciotomy, these 
patients are at risk of developing a Volkmann 
contracture. 



Surgical Treatment 



Control of hemorrhage from either the radial 
or the ulnar artery is easily achievable by direct 



pressure. Tourniquets or blind clamping in 
an open wound is not warranted. Prepare the 
arm and hand to the fingertips (to allow intra- 
operative palpation of the radial pulse) and 
a leg (to allow a separate team to harvest) for 
a conduit. The arm should be supported on 
a board but be mobile. The draping should 
allow space for one operator to be positioned 
cephalad to the arm support and one opera- 
tor to be positioned caudad to the arm 
support. 

Exposure of the proximal portions of both 
arteries can be accomplished through an 
5-shaped incision in the cubital fossa (Fig. 
19-16). The distal arteries can be exposed 
through longitudinal incisions over the course 
of the artery just proximal to the hand. 
Conduct of the operation follows general 



chl9.qxd 4/16/04 3:33PM Page 373 



19 • EXTREMITY VASCULAR TRAUMA 



373 




Brachioradialis m 
Radial n 
Brachial a 
Bicipital aponeurosis 

Boon radial n. fl|«lL ^ ^ ' ' 

Brachioradialis m.-HW ' !,' v/pi'Km 



Pronator teres m 

Superf. radia 

Radial a. 41 

Flexor pollicis Ml 
longus m 



ceps m. (medial 
termuscular septum) 
Median n. 
Brachialis m. 

Pronator teres m. 

Flexor carpi radialis m. 
Common interosseous a. 

Flexor digitorum 
sublimis m. 
Inar a. 



Abductor pollicis 
longus m. 

Median i 



IS* : • 

is , M> 'ii\l ' \-Flexor carpi ulnaris m 

"'Wltlli. \Z -P^UInarn. 

M '■ W 

' V lit 

■ I n It 



Flexor carpi radialis t 



r lexor digitorum 
Drofundus m. 

Dorsal cutaneous 
branch ulnar n. 
#-Flexor digitorum 
f superficialis tt. 

Palmaris longus t. 
Ulnar a. & n. 
Palmar carpal lig. 



■ FIGURE 19-15 

The relationship of the radial and ulnar arteries to the important nerves, major muscle groups, and 
tendons. Particularly note the crossing of the proximal ulnar artery by the median nerve and the 
close approximation of the ulnar nerve to the distal two thirds of the ulnar artery. The cross section 
through the upper third of the forearm emphasizes the relatively deep location of the ulnar artery 
compared with the more superficial radial artery. ■ 




guidelines specified previously and elsewhere 
(Shackford and Rich, 2001). Following 
debridement, catheter thrombectomy, and 
regional heparinization, the area of injury 
should be carefully inspected. If both arter- 
ies are injured, repair of the ulnar is less tech- 
nically taxing because of its relatively larger 
size. If only one artery is injured and no sign 
of ischemia is seen in the hand (as docu- 
mented by a comprehensive physical exami- 
nation and confirmed by Doppler signals in 
the palmar arch and digits), ligation is rea- 
sonable (Johnson, Ford andjohansen, 1993) . 
For small "clean" lacerations or puncture 
wounds, lateral suture repair may suffice. More 
severe lacerations will require resection and 
end-to-end anastomosis, which is the most 



common technique used in recent series (see 
Table 19-3) . Reversed autologous saphenous 
vein from the distal leg can be used if signif- 
icant arterial debridement is required or when 
an end-to-end anastomosis will create tension 
on the suture line. 



Results 

Amputation is rare following radial or ulnar 
artery injury. When amputation does occur, 
it is often the result of massive soft tissue 
destruction (with interruption of both the 
radial and the ulnar arteries) associated with 
sepsis or chronic osteomyelitis. Few studies 
document adequate long-term follow-up. 



chl9.qxd 4/16/04 3:33PM Page 374 



374 



IV • SPECIFIC VASCULAR INJURIES 



Ulnar a. 




Ulnar n. 



Flexor carpi ulnaris m. 



■ FIGURE 19-16 

Elective incisions that can be used for approach to the radial and ulnar arteries. A, An S-type 
incision starting along the course of the distal brachial artery, carried through the antecubital fossa 
and continued down on the forearm will give excellent exposure of the proximal ulnar and radial 
arteries, as well as the origin of the common interosseous artery (A). An extension off this incision 
(B) along the course of the radial artery can be used for exposure to the wrist level. A separate 
incision can be used over the course of the ulnar artery (C). B, This drawing demonstrates 
exposure of the ulnar neurovascular bundle within the deep muscle layers, which have been split 
proximally. ■ 



Recent series with adequate documentation 
of follow-up demonstrate good results (as 
determined by complete resolution of 
swelling, pain, and neurologic deficit) fol- 
lowing radial or ulnar repair in 65% of 
patients (see Table 19-4) . Poor results are not 
related to the vascular repair, but to the asso- 
ciated nerve or tendon injuries. In fact, in one 
study, patency of the vascular repair when 
only one artery (either radial or ulnar) was 
injured was 50% (Johnson, Ford, and 
Johansen, 1993) . Despite the high failure rate 
of radial or ulnar repairs, no patients had 
claudication. 



VENOUS INJURIES OF THE 
UPPER EXTREMITY 

Injuries to the subclavian or axillary vein 
should be repaired if the patient's condition 
permits. In most cases, repair will consist of 
lateral venorrhaphy or end-to-end anasto- 
mosis. After appropriate debridement, when 
a direct repair cannot be performed because 
it will result in tension on the suture line or 
will significantly narrow the vein, repair can 
be accomplished with autologous vein patch, 
interposition vein grafting, or a panel or 



chl9.qxd 4/16/04 3:33PM Page 375 



19 • EXTREMITY VASCULAR TRAUMA 



375 



spiral graft made from autologous vein. Early 
patency of these venous repairs is 50% to 90% 
(Meyer and colleagues, 1987; Pappas and 
colleagues, 1997), but long-term patency 
approaches 100% because it appears that the 
thrombus recanalizes and provides adequate 
function (Nypaver and colleagues, 1992; 
Pappas and colleagues, 1997). If the patient 
is in extremis, ligation of the subclavian or 
axillary vein is acceptable with minimal 
long-term sequelae (Timberlake and Kerstein, 
1995). If symptoms of venous claudication or 
severe swelling develop during rehabilitation 
or with the return of vigorous arm function, 
a subclavian venous bypass using autologous 
vein or ajugular venous "turn down"with tem- 
porary distal arteriovenous fistula provides 
satisfactory relief of symptoms. 



COMPARTMENT SYNDROME 
OF THE UPPER EXTREMITY 

A compartment syndrome can develop in 
either the upper arm (triceps, deltoid, or 
along the axillary sheath) or the forearm. The 
forearm compartment syndrome is more 
common. Increased tissue pressure can follow 
either blunt or penetrating trauma because 
of hematoma, post-traumatic transudation of 
serum into the interstitial space, venous 
thrombosis, or reperfusion following ischemia 
(Shackford and Rich, 2001). The possibility 
of a compartment syndrome must always be 
a consideration in a patient who has been 
injured, particularly one with prolonged 
ischemia before reperfusion. 

The diagnosis of compartment syndrome 
should be suspected in any patient com- 
plaining of increasing pain following injury. 
The physical findings include a tense com- 
partment, pain on passive range of motion, 
progressive loss of sensation, and weakness. 
The loss of arterial pulses is a late finding, 
which usually indicates a poor prognosis. Neu- 
rologic signs and symptoms, while helpful, are 
neither sensitive nor specific in the upper 
extremity following arterial injury because 
associated peripheral nerve injury often exists. 
Early diagnosis must be predicated on mea- 
surement of compartment pressures. The 



normal tissue compartment pressure ranges 
from to 9 mm Hg. Much controversy exists 
about what constitutes a pathologic elevation. 
Our approach has been to perform fas- 
ciotomy when compartment pressure exceeds 
30mmHg. 

Treatment consists of complete fasciotomy 
of the involved compartment. For the volar 
compartment, the skin incision begins 1 cm 
proximal and 2 cm lateral to the medial epi- 
condyle. It is carried obliquely across the skin 
crease at the antecubital fossa and continued 
obliquely for the proximal part of the forearm. 
It is then curved medially, reaching the 
midline at thejunction of the middle and distal 
third of the forearm, and is continued in a 
straight line to the wrist crease at a point on 
the medial side of the palmaris longus tendon. 
The incision is then curved obliquely across 
the wrist crease and terminated in the mid 
palm. This allows routine decompression of 
the carpal tunnel. A superficial fasciotomy 
adequately decompresses the volar com- 
partment in most cases. If any doubt exists 
about adequate decompression, intraopera- 
tive measurement of compartment pressures 
should be performed. For the dorsal com- 
partment, the incision begins 2 cm distal to 
the lateral epicondyle. It is carried straight 
distally in the midline for approximately 7 to 
10 cm. The skin edges are undermined and 
the dorsal fascia incised directly in line with 
the skin incision. 



POST-TRAUMATIC CAUSALGIA 



Persistent pain following upper extremity vas- 
cular injury is common due to associated 
peripheral nerve injury and resultant trau- 
matic neuralgia. Some patients may have pain 
that appears to be sympathetically mediated, 
but not all will have causalgia. Causalgia 
(complex regional pain syndrome type 2) 
occurs in about 3% of patients suffering 
peripheral nerve injuries (Costa and Robbs, 
1988) and is often confused with reflex sym- 
pathetic dystrophy (complex regional pain 
syndrome type 1). Several characteristics dis- 
tinguish causalgia: a burning pain noted 
within 24 hours of injury of a large mixed 



chl9.qxd 4/16/04 3:33PM Page 376 



376 



IV • SPECIFIC VASCULAR INJURIES 



nerve with a pain distribution similar to that 
of the nerve. Onset of reflex sympathetic 
dystrophy usually occurs weeks to months 
following the injury, is not always burning in 
character, and has a distribution that does not 
follow a specific anatomic distribution of a 
mixed nerve in the extremity. Causalgia typ- 
ically presents early and is associated with 
hypalgesia in the area of the partial denerva- 
tion followed by constant burning pain that 
can be increased by nonpainful stimuli (allo- 
dynia). Abnormal sympathetic function is 
evident in the region (e.g., vasomotion or 
hyperhidrosis) and the pain can be exagger- 
ated by emotional upset. 

The diagnosis should be suspected 
when the aforementioned characteristics are 
present. It can be confirmed by the relief 
of symptoms with a sympathetic block (Costa 
and Robbs, 1988). Some patients may 
have resolution of the syndrome with a single 
sympathetic block. For symptom recurrence, 
surgical sympathectomy is the treatment of 
choice. 



LOWER EXTREMITY 
VASCULAR INJURIES 



Common Femoral and 
Profunda Femoral Arteries 

SURGICAL ANATOMY 

The common femoral artery emerges from 
under the inguinal ligament as a continua- 
tion of the external iliac artery at the mid- 
point between the anterosuperior iliac spine 
and the pubic tubercle. It is relatively exposed 
with only subcutaneous fat and lymphatic 
tissue overlying. Along its course are three to 
five branches of varying size and location. The 
most prominent are the superficial circum- 
flex iliac and the superficial epigastric, which 
arise within 1 cm of the inguinal ligament. 
Approximately 5 cm below the inguinal liga- 
ment, the common femoral artery bifurcates 
into the superficial femoral and profunda 
femoral arteries. 



The profunda femoral artery usually 
originates as a single posterolateral branch. 
However, more than one profunda branch 
may be present. The lateral femoral circum- 
flex vein crosses the profunda anteriorly and 
transversely within 3 cm of its origin from 
the common femoral artery. At this level, 
the artery usually bifurcates into two large 
branches, the medial and lateral circumflex 
arteries. The proximity of the crossing vein 
demands careful attention when exposing the 
distal profunda. 

Numerous collaterals come from the 
branches of the hypogastric artery to the pro- 
funda femoral artery but are usually not suf- 
ficient to sustain adequate blood flow in the 
presence of an acute occlusion of the common 
femoral artery. The distal branches of the pro- 
funda femoral artery provide collateral flow 
to the popliteal artery through the lateral supe- 
rior genicular artery and the descending 
genicular artery. Following acute occlusion of 
the superficial femoral artery, these collater- 
als are not sufficient to sustain adequate blood 
flow to the lower leg (Fig. 19-17). 

The femoral nerve, composed predomi- 
nantly of the motor fibers of the quadriceps, 
traverses the femoral region along the lateral 
aspect of the femoral sheath and can be 
injured during exposure of the femoral vessels 
if an inadvertently lateral incision is used or 
excessive lateral retraction is present. 



EPIDEMIOLOGY AND ETIOLOGY 

Trauma to the femoral vessels accounts for 
one third of all vascular injuries in military 
series and 7% to 35% in civilian series (Rich, 
Baugh, and Hughes, 1970; Mattox and col- 
leagues, 1989; Humphrey, Nichols, and Silver, 
1994; Hafez, Woolgar, and Robbs, 2001 ) . Pen- 
etrating injuries are more common than 
blunt. Low-caliber gunshot wounds are the 
most common cause of penetrating injuries; 
knife wounds are less common (Hafez, 
Woolgar, and Robbs, 2001). Anterior dislo- 
cation of the femoral head is a rare cause of 
blunt injury. Laceration of the common 
femoral artery can cause severe hemorrhage 
that can be fatal if not tamponaded or 
controlled. In survivable injuries, the femoral 



chl9.qxd 4/16/04 3:33PM Page 377 



19 • EXTREMITY VASCULAR TRAUMA 



377 




Ext. iliac a 
Deep iliac 

Superf. iliac 
circumflex a 
Superf 
epigastric a 

Ascend, branch lat. 7 
circumflex a. | [ 
Transverse branch lat 
circumflex femoral a 
Lat. circumflex 
femoral a. 

Descend, branch lat 
circumflex femoral a 

I 

Perforating branches 

deep femoral a 



Right 
common iliac a. 

Int. iliac a. 
uj — Superior 
'*' gluteal a. 
Inf. gluteal a. 

Common 
femoral a. 

Obturator a. 

Medial circumflex 
femoral a. 



Superf. 
femoral a. 



Deep femoral a. 



Descend, 
genicular a. 



Lat. sup. genicular a 



■ FIGURE 19-17 

This anatomic drawing traces the course of the superficial femoral artery, the main conduit between 
the common femoral and popliteal arteries. In addition to numerous muscular branches, the 
supreme genicular (descending genicular) is an important collateral to the rich anastomosis around 
the knee. ■ 



sheath contains the hemorrhage and the 
vessel thromboses or forms an acute 
pseudoaneurysm. 



CLINICAL FEATURES AND DIAGNOSIS 

Hemorrhage is the most common presenting 
sign. Less commonly, the lacerated or tran- 
sected common femoral artery thromboses 
and distal ischemia results. Associated injury 



to the femoral vein is very common in pene- 
trating trauma. 

The superficial location of the femoral 
bifurcation allows for accurate clinical assess- 
ment by inspection and palpation. In patients 
with active hemorrhage from the femoral 
area, no diagnostic workup is needed; expedi- 
tious control of hemorrhage is required. 
Arteriography is reserved for patients with 
multiple associated injuries and an equivocal 
examination. Emergency department arteri- 



chl9.qxd 4/16/04 3:33PM Page 378 



378 



IV • SPECIFIC VASCULAR INJURIES 



ogram is usually not helpful in this area 
because of the difficulty of adequately visual- 
izing the common femoral artery and the 
femoral bifurcation. If arteriography is 
required, it should be performed formally in 
the angiography suite. 



SURGICAL TREATMENT 

Both groins, the lower abdomen (beginning 
at the umbilicus) , and both lower extremities 
should be completely prepared and draped. 
Preparing the lower abdomen allows for prox- 
imal extension of the incision and more prox- 
imal control if necessary. Having the uninjured 
groin prepared allows access to an alternative 
source of inflow. Preparing the uninjured leg 
provides access to autogenous conduit. 

The common femoral artery is best exposed 
through a longitudinal incision overlying its 
course from the inguinal ligament inferiorly 
for 8 to 12 cm (Fig. 19-18) . Occasionally, prox- 
imal control may require exposure of the 
external iliac artery. This is best accomplished 
through an oblique muscle splitting lower 
quadrant abdominal incision carried down to 
the retroperitoneum where the artery and vein 
can be controlled with medial retraction of 
the peritoneal structures. 

The profunda femoral artery is exposed 
through the same incision used for the 
common femoral artery. Dissection is carried 
distal and anterior along the proximal portion 
of the superficial femoral artery and poste- 
rior laterally to identify the origin of the 
profunda femoral. The proximal 2 cm of the 
artery is easily exposed. Beyond this point, 
ligating and dividing the lateral circumflex 
femoral vein exposes the artery. This vein is 
broad and short and should be carefully 
ligated to avoid significant hemorrhage. 

Severe hemorrhage usually dictates the 
initial steps of the surgical procedure. Proxi- 
mal and distal control prior to exposure of 
the injury site prevents secondary injury to 
the vessels. The use of vascular clamps, Silas- 
tic vessel loops, optical magnification, and 
fine monofilament sutures are essential to 
successful management. There is no role for 
blind clamp placement. Direct repair, when 
possible, is preferred. Longitudinal laceration 




■ FIGURE 19-18 

Exposure of the common femoral artery and its 
branches is best obtained through a 
longitudinal incision directly over the artery. 
Silastic loops double passed around the 
arteries provide control without causing 
secondary arterial trauma. The profunda 
femoral artery is exposed by carrying this 
dissection distally and by ligating the lateral 
femoral circumflex vein. (From Rutherford RB: 
Atlas of vascular surgery: Basic techniques 
and exposures. Philadelphia: WB Saunders, 
1993.) ■ 



or defects may be repaired with a vein patch 
angioplasty. However, if injury is extensive and 
debridement results in a significant loss of 
artery such that there would be tension on 
the repair, an interposition graft should be 
placed. 

Saphenous vein is the first choice for inter- 
position grafting. However, vein diameter may 
not be adequate. Although spiral vein graft 
construction is an alternative, it is time con- 
suming and technically demanding. Dacron 
or polytetrafluoroethylene (PTFE) interposi- 
tion grafts are acceptable alternatives. In 
general, PTFE is preferred because of its 
relative resistance to infection compared to 
Dacron. Short-segment synthetic grafts in this 
area of high flow are durable and have accept- 
able long-term patency rates (Feliciano and 
colleagues, 1985, 1988). 



chl9.qxd 4/16/04 3:33PM Page 379 



19 • EXTREMITY VASCULAR TRAUMA 



379 



Profunda femoral artery injuries should 
be repaired whenever possible. However, if 
serious associated injuries are present or the 
patient is unstable, the vessel should be 
ligated. Long-term sequelae are uncommon 
as long as the superficial femoral artery is 
patent. Proximal injuries to the profunda 
femoral artery may be managed by placing a 
short interposition graft or by proximal liga- 
tion and reimplantation of the vessel to the 
proximal superficial femoral artery. The pro- 
funda femoral artery should be repaired only 
if the patient is stable and the repair is rela- 
tively easy to accomplish. 



RESULTS 

Successful repair of the common femoral and 
profunda femoral arteries is dependent 
on restoration of adequate arterial lumen 
diameter and avoiding infection. Once groin 
wound infection occurs in patients who had 
an arterial repair, the first priority is to deter- 
mine whether the graft or suture line is 
exposed or involved in the infection. If the 
suture line is involved, immediate graft 
removal, ligation of the proximal and distal 
arteries, and extra-anatomic bypass are 
the only acceptable option to prevent life- 
threatening hemorrhage and eventual limb 
loss. If the graft is exposed, but the suture line 
is not and the graft is patent, the graft may be 
salvaged by coverage with a proximally based 
sartorius flap. 

Long-term patency rates of successful 
primary repair and short segment interposi- 
tion grafts are very good. Acute thrombosis, 
though uncommon, usually causes limb- 
threatening ischemia and requires immediate 
treatment. Early stenosis of vein interposition 
grafts is uncommon. These patients should 
have regular follow-up to assess graft patency 
and the adequacy of limb blood flow. Calf 
claudication is the first clinical indication of 
stenosis at the repair site. 

Lower extremity function following vascu- 
lar repair is predominantly determined by the 
severity of associated musculoskeletal and 
nerve trauma. The most disabling associated 
injury is femoral nerve transection. Loss of 
quadriceps function results in significant gait 



problems. Extensive venous injury with venous 
outflow obstruction at the femoral level causes 
venous insufficiency with long-term sequelae 
of venous stasis dermatitis and ulceration. 

Amputation rates following femoral artery 
injury vary from 15% to 35% and are deter- 
mined by the severity of musculoskeletal and 
neurologic injury (Mattox and colleagues, 
1989; Hafez, Woolgar, and Robbs, 2001 ) . Pen- 
etrating injuries are much less likely to result 
in amputation. In contrast, blunt injuries that 
cause vascular disruption usually involve force 
loading sufficient to cause significant neuro- 
logic and musculoskeletal injuries with limb- 
threatening sequelae. The most discouraging 
outcome is successful revascularization of a 
limb, which ultimately requires amputation 
for chronic recurring pressure ulceration and 
infection because of denervation. 



Superficial Femoral Artery 

SURGICAL ANATOMY 

The superficial femoral artery originates in 
the femoral triangle and travels from an ante- 
rior location to the medial aspect of the thigh 
at the adductor canal where it transitions to 
the popliteal artery. It is superficially located 
in the groin and moves deeper as it traverses 
the medial thigh beneath the sartorius muscle 
approaching the adductor magnus muscle. 
The only significant branch is the descend- 
ing genicular artery, which forms a collateral 
anastomosis with the genicular branches of 
the popliteal artery. The superficial femoral 
vein travels in close posteromedial proximity 
to the artery and is frequently duplicated. 
The saphenous nerve, a cutaneous sensory 
nerve to the medial calf and foot, lies ante- 
rior to the superficial femoral artery for most 
of its course. The nerve leaves the artery to 
join the saphenous vein near the adductor 
hiatus. 



EPIDEMIOLOGY AND ETIOLOGY 



Penetrating 



injury of superficial femoral 
artery is more common than blunt. The pres- 
ence of a femoral shaft fracture should alert 



chl9.qxd 4/16/04 3:33PM Page 380 



380 



IV • SPECIFIC VASCULAR INJURIES 



the examining physician of the possibility of 
a superficial femoral artery injury, but fewer 
than 5% of fractures will have vascular trauma 
(Rosental and colleagues, 1975; Romanoff and 
Goldberg, 1979). 



CLINICAL FEATURES AND DIAGNOSIS 

Hemorrhage is the predominant feature of 
penetrating vascular injuries in the thigh, 
whereas thrombosis is the usual presentation 
following a blunt mechanism. 

High-velocity gunshot wounds of the thigh, 
though common in the military setting, 
remain rare in the civilian environment. Con- 
tusion and thrombosis of the superficial 
femoral artery produced by the temporary cav- 
itational effects of high-energy rounds may 
present as either initial or delayed lower 
extremity ischemia. The severity of tissue 
destruction, neurologic deficit, and vascular 
spasm may make peripheral vascular exami- 
nation difficult in this setting. 

Inspection and palpation with attention to 
distal pulses is usually accurate in assessing 
the superficial femoral artery. Frequent re- 
examination, particularly in patients with 
midshaft femur fracture, must be performed 
to avoid missing a delayed arterial thrombo- 
sis. Patients with active hemorrhage from 
penetrating wounds require immediate 
operation for hemorrhage control and diag- 
nosis. Arteriography is reserved for patients 
with equivocal signs of arterial injury, palpa- 
ble but diminished pulses, or the suspicion 
of pseudoaneurysm or arteriovenous fistula. 
Emergency department or operating room 
arteriography is accurate for detecting 
superficial femoral artery injuries and is time 
saving in patients with multiple injuries and 
the need for immediate thoracotomy or 
celiotomy. 



SURGICAL TREATMENT 

Both groins and both legs should be prepared 
and draped. Preparing the contralateral groin 
provides an alternative source of inflow and 
the contralateral leg provides a source for 
autologous conduit. Proximal superficial 



femoral artery injuries are best exposed 
through a longitudinal groin incision similar 
to that used for femoral bifurcation exposure. 
The middle and distal artery can be 
approached through an oblique incision in 
the thigh over the course of the sartorius 
muscle. The muscle is retracted medially and 
the artery found immediately below in the 
adductor (Hunter) canal. Exposure of the 
distal artery at the superficial femoropopliteal 
arteryjunction may require transection of the 
adductor magnus tendon. 

Primary repair is possible for those few 
wounds that produce a small, clean laceration. 
Saphenous vein interposition grafting is the 
best procedure for more severe injuries of the 
superficial femoral artery. Careful vascular 
technique, avoiding undue tension in the 
repair, preserving lumen diameter, and com- 
pletion angiogram are essential to the suc- 
cessful repair of the superficial femoral artery. 
A synthetic graft is an acceptable conduit if 
no vein is available or the patient is too unsta- 
ble to prolong the procedure to harvest a vein. 
Long-term patency rates of PTFE and Dacron 
grafts are significantly lower than that of autol- 
ogous vein graft. 

Primary amputation is rarely indicated in 
the management of superficial femoral artery 
injuries. Extensive crush injury with avulsion 
of the thigh muscles from the femur is one of 
the few indications for lifesaving above- 
knee amputation. Refractory hemorrhage 
into the thigh is striking in these patients and 
attempts at direct surgical control of hemor- 
rhage are usually futile. High above-knee 
amputation may be the only way to control 
life-threatening hemorrhage in this setting. 



RESULTS 

Superficial femoral artery repair, if done prop- 
erly, has long-term patency that approaches 
100% . Associated neurologic injury is uncom- 
mon and femur fractures are usually amenable 
to successful orthopedic management. Fre- 
quent postoperative assessment for graft 
failure or calf compartment syndrome is nec- 
essary to facilitate early reoperation or fas- 
ciotomy should these complications occurs. 
Amputation rates vary from 10% to 30% and 



chl9.qxd 4/16/04 3:33PM Page 381 



19 • EXTREMITY VASCULAR TRAUMA 



381 



depend on the timeliness and success of the 
vascular repair and the severity of associated 
injuries (Mattox and colleagues, 1989; Hafez, 
Woolgar, and Robbs, 2001). 

Patients with superficial femoral artery 
injuries require long-term follow-up. Yearly 
assessment of distal pulses and, if indicated, 
segmental lower extremity arterial pressures 
should be performed. Five percent to twenty 
percent of patients will require some form 
of secondary reconstruction for lower 
extremity arterial insufficiency because of a 
late failure (stenosis or thrombosis) of the 
repair. 



Popliteal and Tibial Arteries 

SURGICAL ANATOMY 

The popliteal artery originates at the adduc- 
tor magnus hiatus as the continuation of the 
superficial femoral artery. Throughout its 
course, the popliteal artery is located deep in 
the popliteal fossa along the posterior aspect 
of the femur, in proximity to the joint line, 
and the tibial plateau. The artery is covered 



proximally by the semimembranous muscle 
and in its midportion by subcutaneous tissue. 
The artery continues distally to the upper calf 
where it terminates at the origin of the ante- 
rior tibial artery at the triceps surae formed 
by the two heads of the gastrocnemius muscle 
and the soleus muscle. Along its course, the 
popliteal artery has six to eight small genicu- 
late branches, which are usually paired. These 
form an anastomotic network around the knee 
(Fig. 19-19). However, in acute occlusion of 
the popliteal artery, these branches are not 
sufficient to provide adequate distal blood 
flow. 

The relationship of the popliteal artery to 
the muscles of the thigh and calf places it at 
risk for severe injury in the dislocation of the 
knee. In full extension of the knee, the 
popliteal artery is on tension across the back 
of the knee joint. Knee dislocation stretches 
the popliteal artery over the posterior edge 
of the tibial plateau resulting in severe intimal 
injury or transection. 

The three tibial vessels have a variable 
origin. In 85% to 90% of patients, the popliteal 
bifurcates into the anterior tibial and tibial 
peroneal trunk arteries. The posterior tibial 



Descend, branch 
tat. circumflex 
femoral a. 

Popliteal a. 



Superior lat 
genicular a 



Inf. lat. genicular a 



Ant. tibial 
recurrent a. 

Ant. tibial a 




Femoral a. 

Descend, 
genicular a. 

Articular branches 
descend, genicular a. 

Superior medial 
genicular a. 



Inf. medial 
genicular a. 

Post, tibial a. 



■ FIGURE 19-19 

Anterior view of the knee with the popliteal artery and its branches. These collaterals are usually not 
sufficient to provide adequate distal perfusion in patients with acute traumatic occlusion. ■ 



chl9.qxd 4/16/04 3:33PM Page 382 



382 



IV • SPECIFIC VASCULAR INJURIES 



and peroneal artery arise 3 to 6 cm distally. 
In 10% to 15%, variations are seen in the 
tibial vessel origins from the popliteal artery 
including an origin of the anterior or poste- 
rior tibial arteries at or above the knee joint 
line. The popliteal and tibial arteries are 
accompanied in their course by single or 
paired veins. 

The anterior tibial artery traverses the 
superior edge of the interosseus membrane 
to enter the anterior compartment of the calf. 
It courses along the membrane accompanied 
by the deep peroneal nerve and the anterior 
tibial vein. This neurovascular bundle lies deep 
to the extensor muscles. The artery contin- 
ues across the ankle joint beneath the exten- 
sor retinaculum to emerge on the top of the 
foot at the dorsalis pedis artery. Along its 
course, the anterior tibial artery gives off 
numerous muscular branches. The dorsalis 
pedis terminates in the superficial plantar 
arch. 

The posterior tibial and peroneal arteries 
originate at the bifurcation of the tibial- 
peroneal trunk in the upper calf deep to the 
soleus muscle. The posterior tibial artery con- 
tinues along the fascia of the deep posterior 
muscle compartment accompanied by the 
tibial and paired posterior tibial veins. It tra- 
verses the ankle joint posterior to the medial 
malleolus and terminates in the medial and 
lateral plantar arteries, which contribute to 
the deep and superficial plantar arches. Anas- 
tomotic connections between the anterior 
tibial artery and posterior tibial artery allow 
for adequate foot perfusion as long as one of 
the vessels remains patent. 

The peroneal artery parallels the posterior 
tibial artery in a lateral course deep to the 
flexor hallucis longus muscle. It is accompa- 
nied by paired veins. Distally it terminates in 
lateral calcanean branches that anastomose 
with distal branches of the anterior tibial and 
posterior tibial arteries. These connections are 
small and may not be sufficient to supply the 
foot in acute occlusion of the other tibial 
vessels. The distal anterior or posterior tibial 
arteries may be supplied by a large terminal 
branch of the peroneal artery either as a 
congenital anomaly or because of collateral- 
ization after chronic occlusion of those vessels. 



EPIDEMIOLOGY AND ETIOLOGY 

Blunt trauma causes most civilian popliteal 
and tibial arterial injuries. Fracture or dislo- 
cation in the area of the knee is the predom- 
inate mechanism. In the military experience, 
penetrating injuries are more common. Occlu- 
sion of a single tibial vessel is well tolerated as 
long as no preexisting occlusion of the other 
vessels is present. 



CLINICAL FEATURES AND DIAGNOSIS 

Thrombosis with distal ischemia is the most 
common presentation of popliteal artery 
injury. Concomitant venous and neurologic 
trauma makes these injuries extremely 
morbid. Most patients with occlusion of the 
popliteal or more than one tibial artery occlu- 
sion have calf and foot ischemia. On the other 
hand, knee dislocation with spontaneous 
reduction may be overlooked unless a thor- 
ough peripheral vascular examination is per- 
formed. In these cases, the dislocation causes 
a shear effect producing intimal injury and 
delayed thrombosis. 

A thorough peripheral vascular examina- 
tion in all injured patients is the key to prompt 
recognition of popliteal and tibial artery 
injuries. Delays in diagnosis are invariably due 
to the lack of a physical examination of the 
pulses augmented with Doppler pressure 
determination when indicated. Ankle Doppler 
pressure determination allows rapid assess- 
ment of the patient with diminished distal 
pulses. Absence of Doppler flow sounds, an 
ankle brachial index of less than 0.8 or a 
20mmHg decrease compared to the unin- 
jured leg all indicate the need for further 
evaluation. 

Duplex scanning has no role in the assess- 
ment of acute arterial trauma, adds nothing 
to physical examination augmented by 
Doppler pressure measurement, and is not 
accurate enough to assist in planning surgi- 
cal treatment. Even when used by experienced 
surgeons or technicians, this technology does 
not obviate the need for arteriography or accu- 
rately predict the success of nonoperative 
therapy. 



chl9.qxd 4/16/04 3:33PM Page 383 



19 • EXTREMITY VASCULAR TRAUMA 



383 



Arteriography is accurate in the evaluation 
of patients with suspected popliteal or tibial 
arterial injury but is time consuming and 
should be reserved for patients with equivo- 
cal physical findings. Emergency department 
or intraoperative angiography is also accurate 
and should be considered when formal 
angiography is not readily available. 



SURGICAL TREATMENT 

Both groins and both lower extremities should 
be prepared and draped. Contralateral saphe- 
nous vein is the conduit of choice for bypass 
and should be readily accessible. Popliteal 
injuries are best approached through a 
generous medial incision (Fig. 19-20). The 
simplest landmarks for the incision are the 
posterior margin of the femur proximally and 
the posterior margin of the tibia below the 



knee. During the medial exposure, care 
should be taken to avoid lacerating the saphe- 
nous vein. Post-traumatic deep venous insuf- 
ficiency is common and this superficial vein 
may become an important collateral route of 
venous drainage. The proximal popliteal 
artery is exposed as it emerges from adduc- 
tor canal. Exposure of the artery in the area 
of the kneejoint requires division of the medial 
head of the gastrocnemius, semimembra- 
nosus, and semitendinosus muscles. The distal 
popliteal artery is exposed with an incision 
along the posterior margin of the tibia 
(Fig. 19-21). 

Tibial vessel exposure requires careful dis- 
section in the upper medial aspect of the calf. 
The origin of these vessels is exposed by con- 
tinuing dissection from the distal popliteal 
artery through the area of the triceps surae. 
The exposure is facilitated by incising the 
soleus muscle longitudinally 2 cm posterior to 



Sciatii 



Posterior Approach 




Popliteal vv. 



■ FIGURE 1 9-20 

The posterior and medial 
approaches to the popliteal artery. 
A, A modified S-shaped incision is 
used in the posterior approach to 
avoid contracture across the knee 
joint. B, The medial approach 
requires a more extensive dissection 
but provides better access to 
proximal and distal vessels. C, Both 
approaches can be successfully 
used in the exposure and repair of 
the popliteal vessels. ■ 



chl9.qxd 4/16/04 3:33PM Page 384 



384 



IV • SPECIFIC VASCULAR INJURIES 



A 





D 





■ FIGURE 19-21 

A, Distal popliteal exposure is obtained through an incision posterior to the tibia. 6 and C, The 
soleus muscle is divided longitudinally to expose the neurovascular bundle. D, The distal popliteal 
artery and anterior tibial artery origin is exposed ligating the anterior tibial vein and retracting the 
popliteal vein posteriorly. E, The tibioperoneal trunk and origins of the peroneal and posterior tibial 
arteries are exposed by retracting the popliteal vein anteriorly. (From Rutherford RB: Atlas of 
vascular surgery: Basic techniques and exposures. Philadelphia: WB Saunders, 1993.) ■ 



chl9.qxd 4/16/04 3:33PM Page 385 



the tibia, taking care to avoid the soleal plexus 
of veins adjacent to the tibia. The anterior 
tibial artery origin is exposed by retracting the 
popliteal vein posteriorly. The anterior tibial 
vein should be carefully ligated. This short 
broad vein is difficult to control if lacerated. 
Once divided, it allows for exposure of the 
origin of the anterior tibial artery and the tibial 
peroneal trunk. The vessels distal to the ante- 
rior tibial origin are best exposed by retract- 
ing the veins anteriorly. Paired veins with 
crossing branches envelop the proximal pos- 
terior tibial and peroneal arteries. Distal 
exposure of these vessels is obtained through 
a medial incision along the posterior margin 
of the tibia down to the space posterior to the 
medial malleolus. The anterior tibial artery 
is exposed through an incision along the 
middle of the anterior compartment. Dissec- 
tion is carried deep between the extensor hal- 
lucis and extensor digitorum muscles to the 
level of the interosseus membrane and the 
artery (Fig. 19-22). 

Popliteal and tibial arterial injuries are 
rarely simple lacerations. Repair usually 
requires saphenous vein interposition. 
Primary repair of the popliteal artery is appro- 
priate in lacerations from knife wounds, 
which result in little arterial disruption. Blunt 
injuries and gunshot wounds should be treated 
by careful debridement of all injured vessel 
wall and tension-free repair with vein inter- 
position grafting (Shah and colleagues, 1985) . 

Arterial repair at the popliteal and tibial 
level should always be evaluated with inter- 
operative completion angiography. Any defect 
in the repair should be immediately addressed 
with either a catheter thrombectomy or a revi- 
sion of the anastomosis. Early occlusion with 
platelet thrombus should be carefully inves- 
tigated to rule out a technical defect in the 
repair. If platelet deposition in the area of 
repair occurs, a continuous infusion of low- 
molecular-weight dextran should be started. 
This is a treacherous clinical problem and 
must be aggressively treated. 

Soft tissue coverage of arterial repairs in the 
region of the knee and calf is essential to suc- 
cessful limb salvage. Infection or exposure of 
vein interposition grafts always leads to throm- 
bosis or hemorrhage and a high rate of limb 
loss. 



19 • EXTREMITY VASCULAR TRAUMA 385 

RESULTS 

Injury of the popliteal and tibial vessel level 
is associated with significant long-term dis- 
ability. The initial outcome is dependent on 
successful arterial repair and the extent of 
soft tissue damage and ischemia. Infections 
of arterial repairs at this level are usually asso- 
ciated with inadequate soft tissue coverage. 
Long-term results are dependent on the 
extent of musculoskeletal and neurologic 
injury. Tibial nerve transection is associated 
with poor long-term results. Early physical 
therapy is essential to maximize the recovery 
of function in all lower extremity vascular 
injuries. 

Lower Extremity 
Compartment Syndrome 

Compartment syndrome may present 12 to 
24 hours after reperfusion. If not promptly 
diagnosed and treated, the risk of limb loss 
or severe dysfunction is high. Calf compart- 
ment syndrome most commonly results from 
prolonged ischemia or a crush injury. Fre- 
quent physical examinations augmented 
with compartment pressure measurements are 
necessary to detect this complication in its 
early stage. The first complaint may be sensory 
loss in the foot. Thigh compartment syndrome 
is rare. Thigh muscle swelling and pain out 
of proportion to the severity of injury are the 
most common findings. 

Mangled Lower Extremity 

Every effort must be made to balance the sur- 
gical reconstruction of the mangled extrem- 
ity with the overall status of the patient both 
immediately following injury and during the 
rehabilitation phases of care. Primary ampu- 
tation should be considered in patients with 
severe soft tissue injury and a dysvascular 
extremity. Objective rating scales or scoring 
systems are an adjunct to clinical judgment 
but not a substitute for careful consideration 
of what is reasonable and appropriate for the 
patient's short-term and long-term recovery 
(Gregory and colleagues, 1985; Johansen 



chl9.qxd 4/16/04 3:33PM Page 386 



386 



IV • SPECIFIC VASCULAR INJURIES 



A 







■ FIGURE 19-22 

A, Proximal exposure of the anterior tibial artery is obtained through an incision along the 
anterolateral aspect of the calf. B, The extensor muscles are retracted anteriorly and posteriorly to 
expose the anterior tibial neurovascular bundle on the interosseous membrane. C, Distal exposure 
is obtained through an incision along the extensor digitorum tendon. D, The anterior tibial artery lies 
adjacent to the tibia beneath the flexor tendons and the extensor retinaculum. (From Rutherford RB: 
Atlas of vascular surgery: Basic techniques and exposures. Philadelphia: WB Saunders, 1993.) ■ 



chl9.qxd 4/16/04 3:33PM Page 387 



19 • EXTREMITY VASCULAR TRAUMA 



387 




■ FIGURE 19-23 

Mangled extremity with severe degloving injury 
associated with an open femur fracture. This 
limb was initially thought to be unsalvageable, 
but with appropriate revascularization and 
debridement, the extremity was saved and was 
functional. ■ 



and colleagues, 1990; Bonanni, Rhodes, and 
Lucke, 1993). Delayed amputation following 
initially successful vascular repair remains an 
unfortunate possibility in patients with exten- 
sive musculoskeletal and neurologic injury. 
Most trauma centers pursue an aggressive 
approach to young patients with these 
complex injuries. Mangled extremities are 
often salvaged with a multidisciplinary 
approach; however, amputation is eventually 
the best choice for a pain-free return to func- 
tional status in some patients (Fig. 19-23) . This 
decision is never easily made and is based on 
a strong physician-patient partnership. 
Setting reasonable expectations immediately 
after injury is the best starting point in 
this process. Early involvement of a multi- 
disciplinary approach also allows for a timely 
decision about amputation. This remains one 
of the most challenging problems for trauma 
surgeons. 



chronic disability. A balanced approach that 
includes ligation for injuries of minor veins 
or for life-threatening injuries in unstable 
patients, and repair or reconstruction 
whenever possible may be the best strategy 
(Nypaver and colleagues, 1992; Timberlake 
and Kerstein, 1995; Zamir and colleagues, 
1998). 



SURGICAL ANATOMY 

Paired veins form from muscle branches in 
the calf and course parallel to the tibial vessels 
and coalesce into the popliteal vein, which may 
also be paired. The calf is also drained by 
numerous subcutaneous veins that flow to 
either the greater or the lesser saphenous vein. 
The greater saphenous veinjoins the common 
femoral vein in the groin and the lesser saphe- 
nous vein travels proximally along the back 
of the calf to join the popliteal vein. Numer- 
ous perforating veins connect the greater 
saphenous to the deep system. Flow is directed 
deep and proximally by valves in both the 
superficial and the deep system. 

The popliteal vein transitions the adductor 
canal to become the superficial femoral vein. 
Numerous muscular branches join this vein. 
The profunda femoral vein drains the quadri- 
ceps and deep muscles and joins the superfi- 
cial femoral vein to form a single large 
common femoral vein. All of the lower extrem- 
ity veins collateralize to form an extensive 
network of alternative venous drainage routes. 
In the absence of disruption of these collat- 
erals, a single-level venous occlusion does not 
prevent adequate venous drainage. 



EPIDEMIOLOGY AND ETIOLOGY 



Venous Injuries of the 
Lower Extremity 

The aggressive approach to arterial injuries 
in the extremities has not been matched with 
equal enthusiasm for venous repair. Instead, 
ligation is frequently performed. However, lig- 
ation may result in thrombosis and venous 
insufficiency, ultimately leading to significant 



Venous injury is usually associated with arte- 
rial injury and is most commonly due to pen- 
etrating trauma. Veins are much more elastic 
than arteries and are less frequently injured 
in blunt force trauma. Fractures, however, can 
lead to lacerations and thrombosis. The low- 
pressure venous system allows for tamponade, 
and significant external hemorrhage is 
uncommon except in large lacerations of the 
femoral and popliteal veins. 



chl9.qxd 4/16/04 3:33PM Page 388 



388 IV • SPECIFIC VASCULAR INJURIES 

CLINICAL FEATURES AND DIAGNOSIS 

Most clinically significant venous injuries 
present as persistent hemorrhage of dark red 
blood from a penetrating wound. In the 
absence of external hemorrhage or the need 
for surgical exploration for concomitant arte- 
rial trauma, the diagnosis of venous injury is 
usually delayed. Venous hypertension fol- 
lowing major vein injury and thrombosis is 
usually well tolerated despite causing distal 
extremity edema. Lower extremity ischemia 
is rare following venous injury and is the result 
of extensive soft tissue damage and loss of col- 
lateral venous flow. Compartment syndrome 
is also uncommon and is usually delayed in 
onset when it occurs. 

Duplex scanning is an effective and accu- 
rate diagnostic modality to assess venous 
patency in blunt-force injuries. Most trauma 
centers periodically perform surveillance 
duplex scanning of the lower extremities of 
trauma patients following major injuries. 
Dynamic computed tomographic scanning 
and arteriography are rarely used to diagnose 
venous injuries. Most venous injuries of clin- 
ical significance are diagnosed at the time of 
surgical exploration for arterial trauma due 
to the high rate of coincidental injuries. 



SURGICAL TREATMENT 

The veins of the lower extremity are exposed 
by the same incisions used to expose the asso- 
ciated arteries. Direct pressure and proximal 
and distal dissection in adjacent tissue allows 
hemostatic control. Clamp application should 
be carefully performed or control obtained 
with Silastic vessel loops to avoid secondary 
venous trauma. 

The choice of technique for venous repair 
depends on the patient's overall status, the 
extent of venous and soft tissue injury, and 
the duration of ischemia from associated arte- 
rial injury. Life-threatening associated injuries 
or hemodynamic instability mandate ligation 
of venous injuries. Extensive soft tissue injury 
and loss of collateral venous flow mandate 
venous reconstruction. Prolonged ischemia 
from arterial occlusion leads to a dilemma 
when venous injury is associated. If possible, 



the vein should be repaired first. Simple lac- 
erations may be repaired by lateral suture, 
taking care to avoid stenosis. Occasionally, end- 
to-end repair is possible. 

If extensive venous reconstruction is 
required and associated arterial injury is 
present, it is best to restore arterial perfusion 
with a shunt. Venous interposition grafts or 
venous panel grafts should be used to restore 
adequate lumen diameter. Panel grafts are 
constructed by harvesting a sufficient length 
of contralateral saphenous vein, opening the 
vein longitudinally, wrapping it in a spiral 
fashion around an appropriately sized chest 
tube, and sewing the vein into a conduit for 
interposition in the injured vein. This tedious 
and time-consuming procedure requires 
loupe magnification and precise technique. 
The use of synthetic grafts for the repair of 
lower extremity venous injuries is to be 
avoided because of the certainty of early 
thrombosis. 

The use of small arteriovenous fistula 
upstream from the venous repair to maintain 
high flow has been suggested. This technique 
is time consuming and unproven in its effi- 
cacy in the management of venous injuries. 
A low-molecular-weight dextran infusion to 
limit platelet adhesion carries a low risk of 
bleeding complications and may help main- 
tain early patency of the repaired vein. Dextran 
40 is infused at 40 mL per hour for 24 hours. 
Full anticoagulation with intravenous hepa- 
rin is associated with a significant risk of bleed- 
ing and is best reserved for proven deep 
venous thrombosis. 



RESULTS 

A traditional skepticism exists among surgeons 
about the likelihood of venous patency after 
reconstruction for trauma. Well-performed 
repairs remain patent at an encouraging rate. 
Early patency depends on the avoidance of 
stenosis and prevention of thrombosis. If early 
thrombosis occurs, recanalization occurs in a 
significant number of patients. If ligation is 
required, early postinjury edema can be 
minimized by placing the lower extremity in 
a continuous passive mobilization device. 
Pulmonary emboli are uncommon following 



chl9.qxd 4/16/04 3:33PM Page 389 



19 • EXTREMITY VASCULAR TRAUMA 



389 



venous repair (Nypaver and colleagues, 1992; 
Timberlake and Kerstein, 1995; Zamir and col- 
leagues, 1998), but chronic venous insuffi- 
ciency is common. 



REFERENCES 

Bonamii F, Rhodes M, Lucke JF: The futility of pre- 
dictive scoring of mangled lower extremities. 
J Trauma 1993;34:99-105. 

Costa MC, Robbs JV: Nonpenetrating subclavian 
artery trauma. J Vase Surg 1988;8:71-75. 

Endean ED, Veldenz HC, Schwarcz TH, Hyde GL. 
Recognition of arterial injury in elbow disloca- 
tion. J Vase Surg 1992;16:402-406. 

Feliciano DV, Herskowitz K, O'Gorman RB, et al: 
Management of vascular injuries in the lower 
extremity. J Trauma 1988;28:319-328. 

Feliciano DV, Mattox KL, Graham JM, Bitondo CG: 
Five-year experience with PTFE grafts in vascu- 
lar wounds. J Trauma 1985;25:71-82. 

Graham JM, Mattox KL, Feliciano DV, DeBakey 
ME: Vascular injuries of the axilla. Ann Surg 
1982;195:232-238. 

Granchi T, Schmittling Z, VasquezJ, et al: Prolonged 
use of intraluminal arterial shunts without sys- 
temic anticoagulation. Am J Surg 2000;180:493- 
497. 

Gregory RT, GouldRJ,PecletM,etal: The mangled 
extremity syndrome (MES): a severity grading 
system for multi-system injury of the extremity. 
J Trauma 1985;25:1147-1150. 

Hafez HM, Woolgar J, Robbs JV: Lower extremity 
arterial injury: results of 550 cases and review of 
risk factors associated with limb loss. J Vase Surg 
2001;33:1212-1219. 

Hardin WD, O'Connell RC, Adinolfi MF, Kerstein 

MD: Traumatic injuries of the upper extremity: 
determinants of disability. Am J Surg 1985;150: 
266-270. 

Humphrey PW, Nichols WK, Silver D: Rural vas- 
cular trauma: a twenty year review. Ann Vase Surg 
1994;8:179-185. 

Johansen K, Dames M, Howey T, et al: Objective 
criteria accurately predict amputation following 
lower extremity trauma. J Trauma 1990;30:568- 
573. 

Johnson M, Ford M, Johansen K: Radial or ulnar 
artery laceration. Repair or ligate? Arch Surg 
1993;128:971-975. 



Mattox KL, Feliciano DV, Burch J, et al: Five thou- 
sand seven hundred sixty cardiovascular injuries 
in 4459 patients. Epidemiologic evolution 1958 
to 1987. Ann Surg 1989;209:698-674. 

Meyer J, Walsh J, Schuler J, et al: The early fate of 
venous repair after civilian vascular trauma. A 
clinical, hemodynamic, and venographic assess- 
ment. Ann Surg 1987;206:458-464. 

Nypaver TJ, Schuler JJ, McDonnell P, et al: Long- 
term results of venous reconstruction after 
vascular trauma in civilian practice. J Vase Surg 
1992;16:762-768. 

Pappas PJ, Haser PB, Teehan EP, et al: Outcome 
of complex venous reconstructions in patients 
with trauma. J Vase Surg 1997;25:398-404. 

Rich NM, Baugh JH, Hughes CW: Acute arterial 
injuries in Vietnam: 1 ,000 cases. J Trauma 1970; 
10:359-369. 

Rich NM, Spencer FC: Subclavian artery injuries. 
In: Vascular Trauma. Philadelphia: WB Saunders, 
1978:307-329. 

Romanoff H, Goldberger S: Combined severe vas- 
cular and skeletal trauma: management and 
results. J Cardiovasc Surg 1979;20:493-498. 

RosentalJJ, Caspar MR, Gjerdrum TC, Newman J: 
Vascular injuries associated with fractured femur. 
Arch Surg 1975;110:494-499. 

Rozycki GS, Tremblay LN, Feliciano DV, 
McClelland WB: Blunt vascular trauma in the 
extremity: Diagnosis, management, and outcome. 
J Trauma 2003;55(5) :814-824. 

Sampson LN, Britton JC, Eldrup-Jorgensen J, et al: 
The neurovascular outcome of scapulothoracic 
dissociation. J Vase Surg 1993;17:1083-1089. 

Shackford SR, Rich NM: Peripheral vascular injury. 
In: Mattox KL, Moore EE, Feliciano DV, eds. 
Trauma. Philadelphia: WB Saunders, 2001 :101 1- 
1046. 

Shah DM, Naraynsingh V, Leather RP, et al: 
Advances in the management of popliteal vas- 
cular blunt injuries. J Trauma 1985;25:793-799. 

Sparks SR, DeLaRosa J, Bergan JJ, et al: Arterial 
injury in uncomplicated upper extremity dislo- 
cations. Ann Vase Surg 2000;14:110-113. 

Timberlake GA, Kerstein MD: Venous injury: to 
repair or ligate, the dilemma revisited. Am Surg 
1995;61:139-145. 

Zamir G, Berlatzky Y, Rivkind A, et al: Results of 
reconstruction in major pelvic and extremity 
venous injuries. J Vase Surg 1998;28:901-908. 



ch20.qxd 4/16/04 3:31PM Page 393 





Special Problems 



ERIC R. FRYKBERG 



" 



O 

o 



POPLITEAL ARTERY INJURIES 
History and Epidemiology 
Diagnostic Issues 

General principles and modalities 
Posterior Knee Dislocation 
Treatment 

General principles and techniques 

Surgical adjuncts 

Nonoperative observation 
MANAGEMENT OF VENOUS INJURIES 
History and Epidemiology 
Diagnosis 
Treatment 

General principles 

Venous repair considerations 

Venous ligation considerations 

Roles of ligation and repair 
COMBINED VASCULAR AND SKELETAL EXTREMITY TRAUMA 
Epidemiology and Prognostic Factors 
Diagnosis 
Treatment 

Indications for Amputation 
VASCULAR GRAFTS: ROLE AND COMPLICATIONS 
FAILED RECONSTRUCTION OF ARTERIAL TRAUMA 



393 



ch20.qxd 4/16/04 3:31PM Page 394 



394 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



There are a number of distinct clinical 
presentations, problems, and issues 
in vascular trauma that pose special 
challenges in diagnosis and management for 
the surgeon who is confronted with these 
injuries. Each of these special problems is 
plagued by relatively poor outcomes, even in 
the most experienced trauma centers. They 
tend to be uncommon and complex and 
require rapid detection and treatment, mul- 
tidisciplinary prioritization, and innovative 
management techniques. The purpose of this 
chapter is to provide a thorough knowledge 
of the history, epidemiology, current litera- 
ture, and suggested approaches for some of 
the most difficult of these problems to opti- 
mize outcome. 



POPLITEAL ARTERY INJURIES 

The special challenge of injury to the 
popliteal artery lies primarily in the anatomy 
of this vessel, which begins as the continua- 
tion of the superficial femoral artery as it 
courses through the hiatus of the adductor 
magnus muscle. Covered proximally by the 
semimembranosus muscle, it lies only in sub- 
cutaneous tissue in the popliteal fossa behind 
the knee joint, situated between the two heads 
of the gastrocnemius muscle. It is in this fossa 
that the popliteal artery is especially vulnera- 
ble to stretch and direct injury from extrin- 
sic forces and skeletal distortions, such as 
fractures and knee dislocations, being teth- 
ered proximally and distally to the femur and 
tibia by tendons of the adductor and soleus 
muscles. It most often bifurcates twice. As 
the anterior tibial artery branches laterally 
through the interosseous septum, the main 
artery continues for another 2 to 3 cm as the 
tibioperoneal trunk. This vessel then termi- 
nally bifurcates into the peroneal and poste- 
rior tibial arteries. Several geniculate, sural, 
and muscular collateral vessels branch from 
the popliteal artery behind the knee, which 
anastomose in a rich network with branches 
of the profunda femoris artery proximally and 
tibial arteries distally. However, this collateral 
supply is frail and subject to obliteration and 



thrombosis by injury to the main artery and 
surrounding tissues. These collaterals cannot 
maintain viability of the leg and foot on their 
own. 

The fact that the popliteal artery is a true 
end artery with a tenuous collateral support 
explains why injury to it is so dangerous, and 
why such injury has long been recognized as 
the most limb threatening of all peripheral 
vascular trauma. Nonetheless, several recent 
advances in diagnosis and treatment of 
popliteal artery injury have led to dramatic 
reductions in limb loss and limb morbidity. 

History and Epidemiology 

General Albert Sidney Johnston died in the 
American Civil War during the Battle of 
Shiloh in April 1862 of exsanguination from 
a gunshot wound to the popliteal artery, an 
injury that currently would be considered 
quite treatable and not life threatening. In 
1906, the first use of autogenous vein to repair 
an arterial injury was reported by Goyanes for 
a traumatic aneurysm of the popliteal artery. 
Injuries of this artery represented 12% of all 
arterial injuries in British troops in World 
War I, 20% of arterial injuries in American 
troops in World War II, 26% of those in 
the Korean War, and 217 (21.7%) of 1000 
arterial injuries in the Vietnam War. These 
were virtually all due to penetrating trauma, 
from bomb and land-mine fragments and 
high-velocity gunshots. Over the past 25 years, 
popliteal artery injuries in the civilian sector 
account for approximately 20% of all extrem- 
ity arterial injuries reported in the published 
literature, and as many as 20% to 75% of cases 
in this setting are caused by blunt mechanisms. 
The standard treatment of all extremity 
arterial trauma before the Korean War was 
ligation. DeBakey and Simeone (1946) doc- 
umented this approach to result in limb loss 
in 72.5% of all popliteal artery injuries in 
World War II, the highest of any extremity 
artery. Additionally, many salvaged limbs had 
severe functional disability. During the Korean 
and Vietnam Wars, when acute surgical repair 
replaced ligation for arterial trauma, the inci- 
dence of amputation following popliteal 



ch20.qxd 4/16/04 3:31PM Page 395 



20 • SPECIAL PROBLEMS 



395 



artery injury improved substantially to only 
29.5%, with fewer problems of morbidity and 
disability in salvaged limbs. As repair tech- 
niques and the use of surgical adjuncts have 
improved, there has been further substantial 
improvement in outcome since Vietnam fol- 
lowing repair of highly destructive combat 
injuries of this artery, with limb salvage now 
approaching 90% (Table 20-1). 

As this military experience with arterial 
repair was adopted in the civilian sector, the 
same improvements in limb salvage have been 
realized since the 1950s, despite the higher 
incidence of more destructive blunt trauma 
in this setting. Fabian and colleagues (1982) 
reported 165 civilian popliteal artery injuries 
treated over 30 years, showing an improve- 
ment in amputation rates from 74% to 6% 
during this period. Daugherty and colleagues 
(1978) documented a reduction of amputa- 
tion rates from 54% to 9% among 24 civilian 
popliteal artery injuries over a 1 0-year period. 
Thomas and colleagues (1989) similarly 
showed a reduction in limb loss from 30% 
before 1980 to 15% after 1980 in their review 
of 610 cases of civilian popliteal artery injuries 
in 25 published series. During the 1980s, four 
published civilian series reported 78 cases of 
both penetrating and blunt popliteal artery 
injury without a single amputation. Although 
there has been a tendency toward improve- 
ment in limb salvage as time has progressed, 
continued reports of high rates of limb loss 
even in recent years emphasize how danger- 
ous these injuries remain (Table 20-2) . 



Diagnostic Issues 

GENERAL PRINCIPLES AND 
MODALITIES 

The time interval from popliteal artery injury 
to repair is the most important factor in limb 
salvage. Virtually all reports document that 
the most common reason for limb loss in 
this setting is a delay in recognition and revas- 
cularization. This is because of the time- 
dependent nature of the major consequences 
of vascular injury, tissue ischemia, and hem- 
orrhage, to which the popliteal circulation is 
especially vulnerable. Therefore, a prompt 
and accurate diagnosis of popliteal artery 
trauma, within 6 hours of injury, is an extremely 
important factor influencing outcome. 

Diagnosis can be made in most cases by phys- 
ical examination, as long as the significance 
of the clinical manifestations of popliteal 
artery injury is understood. Obvious physical 
findings of arterial injury, also known as hard 
signs, are present in 70% to 90% of these cases, 
including active hemorrhage, large, expand- 
ing, or pulsatile hematoma, bruit or thrill, 
absent distal pulses, and distal ischemia (pain, 
pallor, paralysis, paresthesias, and coolness) . 
These findings must never be ignored. In the 
setting of uncomplicated penetrating trauma 
to the lower extremity, any of these signs 
mandate immediate surgery, because the 
probability of a major arterial injury requir- 
ing repair approaches 100%, and the 
penetrating wounds clearly show where that 



TABLE 20-1 

MILITARY EXPERIENCE WITH POPLITEAL ARTERY INJURIES* 



Author 



Year 



Conflict 



No. Cases 



No. Amputations (%) 



Makins 


1922 


WWI 


144 


62 (43) 


DeBakey, Simeone 


1946 


WWII 


502 


364 (72.5) 


Hughes 


1958 


Korea 


68 


22 (32.4) 


Rich 


1970 


Vietnam 


217 


64 (29.5) 


D'Sa 


1980 


Ireland 


32 


4(12.5) 


Sfeir 


1992 


Lebanon 


118 


14(12) 


Total 






1081 


530 (49) 



*AII injuries were ligated in WWI and WWII and were repaired in all remaining series. WWI, World War I; WWII, World War II. 



ch20.qxd 4/16/04 3:31PM Page 396 



396 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



TABLE 20-2 














MANAGEMENT RESULTS OF CIVILIAN POPLITEAL ARTERY INJURIES 




Author 


Year 


No. Cases 


No. Penetrating 


No. Blunt 


No. 


Amputations (%) 


Conkle 


1975 


27 


13 


14 




12 (44) 


Daugherty 


1978 


24 


11 


13 




8(33) 


O'Reilly 


1978 


49 


49 







6(12) 


Lim 


1980 


31 


19 


12 







Holleman 


1981 


32 


18 


14 




4(12.5) 


Fabian 


1982 


165 


125 


40 




44 (27) 


Jaggers 


1982 


61 


49 


12 




9(15) 


Snyder 


1982 


110 


81 


29 




14(13) 


McCabe 


1983 


24 


5 


19 




4(17) 


Orcutt 


1983 


37 


20 


17 




6(16) 


Yeager 


1984 


10 


5 


5 







Shah 


1985 


30 





30 







Downs 


1986 


63 


10 


53 




18(29) 


Krige 


1987 


28 


14 


14 




3(11) 


Weimann 


1987 


36 


11 


25 




1 (3.6) 


Armstrong 


1988 


76 


60 


16 




9(12) 


Peck 


1990 


108 


32 


76 




13(12) 


Reed 


1990 


7 


4 


3 







Martin 


1994 


40 


26 


14 




6(15) 


DeGiannis 


1995 


35 


35 







5(14) 


Fainzilber 


1995 


81 


63 


18 




13(16) 


Pretre 


1996 


31 





31 




6(19) 


Harrell 


1997 


38 





38 




14(37) 


Melton 


1997 


102 


62 


40 




25 (25) 


Razuk 


1998 


25 


15 


10 




6(24) 


Total 




1270 


728 (57%) 


543 (43%) 




226(18) 



injury is located. Any further diagnostic tests 
would be superfluous, unnecessarily costly, 
and potentially dangerous in view of the 
adverse impact of the inevitable delay on 
outcome. Exceptions to this include any cir- 
cumstance in which the physical examination 
does not clearly reflect the presence or loca- 
tion of arterial injury (e.g., blunt trauma, 
elderly patient with chronic vascular insuffi- 
ciency, associated skeletal trauma, shotgun 
wounds, thoracic outlet wounds, and estab- 
lished complications of delay), in which case 
arteriographic imaging is warranted. 

Arteriography is now known to be unnec- 
essary in injured extremities that do not 
manifest any hard signs of popliteal artery 
injury, regardless of mechanism or wound 
complexity. In the past, all asymptomatic 
wounds (i.e., those with no hard signs) placing 
the popliteal artery at risk either were surgi- 
cally explored or underwent routine arteri- 



ography. These included penetrating injuries 
in proximity to the artery and all high-risk blunt 
trauma, such as lower extremity crush, distal 
femur or proximal tibia fractures, and poste- 
rior knee dislocations. It has long been known 
that in this setting, occult vascular injury may 
still be present in 10% to 15% of cases. 
However, recent studies have shown that such 
asymptomatic vascular injuries are consis- 
tently nonocclusive and have a benign and self- 
limited natural history with a high rate of 
spontaneous resolution. They, therefore, 
require neither surgical repair, nor the con- 
siderable expense and resources necessary 
for routine detection. Virtually all limb- 
threatening complications of delayed diagnosis 
of popliteal artery injury are due to overlooked 
hard signs, rather than an absence of relevant 
physical findings, on initial presentation. 

Noninvasive testing with Doppler pressure 
measurements and duplex ultrasonography 



ch20.qxd 4/16/04 3:31PM Page 397 



20 • SPECIAL PROBLEMS 



397 



has been applied to the evaluation of injured 
extremities for popliteal artery trauma, with 
the potential of being less invasive and less 
costly than, while equally accurate, as arteri- 
ography and surgery. However, these modal- 
ities have not realized their theoretical 
potential for a number of reasons, including 
equipment expense, lack of round-the-clock 
availability of the necessary skill and exper- 
tise in most hospitals, and a failure to show 
any advantage over physical examination 
alone. Several authors have documented 
these modalities to have no benefit in this 
setting (Bergstein et al, 1992; Tominaga et al, 
1996). 



Posterior Knee Dislocation 

Posterior knee dislocation has been associated 
with a substantial incidence of popliteal artery 
trauma, for which reason mandatory arteri- 
ography or popliteal exploration has been 
advocated to avoid the high risk of limb loss 
from delayed diagnosis of popliteal artery 
injury. At least six published studies from the 
past decade, which report 264 cases of poste- 
rior knee dislocation, have related the initial 
clinical findings to outcome (Table 20-3) . The 



results demonstrate that only 23% (range, 
13% to 25%) of all cases present with hard 
signs of popliteal artery injury, and 70% of 
this group (range, 18% to 100%) had arter- 
ial injury requiring surgical repair (i.e., 30% 
false-positive rate of physical examination for 
the detection of surgically significant arterial 
injury) . Among the 77% of all cases of pos- 
terior knee dislocation presenting without 
hard signs, there was not a single popliteal 
artery injury that required surgical repair, a 
result confirmed by follow-up studies of up to 
1 year. These findings are consistent with those 
of all other forms of extremity injury in con- 
firming the reliability of physical examination 
to exclude arterial injury. This has enormous 
economic implications when considering the 
expense and morbidity of routine diagnostic 
workup that can be avoided in such a large 
majority of patients, especially considering 
how tight resources are in so many trauma 
centers and other hospitals. Surgical explo- 
ration of the popliteal artery is warranted in 
the minority of patients presenting with hard 
signs, especially when obviously due to arter- 
ial disruption (e.g., pulsatile hemorrhage and 
cool ischemic limb without pulses) . However, 
preoperative arteriography in less obvious 
cases of hard signs (e.g., transient pulse loss 



TABLE 20-3 

PUBLISHED CASES OF KNEE DISLOCATION RELATING PHYSICAL FINDINGS OF 
VASCULAR INJURY TO OUTCOME 







Hard Signs 


Present 




Hard Signs 


Absent 


Author 


Year 


No. KD 


No. 


(%)* 


No. AIRS O^ 


No. 


(%)* 


No. AIRS 


Treiman 


1992 


115 


29 


(25) 


22 (75) 


86 


(75) 


0* 


Kendall 


1993 


37 


6 


(16) 


6(100) 


31 


(84) 





Kaufman 


1992 


19 


4 


(21) 


4(100) 


15 


(79) 


0« 


Dennis 


1993 


38 


2 


(13) 


2(100) 


36 


(87) 


0' 


Miranda 


2000 


32 


8 


(25) 


6(75) 


24 


(75) 





Martinez 


2001 


23 


11 


(48) 


2(18) 


12 


(52) 


0^ 


Total 




264 


60 


(23) 


42 (70) 


204 


(77) 






'Percentage of total knee dislocations. 

♦Percentage of all patients with hard signs. 

♦includes nine minimal arterial injuries with 6 months of average follow-up. 

includes two minimal arterial injuries with 3 and 23 months of follow-up. 

"Includes seven minimal arterial injuries with 1 1 .5 months of average follow-up. 

includes five minimal arterial injuries with 3 months of average follow-up. 

AIRS, arterial injuries requiring surgery; KD, knee dislocations. 



ch20.qxd 4/16/04 3:31PM Page 398 



398 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



and hematoma) may allow avoidance of 
unnecessary vascular exploration in about 
20% of cases by demonstrating an intact artery. 



Treatment 

GENERAL PRINCIPLES AND 
TECHNIQUES 

Prompt transport to the operating room and 
induction of general anesthesia are necessary 
once popliteal artery injury has been docu- 
mented. The patient should be supine and 
the injured leg should be prepared and 
draped into the operative field, as should 
one uninjured extremity in the event that 
autogenous vein must be harvested. The leg 
should be abducted and externally rotated 
with a support under the knee, to facilitate 
the standard longitudinal medial incision 
above the knee for optimal popliteal artery 
exposure. Retraction of the sartorius and semi- 
membranosus muscles posteriorly opens the 
popliteal space where the artery lies, with the 
vein and nerve medial and posterior to it. Divi- 
sion of the medial head of the gastrocnemius 
muscle and tendons of the semimembranosus, 
semitendinosus, and gracilis muscles allows 
exposure of the more distal tibioperoneal 
trunk. Although these structures may be 
repaired, this is not necessary for an excel- 
lent functional result. Arterial repair is per- 
formed by the standard techniques ofvascular 
surgery. 

Hemorrhage from the injured artery should 
be controlled by digital pressure until proxi- 
mal and distal control can be obtained with 
clamps or vessel loops. Obviously damaged 
portions of the artery should be debrided back 
to grossly normal vessel. Balloon catheter 
thrombectomy should be performed proxi- 
mally and distally, followed by distal injection 
of heparinized saline to retard any further 
thrombus formation. Systemic heparinization 
may be used if associated injuries permit. 

Lateral arteriorrhaphy may be performed 
with clean lacerations involving less than 30% 
of the arterial circumference, although 
only 10% 15% of popliteal artery injuries are 
amenable to this. Care must be taken to avoid 



stenosis and thrombosis, and vein patch angio- 
plasty may facilitate this. End-to-end anasto- 
mosis is preferred if it can be done without 
undue tension but is generally not possible if 
more than 2 cm of artery is lost. Geniculate 
collaterals should not be divided to achieve 
mobility because of the detrimental effect this 
may have on limb perfusion. In this setting, 
interposition grafting should be performed, 
preferably using reversed autogenous saphe- 
nous vein. Prosthetic grafts across the knee 
joint tend to have lower patency rates. Arter- 
ial anastomosis and repair are performed with 
a running monofilament nonabsorbable 
suture, achieving intimal coaptation. Surgical 
repair should not be considered complete 
until distal perfusion is clearly documented 
with palpable pulses in the feet. 



SURGICAL ADJUNCTS 

A number of measures are available in addi- 
tion to standard surgical repair of popliteal 
artery trauma, which should further improve 
outcome from these dangerous injuries. The 
liberal use of preoperative and intraoperative 
systemic and regional anticoagulation with 
intravenous heparin has been mentioned. 
Postoperative anticoagulation should almost 
never be necessary, because it cannot substi- 
tute for the meticulous and appropriate tech- 
nique, which is the most important factor in 
a successful surgical repair. Any failure of arte- 
rial repair should mandate exploration and 
revision. Completion arteriography is impor- 
tant to ensure patency and distal runoff, espe- 
cially if there is any doubt about the adequacy 
of revascularization. This can be done on the 
operating table by direct needle puncture of 
the vessel proximal to the injury and injec- 
tion of water-soluble contrast. 

Extra-anatomic bypass should be consid- 
ered when the native vessel bed is unsuitable 
for vascular repair, because of contamination, 
devitalized tissue, or lack of soft tissue cover- 
age. A prosthetic or autogenous vein inter- 
position is tunneled laterally through clean 
tissue planes from uninvolved proximal and 
distal portions of the artery, allowing open 
management of the wound without worry 
about the vessel. 



ch20.qxd 4/16/04 3:31PM Page 399 



20 • SPECIAL PROBLEMS 



399 



Fasciotomy to release excessive pressure 
within the major tissue compartments of the 
lower leg is a critical adjunct to popliteal artery 
repair, because injury to this artery poses 
a high risk of compartmental hypertension 
and tissue loss (see Chapter 27) . Early or pro- 
phylactic fasciotomy in this setting has been 
associated with improved limb salvage and 
function. 

Intraluminal shunting of injured popliteal 
arteries can be a useful adjunct in those cir- 
cumstances in which a delay is necessary for 
skeletal stabilization, soft tissue debridement 
or vein repair. This immediately restores per- 
fusion, allowing these other problems to 
be addressed deliberately without ongoing 
ischemia, before definitive arterial repair is 
performed. 



NONOPERATIVE OBSERVATION 

A select group of arterial injuries are nonoc- 
clusive and manifest no hard signs, and these 
have been shown to have a high rate of spon- 
taneous resolution or nonprogression when 
left untreated. These include intimal flaps, 
vessel narrowing, and small false aneurysms 
and arteriovenous fistulas, in which the artery 
and its runoff remain intact. When found 
on arteriography, the safety of nonoperative 
observation of these asymptomatic arterial 
injuries has been established by long-term 
follow-up averaging 10 years. This category of 
arterial injuries exclusively includes the 10% 
to 15% of arterial injuries known to occur in 
the setting of asymptomatic lower extremity 
trauma that places the popliteal artery at risk, 
such as penetrating proximity wounds and 
high-risk fractures and posterior knee dislo- 
cation. This is what justifies the simple obser- 
vation of asymptomatic lower extremity 
trauma without the need for routine surgery 
or diagnostic imaging for popliteal artery 
injury (see Table 20-3). The safe avoidance 
of this routine diagnostic workup on the basis 
of only a negative physical examination has 
clear advantages in terms of substantial savings 
of cost and resource use, as well as reduced 
limb morbidity. 



MANAGEMENT OF VENOUS 
INJURIES 

History and Epidemiology 

The evolution of the management principles 
and techniques for venous injuries has 
followed the same path as that for arterial 
injuries, although an appreciation for the dis- 
tinct differences in venous response to injury 
and repair did not occur until relatively 
recently. The first successful surgical repair of 
a venous injury is credited to the German 
surgeon Schede in 1882, who reported the 
lateral suture of a femoral vein. Kummel per- 
formed the first successful end-to-end venous 
anastomosis in 1899, and Goodman reported 
four cases of lateral venorrhaphy in World War 
I. Ligation of all injured and uninjured veins 
adjacent to an arterial injury was advocated 
by Makins in World War I, to theoretically 
increase the "dwell time" of blood within the 
injured extremity after arterial ligation. 
Venous ligation remained standard practice 
through World War II and the Korea War, 
although 20 venous injuries underwent 
surgical repair in the latter conflict. In the 
Vietnam War, an aggressive approach toward 
routine repair of all venous injuries was advo- 
cated after reviewing the major complications 
resulting from venous ligation in the Vietnam 
Vascular Registry. The overall rate of venous 
repair rose to 33% in this conflict, and the 
clear benefits of repair were demonstrated. 
The fears of previous years that repair would 
lead to increased levels of thrombophlebitis 
and embolism were proven groundless and 
were shown to occur more often when veins 
were ligated. This military experience rapidly 
spread into the civilian sector. In 1960, Caspar 
and Treiman published the first large series 
of civilian venous injuries, which confirmed 
the problems with ligation and the safety 
and feasibility of routine repair of venous 
injuries. 

The actual incidence of peripheral venous 
injuries is unknown, because many are known 
to be asymptomatic, heal spontaneously, and 
are never discovered. Venous injuries made 
up 39% of all vascular injuries in the Korean 
War and 27% of all vascular injuries in the 



ch20.qxd 4/16/04 3:31PM Page 400 



400 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



Vietnam War. They were most often (86%) 
associated with an adjacent arterial injury in 
both conflicts. These were largely peripheral 
in location, because abdominal and cervical 
venous injuries are highly lethal in military 
settings, and virtually all were due to destruc- 
tive and high-velocity penetrating mecha- 
nisms. In the civilian sector, venous injuries 
similarly comprise between 13% and 51% of 
all vascular injuries, and 35% to 63% of all 
extremity vascular trauma. In this setting, they 
also are most commonly due to penetrating, 
though low-velocity, mechanisms and most 
commonly occur in association with an adja- 
cent arterial injury. Blunt mechanisms cause 
5% to 15% of all civilian venous injuries (Table 
20-4) . Isolated venous injuries are most likely 
to result from stab wounds than from other 
blunt or penetrating agents. Abdominal 
venous injuries comprise as much as 15% of 
all civilian vascular trauma. The superficial 
femoral vein and the popliteal vein are the 
most commonly injured veins overall in 
military and civilian series, respectively, 
consistent with the most common sites of 
arterial injury. The inferior vena cava is the 
most common site of abdominal venous 
injury. 



Diagnosis 

Peripheral venous injuries are most commonly 
found incidentally during exploration for an 



arterial injury. Although they may manifest 
hard signs, these signs are not specific for 
venous trauma. Bleeding from the low- 
pressure venous system is generally easily tam- 
ponaded by surrounding structures, which is 
why venous injuries often are not detected in 
the absence of arterial injuries. When isolated, 
venous injuries most commonly present 
as hemorrhage or large hematoma, which 
prompts the surgical exploration leading to 
their detection. Venous trauma is most likely 
to present with severe bleeding, shock, or 
hypotension when combined with an arterial 
injury or when involving one of the major 
abdominal veins. 

Routine venography following extremity 
trauma has been advocated. Gerlock and col- 
leagues (1976) performed this imaging in 30 
consecutive pati en ts with penetrating extrem- 
ity trauma, detecting five venous injuries 
(17%). However, four of these five injuries had 
associated arterial injuries that would have led 
to their detection without venography. Gagne 
and colleagues (1995) found venography of 
injured extremities to be suboptimal and to 
be difficult to perform, with more than 50% 
of attempts technically unsuccessful and only 
43% of all venous injuries detected. The only 
benefit of routine venography would be in 
its detection of previously unsuspected and 
asymptomatic venous injuries, but there is no 
evidence to support that such occult venous 
trauma results in any adverse sequelae when 
left untreated. In fact, occult venous trauma, 



TABLE 20- 


-4 












EXTREMITY VENOUS INJURIES: 


MECHANISM AND MANAGEMENT 




Author 




Year 


Total No. 


No. Penetrating 


No. Isolated 


No. Repaired 


Rich* 




1970 


377 


377 


53 


124 


Sullivan* 




1971 


26 


26 


8 


21 


Agarwal 




1982 


57 


53 


18 


34 


Phifer 




1984 


25 


25 





19 


Ross 




1985 


22 





1 


12 


Pasoh 




1986 


82 





4 


53 


Borman 




1987 


82 


71 


20 


74 


Meyer 




1987 


36 


34 


2 


36 


Ye Ion 




1992 


79 


78 


31 


31 


Timberlake 




1995 


322 


292 


83 


98 


Total 






1108 


956 (95%) f 


220 (20%) 


502 (45%) 



'Military series. 

+ Based on 1004 injuries that could be evaluated. 



ch20.qxd 4/16/04 3:31PM Page 401 



20 • SPECIAL PROBLEMS 



401 



even in the inferior vena cava, has been shown 
to have no long-term complications, suggest- 
ing that routine diagnostic imaging for this 
purpose is unnecessary and not cost effective. 
On the other hand, Gagne and colleagues 
(1995) found major thromboembolic com- 
plications to occur in 50% of patients with 
documented asymptomatic venous injuries 
following penetrating extremity trauma, 
although whether this was specifically related 
to the venous trauma is not clear. 

Duplex ultrasonography has been applied 
to the detection of arterial injuries following 
extremity trauma by several authors. Gagne 
and colleagues (1995) reported the detection 
of occult venous injuries in 22% of patients 
with asymptomatic penetrating extremity 
trauma, although the benefits of this detec- 
tion were not clear. 

Currently most trauma centers do not 
perform venous imaging of any sort follow- 
ing torso or extremity trauma. In cases pre- 
senting with hard signs, imaging is generally 
contraindicated, because immediate surgery 
is warranted. In the absence of hard signs, 
imaging is unnecessary because there is no 
clear benefit to intervention for occult venous 
injury. Only if subsequent symptoms develop, 
such as venous insufficiency or thromboem- 
bolic events, would venous imaging be 
justified. 



Unlike arterial trauma, minimal debride- 
ment of injured veins is generally necessary. 
Proximal and distal thrombectomy should 
be done by gentle milking and the judicious 
use of balloon catheters, to avoid valvular 
damage. In combined venoarterial extremity 
trauma, especially with obvious ischemic 
changes, intraluminal shunting of the artery 
restores distal perfusion immediately, allow- 
ing deliberate venous repair, as well as any 
necessary soft tissue or skeletal repairs, 
without the danger of ongoing ischemia and 
tissue loss. Another consideration in such 
combined vascular injuries is to shunt the 
vein while first repairing the artery, to provide 
adequate outflow for the artery during its 
repair and optimizing its success. The shunted 
vessel in either case then can be definitively 
repaired. 

A variety of techniques that correspond to 
the same techniques used for arterial injuries 
have been described for repair of venous 
injuries. Lateral venorrhaphy, for partial cir- 
cumferential lacerations, and end-to-end 
anastomosis, for transections with little loss of 
vessel length, are the most common methods, 
as well as the quickest and easiest to perform. 
Uncommonly used and more difficult and 
time consuming are interposition grafting with 
either reversed autogenous vein or prosthetic 
graft and the construction of spiral grafts or 
panel grafts from segments of autogenous 
vein. 



Treatment 

GENERAL PRINCIPLES 

Management of the patient with venous injury 
is essentially identical to that for any vascular 
injury. Prompt digital or manual control of 
any active external bleeding is followed by 
volume resuscitation through large-bore in- 
travenous catheters placed in uninjured 
areas. Longitudinal incisions directly over the 
injured vessels should be made. Digital control 
of the injury, sponge-stick compression, in- 
traluminal balloon catheters, or occluding 
clamps are methods that control venous 
bleedingfrom the area of injury while thevein 
is dissected free and formal proximal and distal 
control can be achieved using clamps or elastic 
vessel loops. 



VENOUS REPAIR CONSIDERATIONS 

The motivation for repair rather than ligation 
of venous injuries began with the observation 
of the sequelae of venous hypertension and 
venous insufficiency following ligation in the 
Korean War. Rich and colleagues (1976) con- 
firmed these observations during the Vietnam 
War, noting a substantially greater incidence 
of extremity edema, stasis dermatitis, ulcera- 
tion, and chronic venous insufficiency in limbs 
that underwent major venous ligation com- 
pared with those undergoing venous repair. 
This difference was especially true for the 
popliteal vein, which provides the major 
channel for venous drainage from the lower 
extremity. Several cases of limb amputation 
were attributed directly to ligation or failed 



ch20.qxd 4/16/04 3:31PM Page 402 



402 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



repair of popliteal vein injuries. An aggressive 
approach toward routine repair of all venous 
trauma, especially in the popliteal system, led 
to dramatic improvements in limb salvage and 
function and dispelled the fears of earlier 
years that repair would lead to problems with 
thrombophlebitis and venous thromboem- 
bolism. A long-term follow-up of 1 10 popliteal 
vein injuries from this conflict (Rich, 1982) 
demonstrated a reduction of significant limb 
edema from 51% in cases of vein ligation to 
only 1 3% in those undergoing vein repair. The 
safety and feasibility of venous injury repair 
had also been shown in the civilian sector by 
Caspar and Treiman (1960) and by experi- 
mental studies. Repair of the inferiorvena cava 
not only prevents lower limb edema but also 
restores venous return to the heart to offset 
hemorrhagic and cardiogenic shock. Pul- 
monary embolism has been reported in only 
2% of these cases. 

The natural history of surgically repaired 
venous injuries is characterized by a substan- 
tial rate of thrombosis in the postoperative 
period that far exceeds that seen following 
arterial repair. This was documented by Rich 
(1970) in the Vietnam War in isolated cases. 
In the civilian sector, Meyer and colleagues 
(1987) found that 39% of their peripheral 
venous repairs thrombosed within 1 week by 
venography, and that this thrombosis rate 
was higher in complex repairs (59%) than in 
simple repairs (21 % ) . Hobson and colleagues 
(1983) reported postoperative thrombosis in 
26% of femoral vein repairs, but a significantly 
higher rate of limb edema (75%) in these 
occluded repairs than in those remaining 
patent (23.5%). Agarwal and colleagues 
(1982) found postoperative thrombosis in 
80% of vein repairs followed by venography, 
and Nypaver and colleagues (1992) found this 
to occur in 28% of their venous repairs. 

Despite these findings, restoration ofvenous 
continuity appears to offer major advantages 
over ligation. Most series have reported few 
if any adverse sequelae in the limbs of patients 
with thrombosed venous repairs, indicating 
that venous repair allows the development of 
venous collaterals. Also, a number of long- 
term follow-up studies have shown a high 
rate of recanalization of thrombosed venous 
repairs, which cannot occur following ligation. 



Nypaver and colleagues (1992) documented 
that 88% of previously thrombosed vein 
repairs were subsequently patent by color-flow 
duplex sonography over a 49-month average 
follow-up, providing a 90% long-term patency 
rate. Phifer and colleagues (1985) showed 
100% patency of five femoral vein repairs over 
follow-ups ranging from 6 to 20 years. 

These results indicate that veins are 
extremely sensitive to injury and surgical 
manipulation, and that meticulous technique 
is essential to minimize postoperative throm- 
bosis. Gentle handling of venous endothe- 
lium, rigorous attention to intimal apposition, 
and precise suture techniques that restore the 
vein lumen to its normal diameter without 
stricture have all been recommended to opti- 
mize patency. The use of vein patch or inter- 
position grafting is suggested if simple repair 
narrows the venous lumen. Completion 
venography has been recommended to ensure 
widely patent surgical repairs, but few prac- 
tice this. Although prosthetic grafts for venous 
injury repair result in especially high rates 
of thrombosis, they have the advantage of 
allowing rapid restoration of continuity and 
reducing operative bleeding in large open 
wounds with extensive soft tissue disruption 
and at fasciotomy sites, by providing imme- 
diate venous drainage and avoiding venous 
hypertension. 

A number of adjuncts to venous injury 
repair have been reported to improve post- 
operative results. Temporary arteriovenous 
shunts or arteriovenous fistulas created distal 
to extremity venous repairs have improved 
patency rates but have the disadvantages 
of increased limb edema from the higher 
venous flow, reduction in arterial flow, and 
the need for a second operative procedure to 
take these down. Systemic anticoagulation and 
antiplatelet therapy has been applied to 
reduce postoperative thrombosis, but Hobson 
and colleagues (1973) showed no effect of 
these measures on improving postoperative 
patency rates. Intermittent pneumatic com- 
pression devices offer theoretical promise but 
have not been studied for any benefit in this 
setting. 



ch20.qxd 4/16/04 3:31PM Page 403 



20 • SPECIAL PROBLEMS 



403 



VENOUS LIGATION CONSIDERATIONS 

Clinical experience with the morbid seque- 
lae of the standard practice of ligation of 
venous injuries during the Vietnam War 
called this practice into question. Several pub- 
lished experimental studies by Stallworth and 
colleagues (1967), Barcia and colleagues 
(1972), Hobson and colleagues (1973), and 
Wright and colleagues (1973, 1974) docu- 
mented that acute femoral venous occlusion 
in uninjured canine limbs resulted in signif- 
icant reductions in arterial inflow. This 
reduced flow returned to baseline within 72 
hours, suggesting venous collateral develop- 
ment, but the jeopardy this poses to an 
injured limb, especially in the presence of a 
fresh arterial repair, is obvious. Clinical inves- 
tigations from military and civilian settings 
confirmed these findings in showing increased 
limb morbidity and limb loss following venous 
ligation, especially in the popliteal system. 

Since the Vietnam War, the enthusiasm for 
routine repair of venous injuries that these 
studies fostered has been tempered by several 
civilian studies showing remarkably uncom- 
plicated outcomes following ligation of major 
veins in injured extremities. Muffins, Lucas, 
and Ledgerwood (1980) showed the absence 
of any clinically significant short- or long-term 
impairment of limb function in 46 patients 
undergoing ligation of injured major veins, 
70% of which were in the lower extremity. 
Eight of these patients had moderate edema 
requiring support stockings, but no limitation 
of activity. Several other authors have since 
documented acceptable levels of limb mor- 
bidity following lower extremity vein ligation, 
including inferior vena cava and iliac veins, 
though with varying levels of postoperative 
edema in up to 50% of patients. There have 
been no instances of limb loss attributable to 
venous ligation in these reports. Studies by 
Meyer and colleagues (1987), Pasch and col- 
leagues (1986), Yelon and Scalea (1992), and 
Timberlake and Kerstein (1995) report a total 
of 440 patients with major peripheral venous 
injury, including the popliteal and femoral 
veins, with no limb loss and no significant long- 
term edema following vein ligation, as well as 
no difference in outcome between patients 
undergoing vein ligation and those under- 



going vein repair. Ligation of major venous 
injury is consistently tolerated without prob- 
lems in the upper extremities and neck, attrib- 
utable to the greater collateral drainage than 
that found in the lower extremities. 

Recent studies of inferior vena cava and iliac 
vein injuries show similar results for repair and 
ligation, although these tend to be more 
severely compromised patients because of 
the shock and blood loss accompanying 
these injuries. Burch and colleagues (1990) 
reported 161 iliac vein injuries, with a higher 
rate of venous morbidity (26%) among cases 
that were ligated (consisting only of limb 
edema and deep vein thrombosis) than in 
those repaired (4.9%, two cases edema and 
two of pulmonary embolism) , but without any 
limb loss. 

Successful outcome of limb salvage and 
function following venous ligation demands 
an aggressive use of several adjunctive mea- 
sures to promote venous drainage. These 
patients should be placed on bed rest, with 
avoidance of dependent positioning of their 
limbs. Lower extremities should be wrapped 
in elastic bandages and elevated for several 
days before carefully beginning ambulation. 
Four-compartment lower leg fasciotomy 
should be applied liberally and prophylacti- 
cally, to offset the very high probability of 
development of compartment syndrome. If 
fasciotomy is not performed, careful serial 
measurement of compartment pressures is 
necessary. Routine anticoagulation has no 
proven benefit. 



ROLES OF LIGATION AND REPAIR 

Although the published evidence appears con- 
tradictory in many ways regarding the rela- 
tive merits of ligation or repair of venous 
injuries, each can clearly be successful under 
certain conditions (see Table 20-4) . Which 
approach is best in any setting requires that 
the reported differences in outcome be under- 
stood and reconciled. Venous ligation results 
in the worst outcome in military series and 
experimental models, which can be explained 
by the greater collateral damage of bone and 
soft tissue, as well as the total venous outflow 
occlusion, which occurs in these settings. In 



ch20.qxd 4/16/04 3:31PM Page 404 



404 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



actual clinical venous injury in the civilian 
setting, the better results of ligation can be 
explained by the simpler, less destructive 
wounds without major associated trauma and 
by the fact that ligation of any single vein does 
not totally occlude limb outflow. 

Most authorities agree that all injuries to 
major veins should undergo surgical repair, 
unless there are more pressing priorities, such 
as life-threatening problems, which preclude 
it. Hemodynamic instability, ongoing hem- 
orrhage, and life-threatening associated 
injuries to other body systems that require 
immediate attention are the most common 
indications to revert to ligation of venous 
injuries. This is a basic tenet of the damage 
control approach to severely injured patients, 
because venous repair always requires more 
time to accomplish than ligation, and complex 
repairs take more time than simple repairs. 
Some authors recommend that ligation be 
performed if repair requires anything more 
complex than lateral suture or end-to-end 
anastomosis, in view of the added time and 
worse outcome of complex repairs. Deaths 
have been attributed to ill-considered attempts 
at complex venous repairs. Ligation of all 
upper extremity veins and unilateral internal 
jugular veins can be performed without a 
problem. Patients generally tolerate ligation 
of any vein in the body as long as adjunctive 
measures are properly applied. Also, ligated 
veins can be reversed by elective definitive 
repair if necessary in those uncommon 
instances that severe complications develop 
in the future. 

It is important to recognize those circum- 
stances in which venous repair should be 
attempted even under suboptimal conditions 
of patient instability or complex trauma. In 
patients with injuries involving massive tissue 
destruction, from blunt or high-velocity pen- 
etrating trauma, repair of a major vein may 
be critical in providing limb outflow, even 
if that repair must be complex or requires a 
prosthetic graft. In combined arteriovenous 
trauma in the same extremity, venous repair 
should be undertaken to provide outflow for 
the arterial repair and optimize its success. 
Any popliteal vein injury merits all possible 
effort to repair rather than ligate due to the 
known severe and limb-threatening morbid- 



ity that is associated with ligation. Bilateral 
internal jugular vein injuries in the neck 
warrant repair of one side to provide critical 
intracranial venous outflow. In critical cir- 
cumstances, temporary intraluminal shunts 
may be used as a damage-control measure to 
quickly restore venous outflow, allowing defin- 
itive repair to be done later when conditions 
stabilize. 

Some authors advocate routine follow-up 
imaging of repaired venous injuries with 
venography or duplex scanning, to detect 
thrombosis early. However, an understanding 
of the natural history of venous repairs makes 
clear why this practice is unnecessary and is 
not used by most centers. The known high 
incidence of postoperative thrombosis is 
typically followed by spontaneous recanaliza- 
tion to provide a very high long-term patency. 
Also, even thrombosed repairs manifest very 
few clinical problems. Imaging should be 
applied only to those few patients in whom 
symptoms of disabling edema and chronic 
venous insufficiency develop, in which case 
anticoagulation or surgical revision may be 
considered. 



COMBINED VASCULAR 
AND SKELETAL 
EXTREMITY TRAUMA 



Extremity trauma that involves both skeletal 
and vascular injuries poses one of the 
most difficult management problems. These 
complex injuries often involve extensive soft 
tissue and nerve damage as well and are some- 
times termed mangled extremities. It is impor- 
tant to understand the unique considerations 
of epidemiology, pathophysiology, prognosis, 
diagnosis, and multidisciplinary priorities in 
this setting, to reduce the substantial risks of 
limb loss that currently prevail. 

Epidemiology and 
Prognostic Factors 

Combined vascular and skeletal extremity 
injuries are relatively uncommon, making up 
only 0.2% of all military or civilian trauma. 



ch20.qxd 4/16/04 3:31PM Page 405 



20 • SPECIAL PROBLEMS 



405 



Vascular and trauma surgeons are more likely 
to see this combined trauma than orthopedic 
surgeons, because only 1.5% to 6.5% of all 
extremity skeletal traumas are associated with 
an arterial injury of the same extremity, 
whereas 10% to 73% of all extremity arterial 
injuries may be associated with skeletal frac- 
tures and dislocations. 

Combined vascular and skeletal extremity 
injuries pose a substantially increased risk of 
amputation and limb morbidity than isolated 
arterial or skeletal injuries. This was docu- 
mented in several military series dating back 
to World War II. These reports showed that 
combined injuries were associated with ampu- 
tation rates ranging up to ten times those from 
isolated arterial extremity injuries, even as the 
outcomes for both simple arterial and com- 



bined combat extremity injuries improved 
over the past 50years (Table 20-5) .McNamara 
and colleagues (1973) also showed a signifi- 
cantly higher incidence of failed vascular 
repair among combined extremity injuries 
(33%) than among isolated arterial injuries 
of the extremity in the Vietnam War (5%). 
Similar striking differences have been re- 
ported in the civilian sector even in recent 
years. At a time when isolated arterial or skele- 
tal extremity trauma can be expected to result 
in limb loss in far less than 5 % of civilian cases, 
the combination of these injuries in the same 
extremity still are associated with amputation 
rates up to 68% in the most experienced 
trauma centers (Table 20-6). Even many sal- 
vaged extremities are significantly disabled. 
These results are largely due to delayed recog- 



TABLE 20-5 

AMPUTATION RATES (%) FOLLOWING COMBAT EXTREMITY ARTERIAL INJURIES 
WITH AND WITHOUT ASSOCIATED SKELETAL TRAUMA 



Author 


Year 


Conflict 


DeBakey 


1946 


World War 


Spencer 


1955 


Korea 


McNamara 


1973 


Vietnam 


Romanoff 


1979 


srae 


Lovric 


1994 


Croatia 



Isolated Arterial Injury 

42 
15 
2.5 
11 





Combined Injury 

60 
55 
23 

36 
10 



TABLE 20-6 

MECHANISM AND OUTCOME OF COMBINED CIVILIAN VASCULAR/SKELETAL 
EXTREMITY TRAUMA 



Author 

Schlickwei 

Van Wijngarden 

Alexander 

Johansen 

Odland 

Lange 

Palazzo 

Howe 

Bongard 

Drost 

Bishara 

Attebery 



Year 

1992 
1993 
1991 
1990 
1990 
1985 
1986 
1987 
1989 
1989 
1986 
1996 



% Penetrating 





9 
<10 
10 
13 
18 
24 
30 
36 
57 
71 



% Amputation 


45 


41.5 


28 


68 


35 


61 


7(0) 


43 


18 


29 


3(0) 


7(0) 



'Numbers in parentheses refer to amputations among penetrating injuries only. 



ch20.qxd 4/16/04 3:31PM Page 406 



406 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



nition of vascular injury, major nerve damage, 
and increased failure of vascular of repair due 
to disruption of collaterals from soft tissue 
injury, soft tissue infection from inadequate 
debridement, failure to provide adequate 
tissue coverage over sutured vessels, and 
delayed recognition and treatment of com- 
partmental hypertension. 

Mechanism of injury is another important 
determinant of outcome in this setting. Vir- 
tually all combined extremity injuries in mil- 
itary series are due to destructive high-velocity 
penetrating mechanisms. These approximate 
the level of tissue damage found in most blunt 
trauma, which has been the predominant 
mechanism for this distinct category of 
extremity trauma in most civilian series. 
However, an increasing incidence of simple 
penetrating trauma has been reported as a 
cause of these injuries in the civilian sector 
over the past decade, ranging from 18% to 
71 % of cases. Some correlation is also evident 
between the extent of penetrating trauma and 
an improved outcome in these series (see 
Table 20-6). Among 119 patients with com- 
bined vascular and skeletal extremity injuries 
reported in three of these series (Bishara and 
colleagues, 1986; Palazzo, 1986; Attebery 
and colleagues, 1996), 63 cases (53%) were 
due to penetrating trauma, and the total 
amputation rate was the lowest ever reported 
(well below 10%), with no amputations among 
any of the cases due to penetrating injury. 

It is generally accepted that penetrating 
trauma has a better outcome than blunt 



trauma, because of less severe and extensive 
associated tissue damage. Therefore, it should 
be no surprise that the increasing trend in 
low-velocity penetration as an etiology for com- 
bined vascular and skeletal extremity trauma 
in the civilian sector appears responsible for 
the most substantial contribution yet to reduc- 
ing limb loss from these devastating injuries. 
Five civilian series of combined extremity 
trauma have been published since 1986 in 
which more than 50% of cases were due to 
penetrating mechanisms (Table 20-7). The 
overall rate of penetration among all 228 
reported cases was 64.5%. The overall ampu- 
tation rate was 17%, but among penetrating 
injuries, itwas only 6%. Only 23% of all ampu- 
tations were in patients with penetrating 
trauma. Among 88 patients with penetrating 
combined injuries reported in three of these 
series, remarkably there were no amputations. 
Although some published series of combined 
extremity trauma report no difference in 
outcome between blunt and penetrating 
mechanisms, and some report a higher ampu- 
tation rate among penetrating injuries, these 
series involve small numbers and particularly 
severe injuries. Nonetheless, they demonstrate 
that a number of variables other than mech- 
anism affect outcome. 



Diagnosis 

Prompt and accurate diagnosis of vascular 
injury is critically important in the setting of 



TABLE 20-7 

OUTCOME OF PREDOMINANTLY PENETRATING COMBINED CIVILIAN 
VASCULAR/SKELETAL EXTREMITY TRAUMA 











No. 


Amputations (%) 


Author 


Year 


No. Patients 


No. Penetrating (%) 


Total 


Penetrating 


Norman 


1995 


30 


30(100) 








Attebery 


1996 


41 


29(71) 


3(7) 





Swetnam 


1986 


36 


24 (67) 


16(44) 


8(33) 


Bishara 


1986 


51 


29 (57) 


1(2) 





Russell 


1991 


70 


35 (50) 


19(27) 


1(3) 


Total 




228 


147(64.5) 


39(17) 


9(6) 



ch20.qxd 4/16/04 3:31PM Page 407 



20 • SPECIAL PROBLEMS 



407 



an extremity fracture or dislocation, because 
prolonged ischemia and delay in restoration 
of blood flow are cited in most studies as major 
contributors to limb loss and limb morbidity. 
The clinical presentation of the patient and 
the physical examination are the key elements, 
and in many cases the only elements, neces- 
sary for diagnosis or exclusion of vascular 
injury in this setting. Contrast-enhanced arte- 
riography is the standard imaging modality 
used to make this diagnosis, although its exact 
role is debated. 

The presence of hard signs (hemorrhage, 
hematoma, bruit or thrill, absent pulse, distal 
ischemia) in uncomplicated extremity trauma 
has been shown to predict major vascular 
injury with an accuracy approaching 100% 
and therefore generally mandates immediate 
operation without any imaging. However, 
combined skeletal and soft tissue disruption 
substantially reduces the positive predictive 
value (i.e., hard signs present) of physical 
examination for surgically significant vascu- 
lar injury. Applebaum and colleagues (1990) 
reported 53 cases of complex blunt extrem- 
ity trauma, documenting vascular injuries in 
39% of all cases manifesting hard signs, but 
in only 13% of cases did these injuries require 
surgical repair. This represented an 87% false- 
positive rate of physical examination for the 
detection of surgically significant vascular 
injury, the only ones to require detection and 
treatment, similar to that reported in other 
series. This can be explained by the fact that 
fractures, soft tissue disruption, traction, and 
distortion of arteries by bony fragments, 
nerve injuries, and compartment syndrome, 
all common features of complex limb trauma, 
can cause hard signs in the absence of injury 
to a major artery. Arteriography is recom- 
mended in complex extremity trauma that 
manifests hard signs, to exclude an arterial 
injury to avoid as much as an 87% rate of 
unnecessary vascular exploration in these 
already compromised limbs (Figs. 20-1 and 
20-2). 

On the other hand, arterial imaging does 
not appear necessary in complex extremity 
trauma without hard signs. Current evidence 
indicates that a negative physical examination 
result (i.e., no hard signs) excludes surgically 
significant arterial injury as reliably as both 




■ FIGURE 20-1 

Arteriogram performed for displaced 
supracondylar femur fracture manifesting 
hematoma and uncertain distal pulses, 
confirming intact popliteal artery and runoff and 
sparing unnecessary vascular exploration. ■ 



arteriography and surgical exploration. 
Nonocclusive vascular injuries may still occur 
but are known to have a benign natural history 
and therefore do not require detection. The 
economic advantages of avoiding routine 
arteriography in this group of patients are 
obvious, as discussed in previous sections of 
this chapter (see "Popliteal Artery Injuries" 
and "Posterior Knee Dislocation," earlier in 
this chapter) . 

When indicated for complex extremity 
trauma, arteriography is best performed as a 
percutaneous hand-injected study right in the 
trauma center or on the operating table with 
the extremity already prepared and draped 
(see Fig. 20-2) . This technique is easily per- 
formed by any surgeon, using a simple intra- 
venous catheter slipped over a needle directly 
into the femoral artery at the groin or brachial 
artery above an occluding blood pressure cuff, 
with a radiographic plate placed under the 
injured extremity, and injecting 30 mL of 
water-soluble contrast while the x-ray film is 
exposed by a portable unit. Substantial time 
is saved by this procedure, as formal arteri- 
ography in the radiology suite takes at least 1 
to 3 hours even in major trauma centers. This 



ch20.qxd 4/16/04 3:31PM Page 408 



408 



V • SPECIAL PROBLEMS AND COMPLICATIONS 




■ FIGURE 20-2 

Percutaneous one-shot hand-injected 
arteriogram on operating table following 
comminuted tibial plateau fracture with 
hematoma and poorly palpable distal pulses, 
showing occluded anterior tibial artery, 
narrowed tibioperoneal trunk, and intact 
posterior tibial artery with nonocclusive intimal 
defect (arrow). No vascular repair was 
necessary, skeletal repair was immediately 
performed, and distal perfusion remained intact 
without subsequent problems. ■ 



is critical to optimize limb salvage. On-table 
arteriography is also highly accurate, provides 
excellent resolution, and allows immediate 
treatment to begin on either the vascular or 
the skeletal injury, depending on the arteri- 
ogram results. 

Although noninvasive vascular studies with 
Doppler pressure monitoring and duplex 
ultrasonography have been applied to the eval- 
uation of complex extremity trauma for 
vascular injury, there is as yet no clear role for 
these modalities. In fact, the expertise 
required for interpretation of these tests, their 
lack of round-the-clock availability in most 



hospitals, and the significant swelling, skele- 
tal disruption, and bulky splints and dressings, 
which characterize these extremities, cast 
doubt on the utility and accuracy of nonin- 
vasive tests in this setting. Any use of these 
tests for diagnosis or exclusion of vascular 
trauma should be within the context of a con- 
trolled study, and their results should be inter- 
preted with caution. Arteriography remains 
the standard modality of choice for evalua- 
tion of high-risk complex extremity trauma 
for vascular injury. 



Treatment 

Combined vascular and skeletal trauma 
requires a multidisciplinary approach to treat- 
ment, which can succeed only with the smooth 
and coordinated interaction between the 
various specialties involved in caring for the 
skeletal, soft tissue, and vascular injuries, 
as well as for the patient as a whole. All life- 
threatening injuries must be treated as a first 
priority. Once the extremity is addressed, 
orthopedic surgeons and plastic surgeons 
should be involved integrally in the treatment 
decisions, along with the trauma or vascular 
surgeon, as soon as a diagnosis of combined 
extremity trauma is established. 

Prompt restoration of blood flow within 6 
hours of any extremity vascular injury is the 
most critical of the many factors that deter- 
mine limb salvage and function. Clinical and 
experimental studies consistently demon- 
strate a direct linear relationship between the 
time interval to extremity reperfusion and the 
amputation rate. Associated skeletal and soft 
tissue injury makes this time factor even more 
important. A small number of retrospective 
studies fail to show a correlation between time 
delay and outcome, but this again emphasizes 
that multiple variables are at play in these dev- 
astating extremity injuries. The weight of evi- 
dence mandates that rapid diagnosis, based 
on the clinical manifestations and selective 
application of one-shot on-table arteriography, 
be followed as expeditiously as possible by 
restoration of blood flow. 

Prioritization of management of the vas- 
cular and skeletal extremity injuries has been 
subject to debate and uncertainty, leading to 



ch20.qxd 4/16/04 3:31PM Page 409 



20 • SPECIAL PROBLEMS 



409 



wide variations in practice. Early studies rec- 
ommended that skeletal repair take priority 
in combined extremity trauma, to avoid the 
potential disruption of an initial fresh vascu- 
lar anastomosis by subsequent manipulation 
of bone fragments or length discrepancies in 
any initial vascular repair caused by subse- 
quent stabilization of comminuted, unstable 
skeletal injuries. It was believed that some delay 
in reperfusion is acceptable in the absence of 
overt ischemia. However, these conjectures 
have been refuted by much published evi- 
dence. Substantial tissue damage can occur 
in the absence of signs of ischemia, as has been 
made clear by our understanding of com- 
partment syndrome. Disruption of an initial 
vascular repair by subsequent skeletal manip- 
ulation occurs only rarely. Snyder and col- 
leagues (1982) reported this to occur in only 
7% of cases, and Downs and colleagues (1986) 
reported this in only 2% of all cases of com- 
bined extremity trauma undergoing initial vas- 
cular repair, and in each case, the repair was 
immediately revised with no effect on limb 
salvage. Howe and colleagues (1987) reported 
no vascular disruption in 21 such cases. 
Jahnke and Seeley (1953) generally per- 
formed initial vascular repairs in combined 
extremity injuries in the Korean War without 
adverse sequelae. Also, the resistance of 
repaired vessels to disruption has been under- 
estimated. Connolly and colleagues (1969) 
demonstrated that the strength of fresh 
anastomoses of transected canine femoral 
arteries with associated femur fractures 
approximated that of native vessels, resisting 
disruption by either 30 pounds of traction or 
by bone fragment impalement. These data 
are further supported by clinical studies 
(Romanoff, 1979; McCabe, 1983), which 
demonstrate a substantially higher rate of limb 
salvage among combined extremity injuries 
in which vascular repair is undertaken first, 
compared with those in which revasculariza- 
tion is delayed until skeletal stabilization/ 
repair is completed. Most importantly, no dis- 
advantage has ever been documented for 
initial revascularization, whereas the dangers 
of delay are well established. Any delay in revas- 
cularization must be considered a gamble. Of 
course, this makes eminent sense, because 
perfusion, rather than immediate skeletal 



continuity, is the sine qua wow of limb survival. 
Furthermore, as mentioned earlier, published 
studies consistently report that the most 
common reason for limb loss in this setting is 
delay or failure of revascularization, not of 
skeletal repair. 

It must be emphasized that restoration of 
extremity perfusion does not always require 
definitive vascular repair. Temporary plastic 
or Silastic intraluminal shunts placed in 
the severed ends of vessels following distal 
thrombectomy can restore distal perfusion 
within minutes. In fact, a formal vascular repair 
should be avoided in the setting of unstable 
and severely comminuted fractures and dis- 
locations, segmental bone loss, or severe soft 
tissue disruption and contamination. Shunt- 
ing in these cases allows deliberate attention 
to wide debridement and skeletal stabilization 
and fixation without ongoing ischemia, after 
which definitive vascular repair can be per- 
formed (Fig. 20-3). This avoids major stress 
on a vascular suture line from bone manipu- 
lation or undue tension or slack on the 
repaired vessel when the limb is fixed at its 
proper length. Alternatively, initial revascu- 
larization can and should be accomplished by 
immediate definitive arterial repair in the 
setting of stable skeletal injuries in which 
minimal subsequent manipulation and length 
discrepancy is anticipated (Fig. 20-4) .The con- 
sensus in the literature now strongly favors 
limb revascularization as the immediate man- 
agement priority in all combined extremity 
trauma, as the aforementioned considerations 
render moot any possible disadvantages. How 
the revascularization is accomplished is a 
matter of judgment, which depends primar- 
ily on the nature of the skeletal and soft tissue 
injuries. 

Current evidence suggests that asympto- 
matic nonocclusive arterial injuries found on 
extremity arteriography are safe to observe 
nonoperatively in the setting of combined 
extremity trauma, where they have been 
shown to have the same benign natural history 
as in uncomplicated penetrating extremity 
and neck trauma. Three published series 
(Applebaum, 1990; Norman, 1995; Attebery 
and colleagues, 1996) report a total of 98 
asymptomatic arterial injuries in extremities 
with associated skeletal trauma from both 



ch20.qxd 4/16/04 3:31PM Page 410 



410 V • SPECIAL PROBLEMS AND COMPLICATIONS 





* ■ . . 


« 9 




BUn 




■ FIGURE 20-3 

/A, Unstable elbow dislocation with brachial artery avulsion and loss of overlying skin. Distal 
perfusion immediately restored by intraluminal brachial artery shunt, allowing cross-joint external 
fixation and soft tissue debridement. 6, Artery was then repaired with autogenous vein graft, and a 
pedicle flap provided immediate coverage. ■ 



ch20.qxd 4/16/04 3:31PM Page 411 



20 • SPECIAL PROBLEMS 



411 




■ FIGURE 20-4 

A, Midshaft femur fracture with superficial femoral 
artery thrombosis manifesting distal pulse deficit and 
ischemia. B, Completion arteriogram showing vascular 
injury repair by resection and autogenous saphenous 
vein graft interposition (arrow) before the skeletal 
injury was addressed. C, Intramedullary rod fixation of 
femur fracture was then performed without vascular 
complication. ■ 



ch20.qxd 4/16/04 3:31PM Page 412 



412 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



blunt and penetrating mechanisms, of which 
only one (1%) underwent surgical repair, and 
that was a nonocclusive intimal flap of the 
distal radial artery, which was probably unnec- 
essary. There was no limb loss, limb morbid- 
ity, or subsequent vascular problems. Attebery 
and colleagues (1996) followed 15 asympto- 
matic nonocclusive arterial injuries (35% of 
all 41 vascular injuries associated with extrem- 
ity skeletal trauma) for a mean interval of 6.5 
months, and none ever required intervention 
or became symptomatic. This series reported 
one of the lowest rates of limb loss in this 
setting (7.3%), and this was attributed in part 
to the avoidance of unnecessary vascular 
exploration in so many of these severely 
injured limbs. These data add support to the 
avoidance of diagnostic arteriography in 
complex extremity trauma that does not man- 
ifest hard signs of vascular injury. 

The optimal method of fracture manage- 
ment in combined vascular and skeletal 
extremity trauma has evolved over the past 
few decades. The military experience from the 
Vietnam War demonstrated substantially 
higher risks of limb loss following internal 
skeletal fixation than was found after exter- 
nal fixation (Rich, 1971; McNamara and col- 
leagues, 1973). At least 50% of amputations 
following internal fixation were due to infec- 
tion. The civilian experience demonstrates 
acceptable results with both internal and exter- 
nal fixation, most likely because of the less 
extensive bone damage in this setting. This 
evidence suggests that combined extremity 
injuries with a high risk of infection (e.g., open, 
contaminated, extensive soft tissue injury) , 
comminuted or unstable skeletal trauma, or 
those in unstable patients who require rapid 
treatment undergo external skeletal fixation, 
either as a definitive or as a temporizing 
measure. Otherwise internal fixation is appro- 
priate, either immediately or as a later defin- 
itive measure. 

Liberal use of a variety of surgical adjuncts 
has shown some correlation with improved 
limb salvage following combined extremity 
trauma, just as it has in isolated extremity vas- 
cular injuries. Completion intraoperative arte- 
riography should be performed routinely 
before completing vascular repairs to docu- 



ment arterial patency and runoff, because any 
technical errors in this tenuous limb could 
easily lead to limb loss. Four-compartment fas- 
ciotomy should be applied liberally and very 
early or prophylactically in this setting because 
of the especially high risk of compartment syn- 
drome following reperfusion. Extra-anatomic 
bypass or pedicle or free tissue flap coverage 
may be necessary to protect vascular repairs 
in the setting of severe contamination and soft 
tissue injury or loss (see Fig. 20-3). 



Indications for Amputation 

Extensive and prolonged attempts to salvage 
extremities with severe and complex injuries 
may actually harm patients in a variety of ways, 
especially if these efforts ultimately end in 
amputation anyway. Financial costs, hospital 
and intensive care unit days, infectious com- 
plications, number of operative procedures, 
time lost from work, permanent disability, and 
even death have all been shown to be signif- 
icantly greater when limb salvage becomes 
unnecessarily prolonged compared with early 
amputation. Those combined extremity 
injuries that ultimately result in limb loss or 
limb dysfunction can largely be predicted 
within a few days of injury by a number 
of prognostic factors that closely relate to 
outcome. Transected major nerves and 
Gustilo III-C injuries (open comminuted 
tibiofibular fractures with arterial injury) are 
the most common indications for considera- 
tion of immediate amputation (Box 20-1). 
Primary amputation at the time of presenta- 
tion or early amputation within a few days 
should be considered strongly whenever these 
prognostic factors are present. The sophisti- 
cation of limb prostheses, the early return to 
work, short hospitalization, and lower costs 
and complications following early amputation 
are usually preferable to salvage attempts, 
which may take months or years and still have 
uncertain success. 

Primary amputation without any attempt 
at limb salvage is reported in 10% to 22% of 
cases of complex extremity trauma, and such 
immediate amputations account for more 
than 50% of all amputations following these 



ch20.qxd 4/16/04 3:31PM Page 413 



20 • SPECIAL PROBLEMS 



413 




COMBINED VASCULAR/SKELETAL EXTREMITY TRAUMA: 
PREDICTIVE FACTORS FOR ULTIMATE LIMB LOSS OR 
DYSFUNCTION 

Transected tibial or sciatic nerve 

Transection of two of three upper extremity nerves 

Gustilo lll-C skeletal injuries 

Prolonged ischemia (>6-12hr) 

Below-knee arterial injury 

Multiple fractures 

Extensive soft tissue loss 

Crush injury 

Severe contamination 

Elderly with medical comorbidity 

Shock and life-threatening associated injuries 



injuries. The decision to perform early ampu- 
tation is one of the most difficult for trauma 
surgeons. Although a number of predictive 
scoring indices have been developed, using 
factors known to correlate with limb salvage 
(see Box 20-1) , none are sufficiently reliable 
as prospective tools to make the decision for 
us. In the end, it must be a matter of judg- 
ment based on each individual case. This deci- 
sion should always involve and require the 
assent of the entire team involved in the care 
of the patient, including the trauma, vascu- 
lar, orthopedic, and plastic surgeons, reha- 
bilitation specialist, psychologist, nursing, 
and especially the patient and family. 

A major consideration in the decision for 
amputation is whether the injury is in the 
upper or lower extremity. The upper extrem- 
ity is more tolerant than the lower of deficits 
in protective sensation, nerve function, and 
length discrepancy, and prostheses are less sat- 
isfactory. Therefore, amputation is generally 
less necessary in the upper extremity for any 
given level of tissue damage. 

There are extremity injuries of such sever- 
ity that a decision for primary amputation is 
not difficult at all (Fig. 20-5). Any obvious 
impossibility or futility of revascularization, 




■ FIGURE 20-5 

Gustilo lll-C complex extremity crush injury in a 
64-year-old diabetic man with absent pulses 
and severe ischemia and no arterial filling on 
arteriogram. Primary above-knee amputation 
was performed immediately without attempting 
limb salvage. ■ 



ch20.qxd 4/16/04 3:31PM Page 414 



414 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



transected major nerves, or associated life- 
threatening injuries that prevent any atten- 
tion to the limbs are clear indications for 
immediate amputation. Nerve transection 
must be confirmed by direct visualization, 
because vascular insufficiency by itself may 
cause profound nerve deficits. 

However, most complex extremity injuries 
are not that clear-cut. In these cases, revascu- 
larization should be performed immediately 
to prevent further tissue damage (including 
aggressive use of surgical adjuncts such as fas- 
ciotomy), the skeleton should be quickly sta- 
bilized by either traction or external fixation, 
and the limb then should be observed over 
the next 48 hours for the level of function that 
returns. If either revascularization fails, tissue 
loss is profound or worsens, systemic sepsis or 
crush syndrome develops, or profound neu- 
rologic dysfunction persists, then amputation 
should be performed. If improvement is 
noted, each successive stage of limb salvage 
should be assessed just as critically to mini- 
mize unnecessarily prolonged, costly, and 
futile efforts. The ultimate goal is to return 
the patient to a comfortable and productive 
life as quickly as possible. 



VASCULAR GRAFTS: 

ROLE AND COMPLICATIONS 



Simple repairs of vascular injuries are always 
preferred if at all possible, including lateral 
suture and resection with end-to-end anasto- 
mosis. Interposition or patch grafting is used 
when simple repair is not possible or not 
preferable, but restoration of the circulation 
is attainable. Any segmental loss of vessel of 
more than 2 cm, as is seen in complex injuries 
with extensive tissue damage, is generally an 
indication for vascular interposition. In true 
end arteries, such as the popliteal artery, lig- 
ation of tenuous collaterals to achieve ade- 
quate mobilization for primary anastomosis 
is best avoided, and interposition grafting is 
preferred. Patch grafting of a partially lacer- 
ated vessel is occasionally useful to avoid steno- 
sis in smaller vessels such as the brachial or 
popliteal arteries and allows avoidance of 
resection. 



In the Vietnam War, Rich and colleagues 
(1970) reported interposition grafting as the 
most common method of surgical repair of 
arterial injuries, with autogenous vein being 
used in 46% of cases and prosthetic grafts 
in only 0.4%. Several civilian series since 
then have corroborated these findings. Most 
authors agree that autogenous reversed 
saphenous vein is the conduit of choice for 
arterial interposition grafting because of its 
high patency rates and low incidence of infec- 
tion with antibiotic control. Rich and Hughes 
(1972) reported an 18% amputation rate in 
Vietnam among arterial repairs undergoing 
autogenous vein interposition, compared 
with 12% in Korea. McCready and colleagues 
(1987) reported an 89.5% long-term patency 
rate for autogenous vein grafts in 86 cases of 
extremity arterial injuries, with five graft fail- 
ures leading to amputation and infection the 
cause of three graft failures. Keen and col- 
leagues (1991) reported 134 patients with 
autogenous vein grafts of extremity arterial 
injuries. In follow-ups ranging up to 24 
months, they found a 98% cumulative primary 
patency rate and 99% cumulative secondary 
patency rate, with one amputation resulting 
from graft failure and no perioperative graft 
infections. Autogenous vein is also the inter- 
position graft of choice for the repair of venous 
injuries, though having a somewhat higher 
rate of thrombosis. McCready and colleagues 
(1987) reported that 17 (77%) of 22 autoge- 
nous vein interpositions for extremity vein 
injuries remained patent. 

Prosthetic grafts have been applied increas- 
ingly to the repair of vascular injuries over the 
past 3 decades, following their widespread and 
successful use in elective vascular surgery. 
However, the contaminated nature of trau- 
matic wounds led to doubts about the suit- 
ability of prosthetic grafting, because of the 
assumption that these materials are highly 
prone to infection. Rich and Hughes (1972) 
reported dismal results using prosthetic grafts 
in contaminated combat vascular injuries in 
Vietnam, with a 77% graft failure rate (most 
commonly from infection and thrombosis) 
and a 31% amputation rate, significantly 
worse than autogenous vein in this setting. The 
lack of an endothelial surface is the most likely 
explanation for the higher rate of thrombo- 



ch20.qxd 4/16/04 3:31PM Page 415 



20 • SPECIAL PROBLEMS 



415 



sis of prosthetic grafts. This was supported by 
some experimental studies in animals in the 
1970s. 

However, a number of subsequent clinical 
studies of civilian vascular trauma have demon- 
strated consistently good results with the use 
of both polytetrafluoroethylene (PTFE) and 
Dacron prosthetic grafts placed in contami- 
nated wounds. Shah and colleagues (1984) 
reported only one arterial and one venous 
graft thrombosis in 25 vascular reconstructions 
with PTFE in grossly contaminated wounds 
with no graft infections. In a report of 236 
PTFE grafts placed in contaminated wounds, 
Feliciano and colleagues (1985) showed a 
higher graft thrombosis rate than autogenous 
vein, but no instances of peripheral graft infec- 
tion in the absence of exposure of the graft 
or osteomyelitis. The liberal use of antibiotics 
and adequate full-thickness tissue coverage of 
these grafts is necessary to achieve these 
results. Failure to cover any graft and vascu- 
lar anastomosis inevitably leads to infection, 
thrombosis, and suture line breakdown. 

One advantage of prosthetic grafts over 
autogenous vein lies in the differential 
response of these materials to infection. Auto- 
genous vein and arterial homografts both 
develop transmural necrosis when subject to 
exposure and/or infection, which leads to 
sudden blowout of the anastomosis as sutures 
pull through, with massive life-threatening 
hemorrhage. Infected prosthetic grafts do not 
break down in this disastrous way but gradu- 
ally develop suture pull-through at the anas- 
tomosis with native vessel, leading to contained 
false aneurysm. This allows time for them to 
be removed electively and new revasculariza- 
tion to be performed. For this reason, pros- 
thetic grafts are considered the material of 
choice to be used if revascularization must be 
done in a contaminated field . If they fail, their 
temporary placement may still allow time to 
debride devitalized tissue and clean the field 
of contamination so that vein maybe used sub- 
sequently. 

Other advantages that prosthetic grafts 
have over autogenous vein for repair of vas- 
cular injuries tend to outweigh their higher 
rate of thrombosis in specific circumstances. 
They are more suitable for interposition graft- 
ing of larger vessels without size discrepancy. 



Their ready availability, without the need for 
harvesting and preparation that autogenous 
veins require, is preferable when time is an 
important factor, as in unstable patients. 
Keen and colleagues (1991) dispute this 
latter point, reporting an average harvesting 
time of less than 8 minutes for autogenous 
veins, which had no adverse impact on limb 
salvage. 

There are circumstances in which prosthetic 
grafts are best avoided. Injury to small-caliber 
vessels, such as the brachial or tibial arteries, 
are more amenable to autogenous vein inter- 
position, because prosthetic grafts smaller 
than 6 mm in diameter have a prohibitive rate 
of thrombosis. Grafts that must cross the knee 
or elbow joint should be autogenous vein, 
because prosthetic grafts tend to kink and 
thrombose more readily. Most prosthetic 
grafts used to repair venous injuries appear 
to thrombose early, although Feliciano and 
colleagues (1985) have reported that even 
their temporary use for this purpose may dra- 
matically diminish hemorrhage from major 
soft tissue wounds and fasciotomy sites that 
venous ligation produces, by relieving venous 
outflow pressure. If used as a venous substi- 
tute, prosthetic grafts with external ring 
support should be applied to enhance their 
patency. 

The proper management of infected pros- 
thetic grafts should begin with measures to 
prevent infection altogether. Routine cover- 
age of vascular repairs with intravenous broad- 
spectrum antibiotics, and full-thickness tissue 
coverage of repairs using primary closure, or 
pedicled or free flap tissue transfers are essen- 
tial elements to avoid infection in this setting. 
If immediate closure of wounds is not possi- 
ble because of extensive contamination, devi- 
talized tissue, or extensive soft tissue loss, 
porcine xenografts have been used success- 
fully to temporarily cover vascular repairs until 
the wound is clean, and primary closure or 
flap transfers can be done. Extra-anatomic 
bypass through clean and uninjured tissues is 
another option for immediate revasculariza- 
tion, which permits adequate coverage despite 
a hostile wound and allows for appropriately 
aggressive wound management and an op- 
timal functional and cosmetic result. Auto- 
genous vein is preferred for extra-anatomic 



ch20.qxd 4/16/04 3:31PM Page 416 



416 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



bypass, but externally supported prosthetic 
grafts may also be used. 

Graft infection must be assumed whenever 
obvious purulence is found in its vicinity, espe- 
cially if it becomes exposed and has failed to 
incorporate with surrounding tissue. Arterial 
graft blowout is another manifestation of infec- 
tion. Immediate excision of the graft is nec- 
essary in this setting, and the native vessel 
should be debrided back to uninvolved tissue 
and ligated there to prevent secondary hem- 
orrhage. This should be followed by extra- 
anatomic bypass to restore distal perfusion, 
leaving the infected bed open for debridement 
and dressing to ultimately granulate and heal 
secondarily. Simply placing a new graft in the 
same infected bed is a futile gesture destined 
for failure. Although there are isolated reports 
of successful conservative management of 
exposed and infected prosthetic grafts, this 
approach is not recommended. 



FAILED RECONSTRUCTION OF 
ARTERIAL TRAUMA 

There are two primary measures of success of 
peripheral arterial injury management: limb 
salvage and limb function. Certainly the 
salvage of a viable extremity is the first goal 
of arterial injury management, but that does 
not necessarily mean that a patient is fully 
restored to their normal lifestyle. The neu- 
rologic and skeletal function of the salvaged 
limb, as well as cosmetic appearance, can have 
a major impact on the patient's life and may 
actually be detrimental enough that amputa- 
tion would be better. Therefore, the ultimate 
goal of peripheral arterial injury management 
must be the salvage of aviable, functional, and 
cosmetically acceptable extremity. The deci- 
sions made and the approaches taken in the 
immediate postinjury period actually have the 
greatest impact on ultimate outcome. 

The first problem that could lead to a failed 
repair of an arterial injury is delay in its diag- 
nosis and treatment. The direct correlation 
between delay in revascularization and limb 
loss is related to irreversible tissue damage 
that develops with more than 4 to 6 hours of 



ischemia. Even if the limb remains viable after 
a treatment delay, permanent disability from 
nerve and muscle damage ispossible even with 
successful revascularization. Prompt assess- 
ment of all injured extremities for hard signs 
of vascular injury, appropriate and selective 
use of hand-injected, on-table arteriography, 
and immediate arterial repair are essential to 
minimizing delay. Nonvascular tissue damage 
also adversely affects the salvage of a viable 
and functional limb in the setting of a con- 
comitant arterial injury. The increased rate 
of limb loss that occurs in combined vascular 
and skeletal extremity trauma has been dis- 
cussed in a previous section of this chapter 
(Combined Vascular and Skeletal Extremity 
Trauma). Extensive soft tissue damage dis- 
rupts collaterals and increases the sensitivity 
of an injured extremity to interruption of 
blood flow. Nerve damage affects the ultimate 
function of an injured extremity independent 
of how well it is revascularized. Injury to a 
major vein in an extremity with arterial injury 
increases the chance of failure of the arterial 
repair byjeopardizingvenous outflow. In these 
circumstances, early diagnosis and revascu- 
larization and optimal use of surgical adjuncts 
to improve the success of vascular repair are 
critically important factors. 

Technical errors may lead to failure of arte- 
rial repair and to limb loss or limb dysfunc- 
tion, and therefore must be stringently 
avoided. Strict attention is necessary to a 
number of technical factors, including gentle 
dissection, thorough debridement, appro- 
priate prioritization of multiple injuries, 
meticulous technique in suturing arteries and 
veins, proximal and distal thrombectomy, 
regional and systemic heparinization to pre- 
vent further thrombosis, proper choice of 
repair technique to avoid stenosis, undue 
tension and collateral damage, full-thickness 
tissue coverage of the repair, and confirma- 
tion of restoration of blood flow by palpation 
of pulses and clinical signs of normal perfu- 
sion. Completion arteriography should be 
done routinely to detect those unsuspected 
problems with the vascular repair or distal 
runoff that even the most experienced centers 
find in up to 16% of cases, which can be im- 
mediately fixed and will avert a subsequent 



ch20.qxd 4/16/04 3:31PM Page 417 



20 • SPECIAL PROBLEMS 



417 



failure of the arterial reconstruction. Certainly, 
any doubts about the arterial repair must be 
assessed by an intraoperative arteriogram. 
Anticoagulation should not be necessary fol- 
lowing arterial repairs and cannot substitute 
for technical perfection. 

Postoperative surveillance of the arterial 
reconstruction with frequent checks of pulses, 
clinical signs of perfusion, bleeding, and 
Doppler pressure monitoring is necessary to 
detect any thrombosis or anastomotic dis- 
ruption early and permit immediate revisions. 
Any sign of perfusion deficit or active bleed- 
ing must be investigated promptly by arteri- 
ography or operative exploration. The cause 
of a failed reconstruction should be deter- 
mined, to allow appropriate repair. Any throm- 
bosis must be assumed to be due to technical 
problems, and the anastomosis or suture lines 
should be redone following distal thrombec- 
tomy. An intraoperative arteriogram must 
confirm an adequate repair before comple- 
tion of the surgery. If infection is found as the 
cause of failed reconstruction, consideration 
must be given to extra-anatomic bypass fol- 
lowing excision of the infected portion of 
vessel and ligation in a clean field. 

Compartment hypertension can be insidi- 
ous in its presentation and devastating in how 
much tissue and limb function it can destroy 
following repair of arterial injuries, even with 
successful revascularization and in the pres- 
ence of normal pulses. Any suspicion of this 
problem mandates immediate and complete 
fasciotomy of the injured extremity. It is best 
to perform fasciotomy early, and even pro- 
phylactically, in extremities known to be at risk 
for compartment syndrome, to avert its onset 
altogether. Those factors posing a high risk 
for compartment syndrome are well estab- 
lished (see Chapter 27). 

Each instance of failure of arterial recon- 
struction following extremity vascular injury 
substantially reduces the chance of ultimate 
limb salvage. Prevention of these failures 
through optimal diagnosis and initial man- 
agement is the best way to minimize limb loss. 
If a failure occurs, limb salvage and good limb 
function is still possible if recognized and 
treated promptly. 



REFERENCES 

Agarwal N, Shah PM, Clauss RH, et al: Experience 
with 115 civilian venous injuries. J Trauma 
1982;22:827. 

Alexander JJ, Piotrowski JJ, Graham D, et al: 
Outcome of complex orrthopedic and vascular 
injuries of the lower extremity. Am J Surg 
1991;162:111. 

Applebaum R, \fellin AE, Weaver FA, et al: Role of 
routine arteriography in blunt lower extremity 
trauma. Am J Surg 1990;1 60:221. 

Armstrong K, Sfeir R, Rice J, et al: Popliteal vas- 
cular and war: Are Beirut and New Orleans 
similar? J Trauma 1988;28:836. 

AtteberyLR, DennisJW, Russo-Alesi Fetal: Chang- 
ing patterns of arterial injuries associated with 
fractures and dislocations. J Am Coll Surg 
1996;183:377. 

Barcia PJ, Nelson TG, Whelan TJ: Importance of 
venous occlusion in arterial repair failure: An 
experimental study. Ann Surg 1972;1 75:223. 

BarrosD'Sa AA, HassardTH, Livingston RH, etal: 
Missile-induced vascular trauma. Injury 
1980;12:13. 

Bergstein JM, Blair JF, Edwards J, et al: Pitfalls in 
the use of color-flow duplex ultrasound for 
screening of suspected arterial injuries in pen- 
etrated extremities. J Trauma 1992;33:395. 

Bongard FS, White GH, Klein SR: Management 
strategy of complex extremity injuries. Am J Surg 
1989;158:151. 

Bishara RA, Pasch AR, Lim LT, et al: Improved 
results in the treatment of civilian vascular 
injuries associated with fractures and disloca- 
tions. J Vase Surg 1986;3:707. 

Bishara RA, Pasch AR, Lim LT, et al: Improved 
results in the treatment of civilian vascular 
injuries associated with fractures and disloca- 
tions. J Vase Surg 1976;3:707. 

Borman KR, Jones GH, Snyder WH: A decade of 
lower extremity venous trauma. Am J Surg 
1987;154:608. 

Burch JM, Richardson RJ, Martin RR, et al: Pene- 
trating iliac vascular injuries: Recent experience 
with 233 consecutive patients. J Trauma 
1990;34:1450. 

Conkle DM, Richie RE, Sawyers JL, et al: Surgical 
treatment of popliteal artery injuries. Arch Surg 
1975:110:1351. 



ch20.qxd 4/16/04 3:31PM Page 418 



418 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



Connolly JE, Williams E, Whittaker D: The influ- 
ence of fracture stabilization on the outcome of 
arterial repair in combined fracture -arterial 
injuries. Surg Forum 1969;20:450. 

Daugherty ME, Sachatello CR, Ernst CB: Improved 
treatment of popliteal arterial injuries. Arch Surg 
1978;113:1317. 

DeBakey ME, Simeone FA: Battle injuries of the 
arteries in World War II: An analysis of 2,471 cases. 
Ann Surg 1946;1 23:534. 

DeGiannis E, Levy RD, Sofianos C, et al: Arterial 
gunshot injuries of the extremities: A South 
African experience. J Trauma 1995;39:570. 

Dennis JW, Frykberg ER, Veldenz HC, et al: 
Reassessing the role of arteriograms in the man- 
agement of posterior knee dislocations. J Trauma 
1993;35:692. 

Downs AR, MacDonald P: Popliteal artery injuries: 
Civilian experience with sixty-three patients 
during a 24 year period (1960 through 1984). 
JVascSurgl986;4:55. 

DrostTF,RosemurgyAS,ProctorD,etal: Outcome 
of treatment of combined orthopedic and arte- 
rial trauma to the lower extremity. J Trauma 
1989;29:1331. 

Fabian TC, Turkleson ML, Connelly TL, etal: Injury 
to the popliteal artery. Am J Surg 1982;143:225. 

Fainzilber G, Roy-Shapir A, Wall MJ, et al: Predic- 
tors of amputation for popliteal artery injuries. 
Am J Surg 1995;170:568. 

Feliciano DV, Accola KD, Burch JM, et al: 
Extraanatomic bypass for peripheral arterial 
injuries. Am J Surg 1989;158:506. 

Feliciano DV, Cruse PA, Spjut-Patrinely V, et al: Fas- 
ciotomy after trauma to the extremities. Am J 
Surg 1988;156:533. 

Feliciano DV, Mattox KL, Graham JM, et al: Five- 
year experience with PTFE grafts in vascular 
wounds. J Trauma 1985;25:7l. 

Frykberg ER: Popliteal vascular injuries. Surg Clin 
North Am 2002;82(l):67-89. 

Frykberg ER, Crump JM, Dennis JW, et al: Non- 
operative observation of clinically occult arter- 
ial injuries: A prospective evaluation. Surgery 
1991;109:85. 

Gagne PJ, Cone JB, McFarland D, et al: Proximity 
penetrating extremity trauma: The role of duplex 
ultrasound in the detection of occult venous 
injuries. J Trauma 1995;39:1157. 

Caspar MR, Treiman RL: The management of 
injuries to major veins. Am J Surg 1960;100: 
171. 



Gerlock AJ, Thai ER, Snyder WH: Venography in 
penetrating injuries of the extremities. AJR 
1976;126:1023. 

Harrell DJ, Spain DA, Bergamini TM, et al: Blunt 
popliteal artery trauma: A challenging injury. 
Am Surg 1997;63:228. 

Hobson RW, Croom RD, Rich NM: Influence of 
heparin and low molecular weight dextran on 
the patency of autogenous vein grafts in the 
venous system. Ann Surg 1973;178:773. 

Hobson RW, Howard EW, Wright CB, et al: Hemo- 
dynamics of canine femoral venous ligation: 
Significance in combined arterial and venous 
injuries. Surgery 1973;74:824. 

Hobson RW, \feager RA, Lynch TG, et al: Femoral 
venous trauma: Techniques for surgical man- 
agement and early results. Am J Surg 
1983;146:220. 

HollemanJH, KillebrewLH: Injury to the popliteal 
artery. Surg Gynecol Obstet 1981;153:392. 

Howe HR, Poole GV, Hansen KJ, et al: Salvage of 
lower extremities following combined orthope- 
dic and vascular trauma: A predictive salvage 
index. Am Surg 1987;53:205. 

Hughes CW: Arterial repair during the Korean War. 
Ann Surg 1958;147:555. 

Jaggers RC, Feliciano DV, Mattox KL, et al: Injury 
to popliteal vessels. Arch Surg 1982;1 17:657. 

Jahnke EJ, Seeley SF: Acute vascular injuries in the 
Korean War: An analysis of 77 consecutive cases. 
Ann Surg 1953;138:158. 

Johansen K, Daines M, Howey T, et al: Objective 
criteria accurately predict amputation following 
lower extremity trauma. J Trauma 1990;30: 
568. 

Kaufman SL, Martin LG: Arterial injuries associ- 
ated with complete dislocation of the knee. Radi- 
ology 1992;184:153. 

Keen RR, Meyer JP, Durham JR, et al: Autogenous 
vein graft repair of injured extremity arteries: 
Early and late results with 134 consecutive 
patients. J Vase Surg 1991;13:664. 

Kendall RW, Taylor DC, Salvian AJ, et al: The role 
of arteriography in assessing vascular injuries 
associated with dislocations fo the knee .J Trauma 
1993;35:875. 

Krige JEJ, Spence RAJ: Popliteal artery trauma: 
A high risk injury. Br J Surg 1987;74:91. 

Lange RH, Bach AW, Hansen ST, et al: Open 
tibial fractures with associated vascular injuries: 



ch20.qxd 4/16/04 3:31PM Page 419 



20 • SPECIAL PROBLEMS 



419 



Prognosis for limb salvage. J Trauma 1985;25: 
203. 

Lim LT, Michuda MS, Flanigan DP, et al: Popliteal 
artery trauma: 31 consecutive cases without 
amputation. Arch Surg 1980;115:1307. 

Lin C-H, Weif-C, Levin LS, et al: The functional 
outcome of lower extremity fractures with vas- 
cular injury. J Trauma 1997;43:480. 

Lovric Z, Wertheimer B, Candrlic K, et al: War 
injuries of major extremity vessels. J Trauma 
1994;36:248. 

Makins GH: Injuries to the bloodvessels. In: Offi- 
cial History of the Great War Medical Services: 
Surgery of the War. London: His Majesty's 
Stationery Office, 1922;170-296. 

Martin LC, McKenny MG, Sosa JL, et al: Manage- 
ment of lower extremity arterial trauma. J 
Trauma 1994;37:591. 

Martinez D, Sweatman K, Thompson EC: Popliteal 
artery injury associated with knee dislocations. 
AmSurg2001;67:165. 

McCabe CJ, Ferguson CM, Ottinger LW: Improved 
limb salvage in popliteal artery injuries. J Trauma 
1983;23:982. 

McCready RA, Logan NM, Dangherty ME, et al: 
Long-term results with antogenous tissue repair 
of traumatic extremity vascular injuries. Ann Surg 
1987;206:804. 

Melton SM, Croce MA, Patton JH, et al: Popliteal 
artery trauma: Systemic anticoagulation and 
intraoperative thrombolysis improves limb 
salvage. Ann Surg 1997;225:518. 

McNamara JJ, Brief DK, Stremple JF, et al: Man- 
agement of fractures with associated arterial 
injury in combat casualties. J Trauma 1973;13:17. 

Meyer J, Walsh J, Schuler J, et al: The early fate of 
venous repair after civilian vascular trauma: a 
clinical, hemodynamic, and venographic assess- 
ment. Ann Surg 1987;206:458. 

Miranda FE, Dennis JW, Veldenz HC, et al: Con- 
firmation of the safety and accuracy of physical 
examination in the evaluation of knee disloca- 
tion for popliteal artery injury: A prospective 
study. J Trauma 2000;49:375. 

Miranda FE, Dennis JW, Veldenz HC, et al: Con- 
firmation of the safety and accuracy of physical 
examination in the evaluation of knee disloca- 
tion for injury of the popliteal artery: A prospec- 
tive study. J Trauma 2002;52(2):247-251. 

Mullins RJ, Lucas CE, Ledgerwood AM: The 
natural history followingvenous ligation for civil- 
ian injuries. J Trauma 1980;20:737. 

Norman J, Gahtan V, Franz M, et al: Occult vascu- 
lar injuries following gunshot wounds resulting 



in long bone fractures of the extremities. Am 
Surgl995;61:146. 

Nypaver TJ, Schuler JJ, McDonnel P, et al: 
Long-term results ofvenous reconstruction after 
vascular trauma. J Vase Surg 1992;16:762. 

Odland MD, Gisbert VL, Gustilo RB, et al: Com- 
bined orthopedic and vascular injury in the lower 
extremities: Indicationsforamputation. Surgery 
1990;108:660. 

O'Reilly MJG, HoodJM, Livingston RH, etal: Pen- 
etrating injuries of the popliteal artery. BrJ Surg 
1978;65:789. 

Orcutt MB, Levine BA, Root HD, et al: The con- 
tinuing challenge of popliteal vascular injuries. 
AmJ Surg 1983;146:758. 

Palazzo JC, Ristow AB, CuryJM, et al: Traumatic 
vascular lesions associated with fractures and dis- 
locations. J Cardiovasc Surg 1986;27:688. 

Pasch AR, Bishara RA, Schuler JJ, et al: Results of 
venous reconstruction after civilian vascular 
trauma. Arch Surg 1986;1 21:607. 

PeckJJ, Eastman AB, Bergan JJ, et al: Popliteal vas- 
cular trauma: A community experience. Arch 
Surgl990;125:1339. 

Phifer TJ, Gerlock AJ, Rich NM, et al: Long term 
patency of venous repairs demonstrated by 
venography. J Trauma 1985;25:342. 

Pretre R, Bruschweiler I, RossierJ, etal: Lower limb 
trauma with injury to the popliteal vessels. 
J Trauma 1996;40:595. 

Razuk AF, Nunes H, Coimbra R, et al: Popliteal 
artery injuries: Risk factors for limb loss. Panam 
J Trauma 1998;7:93. 

Reed MK, Lowry PA, Myers SI: Successful repair of 
pediatric popliteal artery trauma. Am J Surg 
1990;160:287. 

Rich NM: Principles and indications for primary 
venous repair. Surgery 1982;91:492. 

Rich NM, Baugh JH, Hughes CW: Acute arterial 
injuries in Vietnam: 1,000 cases. J Trauma 
1970;10:359. 

Rich NM, Hobson RW, Collins GJ, et al: The effect 
of acute popliteal venous interruption. Ann Surg 
1976;183:365. 

Rich NM, Hughes CW: The fate of prosthetic mate- 
rial used to repair vascular injuries in contami- 
nated wounds. J Trauma 1972;21:459. 

Rich NM, Hughes CW, Baugh JH: Management of 
venous injuries. Ann Surg 1970;171:724. 

Rich NM, Metz CW, HuttonJE, etal: Internal versus 
external fixation of fractures with concomitant 
vascular injuries in Vietnam. J Trauma 1971; 
11:463. 



ch20.qxd 4/16/04 3:31PM Page 420 



420 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



Romanoff H, Goldberger S: Combined severe 
vascular and skeletal trauma: management and 
results. J Cardiovasc Surg 1979;20:493. 

Ross SE, Ransom KJ, Shatney CH: The manage- 
ment of venous injuries in blunt extremity 
trauma. J Trauma 1985;25:150. 

Schlickewei W, Kuner EH, Mullaji AB, et al: 
Upper and lower limb fractures with concomi- 
tant arterial injury. J Bone Joint Surg 1992;74: 
181. 

Sfeir RE, Khoury GS, Haddad FF, et al: Injury to 
the popliteal vessels: The Lebanese War experi- 
ence. WorldJ Surg 1992;16:1156. 

Shah DM, Leather RP, Corson JD, et al: Polytetra- 
fluoroethylene grafts in the rapid reconstruction 
of acute contaminated peripheral vascular 
injuries. Am J Surg 1984;148:229. 

Shah DM, Naraynsingh V, Leather RP, et al: 
Advances in the management of acute popliteal 
vascular blunt injuries. J Trauma 1985;25:793. 

Spencer FC, Grewe RF: The management of arte- 
rial injuries in battle casualties. Ann Surg 
1955;141:304. 

Snyder WH: Vascular injuries near the knee: An 
updated series and overview of the problem. 
Surgery 1982;91:502. 

Swetnam JA, Hardin WD, Kerstein MD: Successful 
management of trifurcation injuries. Am Surg 
1986;52:585. 



Thomas DD, Wilson RF, Wiencek RG: Vascular 
injury about the knee: Improved outcome. Am 
Surg 1989;55:370. 

Timberlake GA, Kerstein MD: Venous injury: To 
repair or ligate-the dilemma revisited. Am Surg 
1995;61:139. 

Treiman GS, Yellin AE, Weaver FA, et al: Exami- 
nation of the patientwith a knee dislocation: The 
case for selective arteriography. Arch Surg 
1992;127:1056. 

Van Wijngaarden M, Omert L, Rodriguez A, et al: 
Management of blunt vascular trauma to the 
extremities. Surg Gynecol Obstet 1993;177:41. 

Weimann S, San Nicolo M, Sandblicher P, et al: 
Civilian popliteal artery trauma. J Cardiovasc 
Surg 1987;28:145. 

Wright CB, Hobson RW: Hemodynamic effects of 
femoral venous occlusion in the subhuman 
primate. Surgery 1974;75:453. 

Yeager RA, Hobson RW, Lynch TG, et al: Popliteal 
and infrapopliteal arterial injuries: Differential 
management and amputation rates. Am Surg 
1984;50:155. 

Yelon JA, Scalea TM: Venous injuries of the lower 
extremities and pelvis: repair versus ligation. 
J Trauma 1992;33:532. 



ch21.qxd 4/16/04 3:30 PM Page 421 




Illicit Street Drugs and 
Vascular Injury 



CHARLES E. LUCAS 
ANNA M. LEDGERWOOD 



O INTRODUCTION 

O PERIVASCULAR HEMATOMA AND ABSCESS 

O DIRECT ARTERIAL INJECTION 

O MYCOTIC ANEURYSMS 

O VASCULAR RECONSTRUCTION 

O MYCOTIC ANEURYSMS OF CAROTID VESSELS 

O VENOUS ANEURYSMS 

O VASCULAR INJURY FROM COCAINE 



INTRODUCTION 



The use of illicit street drugs is very common 
in our society, especially amongyoungpeople 
who are most susceptible to trauma. The 
method of administration and the pharma- 
cologic responses of these drugs produce a 
unique pattern of vascular injury. This chapter 
is dedicated to the specific type of vascular 
problems associated with illicit drug usage. 



PERIVASCULAR HEMATOMA 
AND ABSCESS 



A common phenomenon in illicit drug 
users using intravenous access is a missed 
venous puncture that results in the needle 
entering an adjacent artery. This is often 
recognized by the withdrawal of bright red 
blood, known as "a pinky" among the users. 
When recognized, the needle is removed and 

421 



ch21.qxd 4/16/04 3:30PM Page 422 



422 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



direct pressure is placed over the puncture 
site; despite this precaution, a hematoma often 
results. Treatment consists of observation and 
oral antibiotics, which are readily available on 
the street. Formal medical care is avoided 
unless complications ensue. 

Because of the high incidence of contami- 
nation associated with the drug preparation 
including the multiple dilutions, which are 
performed to maximize profits, the hematoma 
has a high risk of becoming seeded from the 
contaminated injectable. This typically leads 
to infection of the perivascular clot and sub- 
sequent abscess formation. Sometimes a frag- 
ment of needle will be found within the abscess 
cavity. Physical examination identifies an area 
of cellulitis overlying the neurovascular 
bundle, which commonly is at the wrist or the 
groin. Transmitted pulsations may lead to the 
suspicion that this is a false aneurysm. When 
confusion exists about the nature of the 
pulsatile inflammatory mass, preoperative 
arteriography is warranted. During operative 
evacuation of these abscesses, the surgeon 
should avoid breaking all the adhesions 
because one end of the abscess cavity will abut 
the arterial wall. Overaggressive drainage of 
abscesses in this setting will lead to bleeding 
from the adjacent artery, where the puncture 
site had been sealed by an established platelet 
and fibrin plug. 

Concomitant antibiotic therapy using 
broad-spectrum coverage is needed. During 
a 12-month interval at the Detroit Receiving 
Hospital, 651 patients had abscesses drained 
by the surgical services; 421 of these patients 
had abscesses that resulted from illicit street 
drug use. Most patients who have a single 
organism cultured will have a Staphylococcus 
aureus infection, which often is resistant to 
methicillin (methicillin-resistant S. aureus) . 
(3-streptococcus is also commonly found as 
an isolated organism. Approximately 25% of 
patients will have a mixed infection with gram- 
negative coliform organisms being part of this 
mixture. Many of the users were using their 
larger veins (mainlining) so the heroin mix 
would be injected into the subcutaneous 
plane (skin-popping) . This compromises the 
surgeon's ability to determine whether the 
underlying cellulitis is related to the skin- 
popping or to an arterial injury. 



DIRECT ARTERIAL INJECTION 



Often patients who hit a "pinky" are already 
under the influence of alcohol or drugs and 
do not recognize that the needle has been 
inserted into an artery. The heroin mix is then 
injected intra-arterially followed by an imme- 
diate burning pain in the distribution area of 
that artery. This results from the emboliza- 
tion of particulate matter that has been used 
to dilute the heroin mix, the so-called "mixed 
jive, "which plugs the distal microvascular tree. 
When small vessel occlusion occurs after a 
radial artery or ulnar artery injection, the 
ischemic necrosis typically involves the skin 
and subcutaneous tissues along the distribu- 
tion of the thumb or the first and second 
fingers after a radial artery injection or along 
the distribution of the third and fourth fingers 
after an ulnar artery injection. This soft tissue 
insult is extremely painful. When patients 
present to an emergency department, the 
underlying etiology often is not recognized 
and the patients are treated with oral anal- 
gesics and sometimes antimicrobials. When 
first seen by a surgeon, there is usually evi- 
dence of full-thickness skin and sometimes 
subcutaneous necrosis. The prime therapeu- 
tic objectives are prevention of superimposed 
infection and amelioration of the constant 
pain. The role of intra-arterial crystalloid irri- 
gation, heparin infusion, or other intra- 
arterial modalities of treatment have not 
been successful in this group of patients. 
Systemic analgesics provide minimal relief. 
Significant amelioration can be achieved by 
sympathetic blockade. Transient upper 
extremity pain relief may be obtained by a stel- 
late block. Patients who experience significant 
relief on two separate stellate blocks likely will 
benefit from a dorsal sympathectomy per- 
formed through a small transaxillary incision. 
Patients undergoing sympathectomy in this 
setting will have increased warmth of the 
involved hand and a striking but not complete 
reduction in pain. 

Sometimes the ischemic changes are asso- 
ciated with rapidly spreading infection that 
involves the adjacent muscles and leads to 
myonecrosis. Such patients will need ampu- 
tation, which following lower limb or groin 



ch21.qxd 4/16/04 3:30 PM Page 423 



21 • ILLICIT STREET DRUGS AND VASCULAR INJURY 



423 



injection is a below-knee amputation; seldom 
is an above-knee amputation needed. When 
myonecrosis occurs in the hand, individual 
digits may need amputation. Unfortunately, 
we have performed several hands and even 
forearm amputations following wrist injections 
or brachial artery injections, respectively. 
Lack of an aggressive approach to myone- 
crosis after injections with contaminated 
street narcotics will lead to severe systemic 
septicemia, rhabdomyolysis, and renal 
shutdown. 



MYCOTIC ANEURYSMS 



In 1851 Koch described the first mycotic 
aneurysm. This occurred in a 22-year-old 
woman with a history of rheumatic fever with 
consequent endocarditis. Bacteria embolized 
from the heart valves into the superior 
mesenteric artery, which became aneurysmal, 
ruptured, and caused death. Autopsy demon- 
strated that the aneurysm was in one of the 
secondary arcades of the superior mesenteric 
artery. In 1885 Osier coined the term "mycotic 
aneurysm" when he described a 30-year-old 
patient who had a history of rheumatic fever 
with endocarditis and then developed four 
thoracic aortic aneurysms. One of these 
aneurysms ruptured causing death. At 
postmortem examination, Osier was 
impressed by the appearance of "fresh fungus 
vegetations" around the aneurysm. These 
were not fungus mounds, but inflammatory 
masses around bacteria-induced infection. 
The cause for mycotic aneurysm formation 
was endocarditis, resulting in embolization of 
bacteria into the vasa vasorum resulting in arte- 
rial wall infection and consequent aneurys- 
mal dilation. 

Mycotic aneurysms (infected pseudoa- 
neurysms) in drug addicts are due to direct 
arterial trauma from an errant needle stick 
resulting in a perivascular hematoma that 
becomes infected. Huebl and Reid in 1966 
described this sequence of events and referred 
to this entity as "aneurysmal abscess." During 
a 20-month interval at the Detroit Receiving 
Hospital, the surgeons excised 52 mycotic 
aneurysms or aneurysmal abscesses in 50 



patients following intra-arterial injection with 
heroin mix, or "mixedjive." Often, the patient 
gave a history of a pinky but more often the 
intra-arterial injection was not recognized. 
The patient typically presented with pain, 
swelling, fever, and leukocytosis around 
the area of injection. About half of the 
patients had obvious pulsation of the inflam- 
matory mass at the time of the original 
examination. The duration of symptoms 
usually was about 1 week. About 25% of 
patients had a decreased or absent peripheral 
pulse distal to the inflammatory mass. Few 
patients (10%) had symptoms of limb 
ischemia. An associated neural deficit is 
usually caused by a direct injection into or 
around the adjacent nerve rather than 
ischemia. The most common missed diagno- 
sis was cellulitis, especially when not seen 
promptly by a surgeon. The resultant admin- 
istration of intravenous antibiotics was 
followed by a lack of rapid response, which 
signaled the presence of the mycotic 
aneurysm. 

When mycotic aneurysm is considered, arte- 
riography is recommended. This confirms the 
diagnosis and serves as a road map defining 
both the site of leakage and the extent of 
collateralization. The operative approach 
should entail careful proximal and distal 
control of all involved vessels. This often is 
difficult because of the intense inflammation 
in the tissues around the abscess. This is due 
to the multiple prior injections in this area. 
Extensive fibrosis from repeated soft tissue 
exposure to mixed jive mistakenly injected 
around the vein impedesrapid safe dissection. 
Consequently, one must dissect very slowly 
after obtaining vascular control, to have as 
much anatomy displayed as possible before 
actually getting into the aneurysmal abscess. 
Very slow and careful dissection helps avoid 
injury to nearby structures, particularly the 
adjacent vein, which is often encased in this 
inflammatory mass. 

Once fully exposed, the mycotic aneurysm 
with the adjacent artery should be excised, 
followed by proximal and distal ligation of 
uninvolved arterial wall. Suture ligation of the 
aneurysm artery without excision likely will 
lead to rebleeding. Aneurysmectomy will be 
tolerated without tissue loss in most patients 



ch21.qxd 4/16/04 3:30 PM Page 424 



424 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



with upper extremity aneurysmal abscesses 
and in mostpatientswith aneurysmal abscesses 
involving the superficial femoral artery or the 
profunda femoral artery. Even the involved 
popliteal artery has been excised without tissue 
loss, although this artery is not commonly 
involved with a mycotic aneurysm. The 
need to perform aneurysmectomy of the 
common femoral artery with triple ligation 
of the common femoral artery, superficial 
femoral artery, and profunda femoral artery 
leads to a high incidence of ischemia 
and necrosis requiring amputation. Attempts 
to predict, from preoperative arteriographic 
findings, the level of resultant ischemia 
after aneurysmectomy are fraught with 
failure. 



VASCULAR RECONSTRUCTION 

Ideally, major artery aneurysmectomy is 
followed by vascular reconstruction. This aim 
however is hindered after mycotic aneurys- 
mectomy in narcotic addicts by the sur- 
rounding inflammation and cellulitis. Efforts 
at reconstruction therefore must be directed 
toward extra-anatomic routes. External iliac 
artery mycotic aneurysms are rare; when such 
aneurysms are excised, reconstruction is best 
achieved by a femoral-to-femoral artery bypass 
if neither groin is involved with drug-related 
cellulitis. The timing of extra-anatomic recon- 
struction depends on the patient's presenta- 
tion. When preoperative ischemic pain exists 
and there is an available extra-anatomic route, 
the bypass may be established before aneurys- 
mectomy. When preoperative ischemic pain 
is absent, aneurysmectomy without bypass is 
indicated. 

When common femoral artery aneurys- 
mectomy with triple ligation leads to ischemia, 
the patient will wake up complaining of 
severe, unrelenting pain in the foot. When 
neither the lower abdominal wall nor the distal 
thigh has cellulitis, the patient should be taken 
back to the operating room for placement 
of an extra-anatomic bypass between the 
external iliac artery and the distal femoral 
artery or proximal popliteal artery. The 



extra-anatomic bypass is best performed 
through the obturator foramen. The exter- 
nal iliac artery is most easily exposed through 
an oblique suprainguinal incision in Langer's 
lines followed by retroperitoneal dissection, 
which also allows access to the obturator 
foramen. The thigh incision must stay distal 
to the inflammatory changes abutting the 
groin cellulitis. This procedure is technically 
challenging and therefore dangerous because 
of the extensive collateral circulation associ- 
ated with the groin cellulitis. The surgeon 
should detach the obturator membrane from 
its anterior and medial osseous insertion, 
thereby minimizing the threats of venous 
hemorrhage and neural contusion. The 
danger of rerouting through the obturator 
foramen has resulted in the recommendation 
that the lateral femoral triangle be used for 
this bypass. This approach, whereby, the graft 
goesjust medial to the anterior superior spine, 
is really not extra-anatomic and is prone to 
failure in patients who have cellulitis involv- 
ing the femoral triangle. Avein is almost never 
available in these patients because they have 
already destroyed their veins with prior injec- 
tions. Consequently, one must use a synthetic 
graft. We prefer to use the Dacron graft, 
although the long-term patency rates between 
Dacron and PTFE grafts are not different. 

The long-term success of extra-anatomic 
bypass grafts in these patients is directly 
related to recidivism of the drug usage. When- 
ever a patient goes back to using heroin mix 
and resorts to intravascular injection, the like- 
lihood for thrombosis of the obturator 
foramen bypass graft approaches 100%. When 
thrombosis does occur, some patients have 
ischemia that is tolerable and other patients 
require amputation, which is usually at the 
below-knee level. 

While treating patients with aneurysmal 
abscesses, antibiotic therapy is necessary. Most 
patients will have positive culture results for 
methicillin-resistant S. aureus, whereas a sig- 
nificant number of patients will have positive 
culture results for Pseudomonas aeruginosa. 
About 20% of the patients will have a mixed 
flora within the aneurysmal abscess, so 
broad-spectrum antibiotics are necessary. 
Likewise, the skin over the aneurysmal 



ch21.qxd 4/16/04 3:30 PM Page 425 



21 • ILLICIT STREET DRUGS AND VASCULAR INJURY 



425 



excision site should be left open to heal by 
second intent. 



VENOUS ANEURYSMS 



MYCOTIC ANEURYSMS OF 
CAROTID VESSELS 

The potential for an infected pseu- 
doaneurysm extends to any artery being 
injected. Some mainliners have avoided 
using the groin as a site of injection for fear 
of losing their legs. Somehow the thought 
that the blood supply to the brain could be 
compromised by carotid artery injection is 
not considered. The differential diagnostic 
challenge for a mycotic aneurysm involving 
the common or innominate arteries is the 
same as in the extremities. The lack of reso- 
lution of an inflammatory mass to antibiotic 
therapy should highlight the fact that this 
may be an aneurysmal abscess. The princi- 
ples of treatment are the same. The surgeon 
must obtain proximal and distal control. 
Unfortunately, after excision of the infected 
pseudoaneurysm of the carotid or innomi- 
nates arteries, there is never a plane that is 
not involved with cellulitis, so there is no 
potential for placing a bypass graft. Fortu- 
nately, these patients usually have good col- 
lateralization and do not develop evidence of 
cerebral ischemia after mycotic aneurysmec- 
tomy of aneurysms of the innominate artery, 
external carotid artery, common carotid 
artery, or internal carotid artery. The post- 
operative care requires the same con- 
siderations regarding antibiotics with 
broad-spectrum coverage and leaving the 
skin open to heal by second intent. 

Occasionally, patients with mycotic 
aneurysms will have a fistula between the 
artery and the vein. The principles of care in 
such patients are the same as those for either 
a mycotic arterial aneurysm or a venous 
aneurysm. Proximal and distal control is nec- 
essary to get the arterial component isolated. 
The venous component should then be con- 
trolled proximally and distally before enter- 
ing the artery and doing the aneurysmectomy. 
The care in dissection is especially important 
in patients with an arteriovenous fistula. 



Septic phlebitis is a common coexistent con- 
dition in patients with drug injection-related 
cellulitis caused by mainlining or skin- 
popping. By the time the patient goes to the 
emergency department with complications 
from missed hits, several misses have occurred 
over the many previous weeks and months. 
The suspicion that a patient has something 
more than simple cellulitis is enhanced by the 
appearance of systemic sepsis that exceeds the 
severity typical of localized cellulitis. The pres- 
ence of bilateral lung abscesses typifies the 
patient who has a venous aneurysm that is 
embolizing bacteria to the lungs. These 
changes in the lung are not caused by blood 
clots but are caused by embolization of bac- 
teria. Anticoagulation should be avoided in 
this setting because the patient may also have 
endocarditis and small intracerebral infarcts 
from bacterial embolization. Anticoagulation 
may cause one of these intracerebral infarcts 
to hemorrhage. The surgical approach for 
mycotic venous aneurysm is excision plus 
proximal and distal venous ligation. Again, the 
dissection should be done very carefully to 
avoid injury to adjacent arteries and nerves. 
Fogarty catheterization may be helpful in 
retracting infected clot after venous control 
is obtained. After excision of a venous pseudo- 
aneurysm, the patient is maintained on antibi- 
otics and the limb is elevated. The skin is 
allowed to heal by second intent. Long-term 
care involves support wraps until the wounds 
have healed, after which time lifelongwearing 
of a customized venous support hose is nec- 
essary. These patients are not candidates for 
long-term anticoagulation, and they are not 
candidates for later vein graft inner position 
for their venous insufficiency. 



VASCULAR INJURY FROM 
COCAINE 

Cocaine in various forms has become a very 
popular substance abuse agent in all walks of 
life. The vasospastic effects of cocaine produce 



ch21.qxd 4/16/04 3:30PM Page 426 



426 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



a multitude of clinical problems that involve 
the vascular surgeon. The intense vasocon- 
strictive effects causes multiple-organ dys- 
function in young patients. These include 
myocardial infarction, cardiac arrhythmia, 
acute renal failure, cerebral vascular ischemia, 
and rhabdomyolysis. Thrombosis of small 
vessels is typically associated with ischemia of 
the distal part and is treated symptomatically 
with plasma volume expansion and observa- 
tion. Occasionally, the ischemia will lead 
to severe myonecrosis, necessitating 
amputation. 

Cocaine may also cause thrombosis of 
larger vessels including the abdominal aorta. 
This occlusion is thought to result from 
spasm of the vasa vasorum, resulting in an 
intimal injury followed by platelet deposition 
and clot formation. We treated one patient 
who had cocaine-induced thrombosis of the 
abdominal aorta, both renal arteries, the 
right iliac artery, the profunda femoris 
artery, and the popliteal artery, in addition 
to having distal small vessel occlusion with 
rhabdomyolysis and renal failure. Aggressive 
surgical therapy was needed to preserve life 
and threatened tissues. When the cocaine- 
induced thrombosis threatens the distal 
part, emergency operation with thrombec- 
tomy is indicated. Alternatively, when the 
occlusion is not associated with distal 
ischemia, nonoperative therapy with a full 
course of heparinization will result in com- 
plete resolution of the cocaine-induced 
thrombus. One must be certain that such 
patients do not have other intracerebral 
embolic infarcts from prior heroin use 
before anticoagulation unless the hepari- 
nization lead to intracerebral hemorrhage. 
Fortunately, most patients do not combine 



cocaine and heroin or use them in temporal 
proximity. 



ACKNOWLEDGMENTS 



This work was supported by the Interstitial 
Fluid Fund (account 4-44966) . 



REFERENCES 

Fromm SH, Lucas CE: Obtura tor bypass for mycotic 
aneurysm in the drug addict. Arch Surg 
1970;100:82-83. 

Huebel H, Read C: Aneurysmal abscess. Minn Med 
1966;46:11-16. 

Johnson JR, Ledgerwood AM, Lucas CE: Mycotic 
aneurysm: New concepts in therapy. Arch Surg 
1983;118:577-582. 

Johnson JE, Lucas CE, Ledgerwood AM,Jacobs LA: 
Infected venous pseudoaneurysm: A complica- 
tion of drug addiction. Arch Surg 1984;1 19:1097- 
1098. 

Koch L: German "Ueber Aneurysma Dir Arteriae 
Mesenterichae Superioris, Inaug Dural-Abhand- 
lung." Erlangen J Barfus' Schen Universitates- 
Buchdruckerei 1851:5-23. 

Ledgerwood AM, Lucas CE: Mycotic aneurysm of 
the carotid artery. Arch Surg 1974;109:496-498. 

Osier W: The gulstonian lectures on malignant 
endocarditis. Br Med J 1885;1:467. 

Shanti CM, Lucas CE: Cocaine and the injured 
patient. Crit Care Med 2003;31: 

Wallace JR, Lucas CE, Ledgerwood AM: Social, eco- 
nomic and surgical anatomy of a drug-related 
abscess. Am Surg 1986;52(7):398-401. 

Webber J, Kline RA, Lucas CE: Aortic thrombosis 
associated with cocaine use: Reportof two cases. 
Ann Vase Surg 1999;13:302-304. 



ch22.qxd 4/16/04 3:28 PM Page 427 





Iatrogenic Vascular Trauma 



SAMUEL R. MONEY 
MICHAEL R. LEPORE, JR 



O INTRODUCTION 

O PERCUTANEOUS VASCULAR ACCESS 

Pseudoaneurysms 

Hemorrhage 

Arterial Closure Devices 

Central Venous Access 

Intra-Aortic Balloon Pump 
O INTRAOPERATIVE VASCULAR INJURIES 

Colorectal Procedures 

Pancreatobiliary Procedures 

Laparoscopic Procedures 

Vascular/Endovascular Surgery 

Orthopedic Surgery 

Neurosurgery 
O SUMMARY 



INTRODUCTION 



Patterns of injury for civilian arterial and 
venous trauma have long been recognized and 
discussed in both the vascular and the trauma 
literature. Most of the discussions are centered 



around arterial injuries resulting from either 
blunt or penetrating forces. These mecha- 
nisms are usually relatively easy to identify 
given the circumstances behind a motor 
vehicle accident or a fall, as is the case in blunt 
traumatic injury. Identification of the level 
of injury in the arterial tree, secondary to 

427 



ch22.qxd 4/16/04 3:28 PM Page 428 



428 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



penetrating trauma, necessitates thorough 
knowledge of arterial anatomy and recogni- 
tion of the possible pathway of injury. Iatro- 
genic patterns of injury are not generally 
considered in conjunction with vascular 
trauma. It should, however, be considered as 
a subheading both trauma and vascular surgery. 

These are the injuries that no clinician likes 
to collect or report in a personal series, but 
of which the vascular surgeon is well aware. 
Complications related to arterial access (e.g., 
pseudoaneurysms, hemorrhage, hematoma, 
and ischemia) or intra-aortic balloon pump 
(IABP) placement and venous access (e.g., 
venous injuries, arterial injuries, and arteri- 
ovenous fistula) are not uncommon. In addi- 
tion, there are patterns of arterial injury that 
are specific to different surgical procedures 
and subspecialties such as colorectal, pan- 
creatobiliary, laparoscopic, orthopedic, and 
neurosurgery. As enthusiasm for new proce- 
dures grows, such as endovascular aortic stent 
grafting for abdominal aortic aneurysms, a 
new pattern of iatrogenic arterial injuries 
evolves. 

We discuss and illustrate some of the more 
commonly encountered iatrogenic vascular 
injuries. Prevention and management of these 
injuries necessitates an understanding of the 
mechanism of injury, as it does with all forms 
of trauma. 



PERCUTANEOUS VASCULAR 
ACCESS 

Complications related to percutaneous vas- 
cular access are not a new phenomenon. 
Translumbar aortography was first introduced 
in the late 1920s and early 1930s by dos Santos. 
The next generation in the evolution of per- 
cutaneous access was fostered by Seldinger 
who introduced the concept of catheter 
exchange over a guidewire in 1953. Since that 
time, technology has continued to advance 
the field. Percutaneous arterial or venous 
access has become an almost routine part of 
clinical patient management. 

During the 1990s, there have been major 
technological advances in the treatment of 
peripheral vascular and coronary arterial 



disease. As a result of these new "endovascu- 
lar" techniques, various types of sheaths (i.e., 
crossover sheaths and shuttle sheaths) with 
increasing diameters have been developed 
to deliver newer and more complex intra- 
luminal devices for advanced endovascular 
procedures. Add the increasing use of anti- 
coagulants and the powerful antiplatelet 
agents (e.g., group Ilb/IIIa platelet receptor 
inhibitors and adenosine diphosphate [ADP] 
receptor inhibitors) and it is no surprise that 
the incidence of iatrogenic arterial and venous 
injuries has mirrored the enthusiasm and 
growth of percutaneous interventional/ 
endovascular techniques. 

The more complex interventions that 
require larger delivery systems (8- and 9- 
French sheaths) are mainly performed by 
femoral arterial approach. Fortunately, for the 
increasing number of patients undergoing 
these procedures, vascular occlusion and 
uncontrolled hemorrhage are the least 
common complications. Vascular occlusion 
secondary to thrombosis or dissection does 
occur but may be treated percutaneously, if 
recognized, as demonstrated in Figure 22-1. 
Most surgeons would agree that vascular 
occlusion or thrombosis that is not amenable 
to percutaneous therapy requires surgical 
exploration with treatment dictated by the 
respective etiology. 

Techniques for radial artery access are being 
used more frequently by interventional car- 
diologists, mainly for diagnostic coronary 
studies. As the emphasis toward endovascu- 
lar intervention continues, peripheral diag- 
nostic angiography and balloon angioplasty 
via radial arterial access are becoming more 
prevalent with lower profile percutaneous 
systems. These procedures are performed 
through a 6- or 7-French introducer sheath. 
As a result, such complications as infection, 
pseudoaneurysm, and thrombosis are not 
uncommon. Infection may occur and require 
some local drainage and antibiotics. Pseudo- 
aneurysm of the radial artery can be treated 
with similar techniques for femoral pseudo- 
aneurysm, discussed later discussion. Throm- 
bosis is typically well tolerated because the 
ulnar artery is usually the dominant artery of 
the hand. Thrombosis that leads to sympto- 
matic hand ischemia and/or major vascular 



ch22.qxd 4/16/04 3:28 PM Page 429 



22 • IATROGENIC VASCULAR TRAUMA 429 




■ FIGURE 22-1 

Left common iliac dissection secondary to a crossover sheath on right. Left common iliac post- 
stent placement on right. ■ 




■ FIGURE 22-2 

A, Wire in radial artery. B, Sheath and wire removed. C, Wire with piece of radial artery attached. 



injury (Fig. 22-2) requires immediate surgi- 
cal intervention using standard techniques of 
thrombectomy and or arterial reconstruction. 



Pseudoaneurysms 

Ever since Seldinger's technique for femoral 
arterial access became more commonplace, 



pseudoaneurysm has been recognized as one 
of the most frequently encountered compli- 
cations. Classic vascular surgical treatmenthas 
required open arterial repair, evacuation of 
the hematoma, and drain placement as 
needed. In turn, this necessitates further hos- 
pitalization for the patient and the accompa- 
nying discomfort inherent to recovery from 
surgery. Open repair still remains the 



ch22.qxd 4/16/04 3:28 PM Page 430 



430 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



standard of care for more complicated femoral 
arterial injuries; however, newer and less inva- 
sive techniques have been developed. 

The technique of ultrasound-guided 
pseudoaneurysm compression was introduced 
in the early 1990s. An experienced sonogra- 
pher is required to apply between 10 and 120 
minutes of compression to the "neck" of the 
pseudoaneurysm. Initial success rates for this 
procedure have been reported between 60% 
and 90%, with no further surgical interven- 
tion required. Unfortunately, recurrence rates 
have been reported at 25% to 30%. Many 
times, the pseudoaneurysm is tender and the 
patients require significant amounts of seda- 
tion before undergoing compression. In addi- 
tion, ultrasound-guided compression has even 
lower success rates for patients who are taking 
anticoagulation or antiplatelet agents. This 
represents a significant number of patients 
who undergo percutaneous procedures with 
concomitant cardiac and peripheral arterial 
disease. 

A combined modality using ultrasound 
guidance and thrombin injection has been 
proven more effective in the treatment of 
pseudoaneurysms. Success rates as high as 
96% have been reported even when nearly 
25% of the patients were anticoagulated. An 
experienced sonographer is still required to 
help identify the respective pseudoaneurysm 
in the appropriate axis for orientation. The 
needle is introduced under real-time ultra- 
sonography and the pseudoaneurysm is punc- 
tured directly with the needle. Appropriate 
orientation is crucial because the supplying 
artery should not be crossed (Fig. 22-3) . The 
thrombin (1000 IU) is then slowly injected 
into the pseudoaneurysm only, and throm- 
bosis can be seen immediately by ultrasound 
(Fig. 22-4) . Observing the ultrasound during 
injection ensures instillation of a minimal 
amount of thrombin. As thrombosis begins 
to occur, injection can be performed incre- 
mentally while the rest of the pseudoaneurysm 
thromboses. The greatest risk is from direct 
thrombin injection into the supplying artery, 
which occurs infrequently given adequate 
sonographic imaging. Many of the patients 
simply require some local anesthetic. This is 
performed as an outpatient procedure and 
the patients can walk 1 hour after injection. 



Common 
Femoral A. 




■ FIGURE 22-3 

Injection of common femoral artery from lateral 
approach to avoid intraluminal thrombin 
injection. ■ 



Newer echogenic needles can reportedly 
improve visualization of the needle, avoid- 
ing thrombin injection into the supplying 
artery. We simply use a 22-gauge spinal 
needle, moving the obturator in and out to 
improve ultrasonographic visualization before 
injection. 

Enthusiasts of endovascular therapy have 
reported on the use of covered stents to 
exclude a pseudoaneurysm or arteriovenous 
fistula. The early success rates were reason- 
able, with an 88% immediate result. On initial 
follow-up, however, there was nearly a 20% 
failure rate. The use of any stent, covered or 
uncovered, in the femoropopliteal region has 
been shown to have relatively poor results 
when compared to surgery; this is why covered 
stents should remain investigational in this 
anatomic region. 



Hemorrhage 

As discussed earlier, uncontrolled hemor- 
rhage from percutaneous access is not a 



ch22.qxd 4/16/04 3:28 PM Page 431 



22 • IATROGENIC VASCULAR TRAUMA 



431 




■ FIGURE 22-4 

Common femoral pseudoaneurysm on left with flow. Thrombosed pseudoaneurysm on right post- 
thrombin injection. ■ 



common phenomenon. The patientwho has 
obvious pulsatile bleeding from the puncture 
site or a growing hematoma should immedi- 
ately have the hemorrhage controlled with 
direct pressure on the artery proximal to the 
puncture, usually over the femoral head. Occa- 
sionally the puncture is too cephalad (exter- 
nal iliac) or the femoral artery is damaged 
too severely for simple compression. This may 
be the case in the presence of severe arterial 
laceration or avulsion. Operative repair using 
the standard vascular principles of adequate 
exposure and proximal/distal vascular control 
should be performed immediately. 

The more dangerous scenario is that of 
insidious and unrecognized hemorrhage. 
Patients who have uncontrolled bleeding 
without the obvious stigmas (e.g., pulsatile 
bleeding and expanding hematoma) initially 
exhibit very subtle clinical signs of hemor- 
rhage. Relative hypotension and mild tachy- 
cardia that transiently improves with 
administration of fluids should alert the astute 
clinician and necessitate further investigation. 
Once suspected, a decreasing hemoglobin 
level verifies the likelihood of a retroperitoneal 
hematoma. Once again, the location of the 
puncture may provide some clues (i.e., above 
the inguinal ligament) to ongoing bleeding. 
An abdominal computed tomographic (CT) 
scan will verify the presence, location, and size 



of the retroperitoneal hematoma (Fig. 22-5) . 
Surgical exploration should rarely be 
required, as the retroperitoneum serves to 
tamponade the bleeding. Adequate resusci- 
tation, reversal of any underlying coagulopa- 
thy, and identification and correction of 
medications that may exacerbate the bleed- 
ing (e.g., Ilb/IIIa antiplatelet agents) should 
be first and foremost before any surgical explo- 
ration is performed. 



Arterial Closure Devices 

Given the growing field of endovascular tech- 
niques, combined with the accompanying 
increase in percutaneous access complica- 
tions, industry has answered with newer prod- 
ucts in an attempt to decrease access 
complications and reduce personnel time 
holding pressure on groins. The goal is to 
achieve immediate hemostasis following per- 
cutaneous arterial access. Different devices 
have been designed, each with its own tech- 
nique toward achieving hemostasis. One 
approach is to "plug" the hole using collagen- 
based materials. Another technique involves 
closing the site with a suture. Still, one other 
involves using chemicals (procoagulant) to 
initiate early hemostasis. The development 
of these devices has created a new set of 



ch22.qxd 4/16/04 3:28 PM Page 432 



432 



V • SPECIAL PROBLEMS AND COMPLICATIONS 




■ FIGURE 22-5 

Computed tomographic scan of retroperitoneal hematoma (H). 




■ FIGURE 22-6 

Angio-Seal device in vessel and post-deployment with intraluminal anchor in position. 



complications. Treating any of the respective 
complications requires some understanding 
of how each device functions. 

Angio-Seal (Sherwood Davis and Geek, St. 
Louis, Missouri) makes use of the collagen 
plug philosophy. The plug is sutured to a small, 
flat, rectangular anchor that is deployed intra- 
luminally (Fig. 22-6). The plug is "tamped" 
down and secured extraluminally for near- 
immediate hemostasis. Initial success rates 
have been reported in the range of 88% to 
92 %. Success rates are claimed to improve with 
experience, as the manufacturers report an 
inherent learning curve. Given the typical 
nature of femoral vessels in this patient pop- 
ulation, leaving anything intraluminally is 
disturbing. Rates of infection, stenosis, and 



vascular occlusion or acute ischemia have been 
reported to be 2% to 3%. The risk of infec- 
tion seems high when compared to simple 
manual compression. However, it continues 
to be used by many interventionalists in con- 
junction with prophylactic antibiotics (not a 
recommendation of the manufacturer). 

The collagen plug technology is shared by 
VasoSeal (Datascope, New Jersey) but in a 
different manner. This device requires pre- 
dilation with measurements of the length of 
the subcutaneous tract. The plug is then 
deployed extraluminally (Fig. 22-7) and pres- 
sure is applied for 2 to 3 minutes. Immediate 
success, defined as hemostasis, was achieved 
in 87% to 95% of the patients. The hematoma 
rate was alarmingly high, at 21% with 



ch22.qxd 4/16/04 3:28 PM Page 433 



22 • IATROGENIC VASCULAR TRAUMA 



433 








A B 

■ FIGURE 22-7 

VasoSeal device with extraluminal placement of collagen plug. 




■ FIGURE 22-8 

Duett device illustrating intraluminal balloon inflated and procoagulant injected after compression 
on right. ■ 



1% requiring surgery. If the plug is not ade- 
quately placed, then the vessel continues to 
bleed into the surrounding tissue. This would 
explain the decreased success rate (58.8%) 
in obese (>90-kg) patients. The device did not 
perform as well for patients on anticoagulants 
or antiplatelet therapy, as success rates were 
79%. In addition, embolization of the colla- 
gen plug, late bleeding, and infection have 
required surgical intervention in as many as 
5% of patients. 

The Duett device (Vascular Solutions, Inc., 
Minneapolis, Minnesota) makes use of a pro- 
coagulant to seal the arterial puncture site. A 
small balloon is inflated on the luminal side 



of the puncture site, to avoid introduction of 
material into the respective vessel, and pro- 
coagulant is injected (Fig. 22-8). Once the 
balloon is deflated, the device is removed and 
2 minutes of manual compression is required. 
A European multicenter registry reported a 
96% deployment rate with successful hemo- 
stasis in 2 to 5 minutes in 95% of the patients 
with the Duett device. The overall complica- 
tion rate was 2.6%, including pseudo- 
aneurysms and complete arterial occlusions. 
Surgical intervention was required in fewer 
than 1% of patients. The use of anticoagu- 
lants or antiplatelet agents was not an exclu- 
sion criterion for the study. 



ch22.qxd 4/16/04 3:28 PM Page 434 



434 V • SPECIAL PROBLEMS AND COMPLICATIONS 




■ FIGURE 22-9 

A, Perclose device in lumen of the vessel. B, Post-deployment with suture in place. 




■ FIGURE 22-10 

A, Angiogram of introducer sheath entering the left common femoral artery just proximal to the 
profunda femoris artery. 6, Angiogram exhibiting high-grade stenosis of common femoral and 
profunda femoris arteries. ■ 



The last, and probably most frequently used, 
device uses suture to close the puncture site. 
Perclose (Perclose Inc., Menlo Park, Califor- 
nia) uses two needles and a preloaded suture 
to puncture the vessel in a cephalad to caudad 
orientation and close the puncture site (Fig. 
22-9) . The ends of the suture come out 
through the device and are tied extracorpo- 
really. A knot pusher then slides the knot down 
to the vessel. Early success rates are reported 
at 85% to 90%. Complications are reported 
at 1.8% and relate to inadequate deployment 
or pseudoaneurysm formation. Device failures 



are usually recognized immediately for lack 
of hemostasis. Given the size of the needles, 
the device does not function on thick calci- 
fied vessels. We have seen delayed injuries, with 
development of severe claudication (Fig. 
22-10), which ultimately leads to common 
femoral endarterectomy and profundoplasty. 
The aforementioned devices are more fre- 
quently being used and this trend will con- 
tinue. Aside from the acute need for surgical 
intervention, some delayed complications 
exist that may mandate surgical intervention. 
Although none of the studies discuss any of 



ch22.qxd 4/16/04 3:28 PM Page 435 



22 • IATROGENIC VASCULAR TRAUMA 435 



the local tissue changes, we have experiences 
with all of them at our institution. The sur- 
rounding scarring and inflammation encoun- 
tered when one of these devices has been used 
is similar to that seen in a "reoperative groin." 



Central Venous Access 

Venous access by either the internal jugular 
or the subclavian vein approach has become 
a frequently performed procedure, with more 
than 3 million central venous catheters 
inserted annually. The obvious risk of pneu- 
mothorax and/or tension pneumothorax is 
a well-known complication of this procedure. 
Prevention of complications requires strict 
adherence to anatomic landmarks. Most 
venous injuries will respond to manual digital 
compression for hemostasis because it is a low- 
pressure system. The most common compli- 
cations, however, are from injuries to adjacent 
structures. 

The internal jugular vein can be 
approached from either side of the neck. It 
is one of the easiest veins on which to obtain 
digital compression. The more common sig- 
nificant injury in this location involves the 
carotid artery, given its proximity (Fig. 22-11). 
Although it is a higher pressure vessel, again, 
digital control is relatively easy here. The 
serious injuries occur with laceration of the 
vessel or when unrecognized cannulation of 
the artery occurs with subsequent large-bore 
dilation for placement of a resuscitation 
catheter (12 French) or a cordis (8 to 10 
French). Additionally, unrecognized cannu- 
lation of the internal jugular vein through a 
portion of the artery may lead to an arteri- 
ovenous fistula (Fig. 22-12). 

Subclavian venous access has the greater risk 
of morbidity and mortality. It is more difficult 
to apply digital pressure to the subclavian vein 
or the subclavian artery, given their relative 
anatomic location posterior to the clavicle 
(Fig. 22-13). Cannulation of the right sub- 
clavian vein is felt to be potentially more 
hazardous secondary to its abrupt angulation 
into the superior vena cava. Passage of the 
dilator can lacerate or perforate the vein or 
the superior vena cava, with an incidence 
reported as high as 1% of the time, leading 




■ FIGURE 22-11 

Illustration depicting carotid artery and internal 
jugular vein relationships. ■ 



to exsanguination and possible death. Passage 
through the artery into the vein can lead to 
arteriovenous fistula (Fig. 22-14) formation 
as well. Aortic perforation and subclavian 
artery aneurysm, though not common, have 
been reported as a consequence of central 
venous access as well. 



Intra-Aortic Balloon Pump 

The IABPwas first instituted, clinically, 30years 
ago, and it has become the most widely applied 
mechanical circulatory assist device, inserted 
in 2% to 12% of all patients, as an adjunct to 
heart surgery. However, it has its own set of 
accompanying vascular complications. Com- 
plications have been reported to occur 
between 12% and 30% of the time. 

The most commonly encountered compli- 
cation is that of ipsilateral lower extremity 
ischemia. Although most patients will improve 
with simple removal of the balloon, some 
patients still require surgical intervention even 



ch22.qxd 4/16/04 3:28 PM Page 436 



436 



V • SPECIAL PROBLEMS AND COMPLICATIONS 




■ FIGURE 22-12 

A, Duplex ultrasound of carotid/internal jugular vein fistula from access injury. 6, Corresponding 
angiogram of fistula filling vein. ■ 



Subclavian A 
Subclavian V. 




■ FIGURE 22-13 

Illustration depicts location of subclavian vein 
and artery behind the clavicle. ■ 



*■•- * 


■ *' ! 


< 






mwm 


<►„■ ^^__ 

Subclavian AV Fistula 



■ FIGURE 22-14 

Duplex ultrasound of subclavian vein/artery 
fistula secondary to access. AVF, arteriovenous 
fistula; SCA, subclavian artery; SCV, subclavian 
vein. ■ 



after it is removed. Other patients are just not 
stable enough to have the balloon removed 
and may need contralateral placement. Ipsi- 
lateral iliac dissection, thrombosis of 
iliac /femoral arteries, and distal embolization 
are the most frequently encountered arterial 
pathology. A dissection may be treated by 



endovascular techniques and stenting. Throm- 
bosis requires immediate surgical attention 
and intraoperative decisions dictated by the 
anatomic location of occlusion. Distal 
embolization may occur and the severity of 
distal embolization will dictate treatment. A 



ch22.qxd 4/16/04 3:28 PM Page 437 



22 • IATROGENIC VASCULAR TRAUMA 



437 



patient who experiences acute popliteal occlu- 
sion leading to a nonviable leg should undergo 
immediate exploration, thrombectomy, and 
potentially bypass. In contrast, patients with 
"blue toes" and intact pulses are best observed 
for further tissue demarcation and improve- 
ment. Luckily, aortic perforation does not 
occur frequently (<1%) as it carries a near- 
100% mortality rate in this population. 



INTRAOPERATIVE VASCULAR 
INJURIES 



Vascular injuries that occur as a consequence 
of another operative intervention are more 
prevalent than is reported. Not many surgeons 
are willing to report on their series of intra- 
operative vascular injuries. However, it is well 
known that certain operations and different 
procedures have inherent risks of vascular 
injuries. The injuries that require intraoper- 
ative, urgent vascular consultation are usually 
severe and life threatening, as most surgeons 
will deal with the less severe injuries 
themselves. 



Colorectal Procedures 

Many colorectal operations require dissection 
into the pelvis. The low anterior resection, 
total proctocolectomy, abdominoperineal 
resection, and especially complicated diver- 
ticulitis place iliac vascular structures at risk. 
The arterial injuries are usually fairly simple 
to recognize and repair following standard vas- 
cular surgical principles. However, venous 
injuries to the iliac veins or inferior vena cava 
may be more challenging and can lead to sig- 
nificant blood loss. Simple ligation or over- 
sewing of the bleeding may slow the bleeding 
enough that compression or packing of the 
pelvis may stop the hemorrhage. By the time 
a vascular surgeon is called, the patient has 
usually bled significantly. Initial packing of the 
pelvis, while resuscitation and correction of 
an underlying coagulopathy can begin, is the 
most prudent first step in this situation. Once 
corrected, the packing can be removed in a 
systematic fashion to identify the source of 



bleeding for subsequent repair. All venous 
repairs should use pledgeted sutures of fine 
Prolene. 



Pancreatobiliary Procedures 

Pancreatobiliary operations can be difficult 
without a thorough understanding of the 
anatomy of the region. The pancreatobiliary 
structures lie in a peritoneal and retroperi- 
toneal location (Fig. 22-15) that places them 
in proximity to the superior mesenteric, 
splenic, renal, and portal veins, as well as the 
superior mesenteric, celiac, splenic, and 
hepatic arteries, in addition to the vena cava 
and aorta. Even though these vascular injuries 
are not often reported, it has been estimated 
that they may approach 4%. 

Injuries of aforementioned vessels occur 
most frequently to the portal vein, followed 
by superior mesenteric vein, right hepatic 
artery, splenic vein, superior mesenteric artery 
(SMA), and common hepatic artery. Injury 
patterns will be dictated by the types and fre- 
quency of the pancreatobiliary operation 
(e.g., Whipple versus laparoscopic cholecys- 
tectomy) performed. Sometimes these vas- 
cular injuries are recognized only after the 
retractors have been pulled back and the 
bowel is ischemic (SMA injury) . 

In the trauma setting of a severely injured 
patient, many injuries may be ligated or over- 
sewn. However, elective pancreatobiliary 
surgery often destroys the collateral connec- 
tions that are required to maintain viability 
after simple ligation. For this reason, it is advis- 
able to repair these injuries when recognized 
and feasible. Venous injuries may require 
simple venorrhaphy or mobilization and 
primary anastomosis. In the case of arterial 
injuries, primary repair is the goal, but not 
always possible given the type of resection and 
length of remaining artery. Autogenous recon- 
struction with saphenousvein is our preferred 
method of repair in this setting. 



Laparoscopic Procedures 

Laparoscopic surgery has become routine 
in surgical practice. Some of the early 



ch22.qxd 4/16/04 3:28 PM Page 438 



438 



V • SPECIAL PROBLEMS AND COMPLICATIONS 




■ FIGURE 22-15 

Pancreatobiliary anatomic relationships and in cross section. Ao, aorta; IVC, inferior vena cava; 
SMA, superior mesenteric artery; SMV, superior mesenteric vein. ■ 



complications from this "minimally invasive" 
technique have completely disappeared. 
However, some of the vascular complications 
have persisted over theyears. The mechanism 
of injury has not changed, even today. 

The overall vascular complication rate is 
quite low (0.08% to 0.1%) when compared 
with that of many other operations. Most com- 
monly, vascular injuries reported in the largest 
series continue to be to the distal abdominal 
aorta, iliac arteries, inferior vena cava, and iliac 
veins. These structures are susceptible to 
injury from introduction of the Veress needle, 
for blind abdominal insufflation, or trocars 
into the lower abdomen. The distance, after 
compression of the periumbilical region with 
a Veress needle or trocar, between the abdom- 
inal wall and the vessels during insertion 
is generally not appreciated by the inex- 
perienced laparoscopist (Fig. 22-16). The 
result is major vascular injury. It is usually 



immediately recognized though, requiring 
conversion to open laparotomy with repair of 
the respective vascular injury. 



Vascular/Endovascular Surgery 

Iatrogenic vascular injuries during vascular 
procedures are again an area that is likely un- 
reported for two reasons. The first is that the 
vascular surgeon is going to repair the injury 
at the time it occurs and the second involves 
the lack of willingness to report vascular com- 
plications. Nonetheless, some well-established 
injuries are known to be associated with dif- 
ferent operations and have been discussed for 
many years. For example, iliac vein injury 
during aortobiliary bypass for aneurysmal 
disease. Endovascular surgery has borne out 
a whole new set of complications relative 
to aortic-stent grafts that are still in the 



ch22.qxd 4/16/04 3:28 PM Page 439 



22 • IATROGENIC VASCULAR TRAUMA 



439 




■ FIGURE 22-16 

Iliac artery injury from Veress needle 
placement. ■ 

discovery phase. The complications associated 
with percutaneous access have been discussed 
at length. 

Thoracoabdominal aortic operations are 
generally recognized, by most, as one of the 
highest risk operations a vascular surgeon 



performs. The large experience of E. Stanley 
Crawford and colleagues at Baylor /Methodist 
set the standard for this procedure. Respira- 
tory failure, renal failure, cardiac complica- 
tions, and stroke have all been well established 
and are beyond the scope of this chapter. Thor- 
ough familiarity with retroperitoneal anatomy 
is imperative if one is to perform these oper- 
ations safely. Some of the most dangerous 
bleeding involves the network of veins that 
should be avoided while approaching the 
aorta. Examples include the lumbar and 
gonadal veins behind the left kidney (Fig. 
22-17) and the azygous system. As in trans- 
abdominal aortic operations, the vena cava 
and iliac veins can lead to massive exsan- 
guination and even intraoperative death if 
injured. These vessels are difficult to control 
and should be packed/compressed initially. 
If the injury is too large, then adequate expo- 
sure with ligation and/or oversewing still 
remains the standard treatment for these 
dreaded injuries. 

Endovascular aortic aneurysm repair is 
still in its infancy in the United States. The 
Europeans, however, have been using multi- 
ple devices since the early 1990s. Certain com- 
plications such as iliac limb occlusion, distal 
migration, and device malfunction are 




Aortic 
Aneurysm 



Gondal V. — ' > — Lumbar ' 

■ FIGURE 22-17 

Gonadal and lumbar veins to be avoided during thoracoabdominal exposure of the aorta. 



ch22.qxd 4/16/04 3:28 PM Page 440 



440 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



specific to different devices, the subject of 
another chapter. Identification of common 
vascular complications to the procedure itself 
is more important. The Eurostar Collabora- 
tors, 56 European centers, identified a 3% vas- 
cular complication rate in more than 1500 
patients. These complications were identified 
as (1) arterial rupture, perforation, or dis- 
section; (2) thrombus, obstruction, or steno- 
sis; (3) embolization; (4) occlusion of renal 
artery; and (5) other injuries. This does not 
include the need for open conversion, which 
was required in 2.5% of the patients, or late 
ruptures as a result of endoleaks. 

The Fogarty balloon catheter was first 
introduced in 1963. Although it is an instru- 
mental part of the vascular surgical arma- 
mentarium, some potential complications 
are associated with its use. The most common 
injuries that have been documented include 
(1) perforation of the artery by the catheter 
tip, (2) rupture of the artery resulting from 
overinflation of the balloon, (3) disruption 
or injury to the arterial intima, (4) emboliza- 
tion of fragments of the ruptured balloon or 
catheter tip, (5) arteriovenous fistula, and (6) 
pseudoaneurysm formation. 

Many of these injuries are the result of 
overzealous inflation and thrombectomy by 
the operator, which can be avoided as follows: 

(1) Before using the balloon in the vessel, 
place a 3-way stopcock on the end of the 
catheter to inflate the balloon to the proper 
volume (size) under direct visualization, then 
turn the stopcock to expel the excess saline; 

(2) tactile feel of catheter resistance to with- 
drawal in conjunction with careful control of 
the syringe to avoid overinflation; (3) if sig- 
nificant resistance is encountered and the 
anatomy unclear, then dilute the syringe with 
some contrast and monitor balloon passage 
fluoroscopically. Another method would be 
to simply proceed with an intraoperative 
angiogram and use an "over-the-wire" balloon 
thrombectomy catheter. 



Orthopedic Surgery 

Common vascular injuries that occur sec- 
ondary to orthopedic injuries such as frac- 
tures and dislocations have been well 
recognized and reported. Given the nature 



of a subspecialty that requires placement of 
rods and screws for fixation of fractures, it is 
amazing that iatrogenic vascular injuries are 
not more common. Aside from complete mis- 
placement of a rod, the most common injuries 
are reported with arthroplasty. 

Total knee arthroplasty has been performed 
for more than 30 years now. The incidence of 
vascular injury is reported to be as low as 
0.03%. This operation is typically performed 
under tourniquet control, so the injury may 
not be recognized initially. The mechanism 
of injury, when direct trauma is not involved, 
has been theorized to be secondary to arter- 
ial stretching or disruption of arterial plaques 
from the tourniquet placement. A thorough 
preoperative vascular assessment for reference 
is not always available when one is consulted 
on these patients acutely. In this case, the best 
approach is to determine the viability of the 
affected leg and compare it to the uninvolved 
extremity as a baseline. The most common 
complication is that of acute occlusion of the 
popliteal artery, requiring immediate explo- 
ration with thrombectomy or potential bypass. 

One of the other well-established vascular 
injuries occurs secondary to screw placement 
for total hip arthroplasty. The vascular injuries 
that have been reported are related to 
intrapelvic extrusion of cement or damage to 
the common iliac vein during reaming for 
prosthesis placement. Orthopedic surgeons 
are well acquainted with the structures that 
are anterior, superior, and posterior to the 
acetabulum but are relatively unaware of those 
that lie medial to it. During the developmental 
period of total hip arthroplasty, after some cat- 
astrophic complications, it became widely rec- 
ognized that the screws for the acetabular 
component placed medial structures at risk. 
Medial to the acetabulum lie the external iliac 
vein, obturator artery, and obturator vein. The 
anterior part of the acetabulum became rec- 
ognized as the danger zone with the highest 
risk for vascular injury during screw place- 
ment. Through education, this complication 
has been reduced dramatically. 



Neurosurgery 

Neurosurgical vascular emergencies are not 
common. Intracranial vascular complications 



ch22.qxd 4/16/04 3:28 PM Page 441 



22 • IATROGENIC VASCULAR TRAUMA 



441 



will be primarily handled by the neurosurgeon 
and obviously not the vascular surgeon. Back 
operations, such as disk surgery or corrective 
scoliosis surgery, have the highest potential 
for iatrogenic vascular complications in neu- 
rosurgery. Typically, large multilevel spine 
operations require adequate exposure. It is 
not uncommon for a general surgeon or a vas- 
cular surgeon to provide anatomic exposure 
for the neurosurgeon. 

In the case of scoliosis surgery, prevention 
of major vessel injury during anterior expo- 
sures to the spine is of major concern. Given 
their anatomic proximity to the spine, the 
aorta and vena cava are at greatest risk. Typ- 
ically, laceration or avulsion type of injuries 
occur secondary to rigorous retraction. 
Penetrating injuries may occur during 
removal of the rim of the disk annulus by the 
neurosurgeon. The recommended preven- 
tive technique is for placement of an elevator 
between the vessels and the spinal column 
during disk removal. Appropriate-length 
screws will avoid further vascular injury as 
well. However, late hemorrhage resulting 
from erosion, leakage, or false aneurysm 
of adjacent vessels has been reported. 
Retroperitoneal exposure to the spine is 
fraught with the same hazards as those dis- 
cussed earlier with relation to thoraco- 
abdominal exposure. The lumbar veins and 
arteries are the most commonly injured 
vessels because of avulsion or laceration. 
Digital pressure should be attempted ini- 
tially, followed by suture ligation if unsuc- 
cessful. These can be troublesome injuries 
that result in a significant amount of blood 
loss. 

Vascular injuries in lumbar disk surgery 
are rare (0.05%) but serious complications. 
They may be delayed in presentation or dif- 
ficult to recognize given the anatomic 
approach. The most commonly seen vascular 
injuries are lacerations of the iliac veins, 
lumbar veins, abdominal aorta, median 
sacral artery, and arteriovenous fistulas. 
Because of the relative rarity of these 
injuries, no large series has been published 
that discusses the surgical approach to any of 
these injuries. It is important to keep the pos- 
sibility in the back of one's mind, when 
recent back surgery has been performed, 
that the potential for vascular injury exists. 



Early recognition of these injuries will help 
avoid unrecognized and ongoing hemor- 



rhage. 



SUMMARY 



Any busy vascular surgeon is fully aware that 
a number of iatrogenic vascular injuries occur 
with varying frequencies based on anatomic 
location. However, reliable data about to the 
true incidence of these complications are not 
easily obtained. As previously discussed, this 
is not a series that any surgeon in any sub- 
specialty would like to collect and report. The 
incidence and prevalence of iatrogenic vas- 
cular injuries are likely even higher than has 
been reported. 

Our population continues to age and the 
greater percentage of the population will be 
older than 55 years within 10 years. That trans- 
lates into more general, vascular, and cardio- 
vascular disease. In turn, a greater number of 
operations are likely to be performed in all 
of the surgical subspecialties. As technologic 
advances continue to develop more devices 
for performance of endovascular techniques, 
there will continue to be a concomitant 
increase in the iatrogenic vascular injuries that 
accompany these techniques. The vascular 
surgeon will need to be well trained to handle 
different types of injuries that will continue 
to evolve with advancing technology. This will 
require a current knowledge base regarding 
to this ever-changing and rapidly developing 
technology. 



REFERENCES 

Arafa OE, Pedersen TH, Svennevig JL, et al: Vas- 
cular complications of the Intraaortic balloon 
pump in patients undergoing open heart oper- 
ations: 15-year experience. Ann Thorac Surg 
1999;67:645-651. 

Bridwell KH, DeWald RL: The Textbook of Spinal 
Surgery, 2nd ed. Philadelphia, Lippincott — 
Raven Publishers, 1997. 

Buth J, Laheij RJF, et al: Early complications and 
endoleaks after endovascular abdominal aortic 
aneurysm repair: Report of a multicenter study. 
J Vase Surg 2000;31:134-146. 



ch22.qxd 4/16/04 3:28 PM Page 442 



442 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



Cikrit DF, Dalsing MC, Sawchuk AP, et al: Vascular 
injuries during pancreatobiliary surgery. Am 
Surg 1993;59:692-697. 

Fruhwith J, Koch G, Mischinger HJ, et al: Vascular 
complications in minimally invasive surgery. Surg 
Laparosc Enclose 1997;7(3):251-254. 

Gonze MD, Sternbergh WC II, Salartash K, et al: 
Complications associated with percutaneous 
closure devices. Am J Surg 1999;178:209-211. 

Goyen M, Manz S, Kroger K et al: Interventional 
therapy of vascular complication caused by the 
hemostatic puncture closure device Angio-Seal. 
Cathet Cardiovasc Intervent 2000;49:142-147. 

Kang SS, Labropoulos N, Mansour MA, et al: 
Expanded indications of ultrasound-guided 
thrombin injection of pseudoaneurysms. J Vase 
Surg 2000;31:289-298. 

Keating ME, Ritter MA, Faris PM: Structures at risk 
from medially placed acetabular screws. J Bone 
Joint Surg 1990;72-A(4):509-511. 

Lazarides MK, Tsoupanos SS, Georgopoulos SE, et 
al: Incidence and patterns of iatrogenic arterial 



injuries. Adecade's experience. J Cardiovasc Surg 
1998;39:281-285. 

Menlhorn U, Kroner A, de Vivie ER: 30 years clin- 
ical intra-aortic balloon pumping: Facts and 
figures. Thorac Cardiovasc Surg 1999;47 
(Suppl):298-303. 

Robinson JF, Robinson WA, Cohn A, et al: Perfo- 
ration of the great vessels during central venous 
line placement. Arch Intern Med 1995;155:1225- 
1228. 

Silber S, Tofte AJ, Kjellevand TO, et al: Final report 
of the European multi-center registry using the 
Duett vascular sealing device. Herz 1999;24(8): 
620-623. 

Sorell KA, Feinberg RL, Wheeler JR, et al: Color- 
flow duplex-directed manual occlusion of 
femoral false aneurysms. J Vase Surg 1993;1 7:571- 

577. 

Svensson LG, Crawford ES, Hess KR, et al: Expe- 
rience with 1509 patients undergoing thora- 
coabdominal operations. J Vase Surg 1993; 
17:357-370. 



ch23.qxd 4/16/04 3:27 PM Page 443 





Compartment Syndromes 



THOMAS S. GRANCHI 
PRISCILLA GARCIA 
KENNETH L. MATTOX 
MICHAEL E. DEBAKEY 





o 


PRESENTATION 




o 


ANATOMY 

Calf 

Thigh 

Arm/Hand 

Abdomen 

Spinal Cord 

Pericardium 




o 


DIAGNOSIS 

Extremity Compartment Measurements 

Noninvasive Assessment of Compartment Compromise 

Laboratory Evaluation 

Pathophysiology 


o 


TREATMENT OF EXTREMITY COMPARTMENT SYNDROME 




Technique of Decompression 


o 


SUMMARY 



443 



ch23.qxd 4/16/04 3:27 PM Page 444 



444 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



Compartment syndrome occurs when 
pressure in a rigid compartment 
exceeds perfusion pressure. It can 
occur in any limb, the anterior chamber of 
the eye, the spinal canal, the pericardium, or 
in the abdomen and is seen in many clinical 
settings, threatening life, limb, sight, and/or 
neurologic function. Despite technology 
advances in noninvasive measurements 
and a better understanding of the anatomy, 
biology, and chemistry of reperfusion injury, 
diagnosis of this complication may still be 
missed or delayed. Operative decompression 
of the compartment is the mainstay of treat- 
ment. Fasciotomy treats compartment syn- 
drome in limbs, laparotomy is the treatment 
for abdominal compartment syndrome, peri- 
cardiotomy treats cardiac tamponade, and 
spinal canal decompression has been used to 
treat reperfusion-related spinal compartment 
syndrome. For these reasons, compartment 
syndrome poses many potential pitfalls for 
trauma, vascular, and orthopedic surgeons. 
Medical treatments to reduce swelling and 
protect against cellular injury may have 
adjunct roles but do not replace the timely 
operative decompression. 

Common clinical presentations include 
reperfusion injury after blunt trauma, vascu- 
lar injury and repair, closed fractures, and elec- 
trical injuries. Additionally, compartment 
syndromes have been reported following use 
of compressive devices, such as the military 
antishock trousers. In these clinical settings, 
the astute clinician suspects compartment syn- 
drome on initial physical examination and 
must then develop diagnostic and therapeu- 
tic actions, to include serial examinations 
of the involved region or extremity, serial 
pressure measurements, and/or use of new 
machines that evaluate tissue perfusion. 

Excessive pain and loss of motor and 
sensory function in a limb are late clinical 
findings. In patients with a high risk for com- 
partment syndrome, but in who repeated 
examination is not feasible, immediate com- 
partment pressure measurement or immedi- 
ate prophylactic fasciotomies should be 
considered. Pressure measurements can 
be graded, but a direct pressure of 25 cm H 2 
or a pressure differential between mean arte- 
rial pressure and compartment pressure of 



more than 50mmHg indicates the need for 
immediate decompression. 

Pericardial compartment syndrome is asso- 
ciated with increasing pressures in the peri- 
cardial sac and can be secondary to venous, 
arterial, or cardiac injury. Beck's triad of ele- 
vated central venous pressure, hypotension, 
and muffled heart sounds is a late manifes- 
tation of the pericardial compartment 
syndrome. Early detection of post-traumatic 
hemopericardium should lead to immediate 
thoracic decompression before the late man- 
ifestations develop. Narrowed pulse pressure 
and cardiac arrest from pericardial tampon- 
ade are very late manifestations and usually 
occur in patients with multisystem injury 
where the attention of the examining physi- 
cian has been diverted, causing delayed 
or missed diagnosis of the compartment 
syndrome. 

Spinal compartment syndrome has been 
evaluated most often in patients undergoing 
operation for extensive thoracoabdominal 
aortic surgery, where drainage of cere- 
brospinal fluid and reducing the spinal canal 
pressure are performed to reduce the inci- 
dence of paraplegia. Post-traumatic paraple- 
gia, even associated with treatments of blunt 
injury of the descending thoracic aorta, has 
not been treated with spinal canal decom- 
pression. However, because paraplegia is 
associated with increased pressures in a 
closed compartment, decreasing pressure dif- 
ferentials, spinal cord swelling, and ischemia/ 
reperfusion conditions undoubtedly con- 
forms to the definition of a compartment syn- 
drome. Further research in patients with 
thoracic aortic injury, post-traumatic para- 
plegia, and direct spinal column injury is 
required to define the post-traumatic spinal 
cord compartment syndrome. One might raise 
the argument that any potential value of use 
of corticosteroids in paraplegia following 
blunt injury to the spinal cord is actually 
an attempt to treat a spinal compartment 
syndrome. 

Abdominal compartment syndrome can 
occur in patients with intra-abdominal injuries 
and hemorrhagic shock but is not directly 
related to these conditions. Abdominal com- 
partment syndrome also occurs in some 
patients with no abdominal injury but who 



ch23.qxd 4/16/04 3:27 PM Page 445 



23 • COMPARTMENT SYNDROMES 



445 



have treatment for remote conditions, such 
as cardiopulmonary bypass and excessive 
fluid resuscitation. In the abdomen, elevated 
compartment pressure is manifested by the 
following triad: 

• Oliguria 

• Reduced cardiac output that does not 
improve with intravascular fluid 
replacement 

• Increased airway pressures 

Organ impairment and increased airway 
pressure can be detected at intra-abdominal 
pressures as low as 1 5 mm Hg. At 25 to 30 mm 
Hg, organ failure is evident and immediate 
laparotomy should be performed (Burch and 
colleagues, 1996) . Measuring intra-abdominal 
pressures will confirm an already suspected 
clinical diagnosis. 



PRESENTATION 

Surgeons caring for trauma patients most com- 
monly diagnose and treat compartment syn- 
dromes, because this condition is often seen 
in association with injury. Pediatric, ortho- 
pedic, plastic, replantation, microvascular, 
vascular, and thoracic surgeons also often 
encounter compartment syndromes. Arterial 
or venous occlusion followed by reperfusion 
injury is a common presentation, whether 
occlusion is secondary to injury or vascular 
control during attempted reconstruction. 
Long bone fractures often precipitate com- 
partment syndrome because of hematoma and 
tissue swelling at the site. Traditionally, calf 
compartment syndromes have been the most 
commonly diagnosed, treated, and reported. 
Gulli and Templeton (1994) report that com- 
partment syndrome occurs in 3% to 17% of 
closed tibia fractures. Compartment syn- 
drome associated with femur injuries is rare 
if the fracture occurs at the shaft and absent 
associated vascular injuries (Schwartz and 
colleagues, 1989; Russel and colleagues, 2002) . 
Compartment syndrome occurring in the 
thigh is often overlooked because of other life- 
threatening injuries that distract the surgeon. 



Pericardial compartment syndromes are often 
detected during the surgeon performed 
ultrasound examination in the emergency 
center. Rarely, a patient with unexplained 
continuing hypotension in the operating 
room following laparotomy will be found to 
have an occult pericardial compartment 
syndrome. 

Although most cases of compartment 
syndrome from vascular etiologies occur with 
arterial injuries, it can also occur with venous 
pathology. There are many reports of com- 
partment syndrome occurring with phleg- 
masia cerulea dolens (Dennis, 1945; Cywes 
andLouw, 1962; Wood and colleagues, 2000). 
Venous bleeding in the calf, thigh, abdomen, 
arm, and neck has also produced compart- 
ment syndromes. The individual fascial and 
muscle compartments in each of these areas 
deserve careful attention. 

In the upper arm and forearm, compart- 
ment syndrome may occur with supracondy- 
lar humerus fractures, intravenous drug 
abuse, electrical injuries, intravenous line 
insertion site complications, prolonged 
tourniquetuse, and even weight lifting (Moore 
and Friedman, 1989) . Historically, when home 
washing machines had mechanical wringers 
attached to the machine, children getting 
arms caught in the wringer was an extremely 
common cause of both humeral fracture and 
compartment syndrome, known then as 
Volkmann 's ischemic contracture. Many of these 
patients will present in ambulatory settings, 
where the index of suspicion may be low. Deep 
pain and tense swelling of the limb should 
prompt further investigation. 

Abdominal compartment syndrome often 
develops in trauma patients who have under- 
gone recent laparotomy and been excessively 
resuscitated for hemorrhagic shock using 
large volumes of crystalloid solution. The 
abdominal cavity will stretch anteriorly and 
superiorly (along the diaphragm) to accom- 
modate visceral edema or accumulating blood 
until it reaches the limits of its compliance. 
At this point, the abdomen becomes a rigid 
compartment and pressure rises sharply, 
impairing organ function. Increased vascular 
resistance and reduced venous return impair 
cardiac output. Reduced renal perfusion 
pressure causes oliguria. Much of the post- 



ch23.qxd 4/16/04 3:27 PM Page 446 



446 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



traumatic renal failure reported in the liter- 
ature during the 1960s and 1970s appears now 
to have been secondary to an abdominal com- 
partment syndrome, which at that time had 
not yet been described. Transference of the 
abdominal pressure to chest and tension on 
the diaphragm increase ventilator and airway 
pressures. Loss of functional residual capac- 
ity and ventilation-perfusion mismatch causes 
hypoxia (Ivatory, Sugerman, and Peiztman, 
2001). 

Several systemic diseases are associated 
with compartment syndromes. Rutgers, van 
derHarst, andKoumans (1991) reported four 
cases on nontraumatic rhabdomyolysis and 
compartment syndrome in young male alco- 
holics receiving treatment with benzodi- 
azepines. Ergotamine use and cocaine 
intoxication have also been implicated in the 
development of compartment syndrome 
(Gilman, Goodman, and Murad, 1989). 
Patients with type I diabetes mellitus can suf- 
fer spontaneous compartment syndrome 
(Lafforgue and colleagues, 1999; Smith and 
Laing, 1999; Silberstein and colleagues, 2001) . 
Systemic diseases or drugs that cause vaso- 
constriction can induce muscle ischemia and 
subsequent compartment syndrome. Local 
factors including hematoma, fluid injection, 
infection, and metastatic melanoma that 
increase mass within the inelastic fascial com- 
partments can also raise intracompartment 
pressure sufficiently to cause the feared 
syndrome (Simmons, 2000). 



ANATOMY 
Calf 

The four muscle compartments of the calf are 
the anterior, lateral, superficial posterior, and 
deep posterior. The anterior compartment is 
bounded by the tibia medially, the interosseous 
membrane posteriorly anterior crural inter- 
muscular septum laterally, and the crural fascia 
anteriorly. It contains the tibialis anterior, the 
extensor digitorum longus, and the extensor 
hallucis longus muscles. It also contains the 



anterior tibial artery and vein, as well as the 
deep peroneal nerve. The lateral compart- 
ment contains the peroneus longus and brevis 
muscles and the superficial peroneal nerve. 
The superficial posterior compartment con- 
tains the bulky soleus muscle. The deep pos- 
terior compartment encloses the tibialis 
posterior, flexor digitorum longus, and flexor 
hallucis longus muscles. The posterior tibial 
vessels and the tibial nerve run within this com- 
partment. Note that the saphenous vein 
courses in the subcutaneous tissue along the 
medial border of the superficial compartment. 
It can be damaged during fasciotomy if 
care is not taken to protect it. Also, the sural 
nerve runs along the posterior lateral border 
of the superficial posterior compartment 
(Clemente, 1981). Lateral and medial inci- 
sions are made throughout the extent of the 
calf, with retraction of the more superficial 
muscles to expose the deep compartment so 
that all compartments are decompressed. His- 
torically, small skin incisions with incisions in 
the superficial fascia only and use of a lateral 
fibulectomy fasciotomy did not adequately 
decompress all calf fascial compartments. 
These procedures have little utility in today's 
trauma armamentarium. 



Thigh 

The thigh has three muscle compartments: 
anterior, medial, and posterior. The anterior 
compartment contains the quadriceps, sar- 
torius, iliacus, and psoas muscles, as well as 
the femoral vessels and nerve and the lateral 
cutaneous nerve. The medial compartment 
encloses the adductor muscles, and the pos- 
terior compartment encircles the biceps 
femoris muscle and the sciatic nerve. Com- 
partment syndrome of the thigh usually 
involves the anterior and lateral compart- 
ments (McGee and Dalsey, 1992) . The gluteal 
muscle group also constitutes a compartment 
that is enclosed by the fascia lata. Gluteal com- 
partment syndrome occurs but is often diag- 
nosed late (Hill and Bianchi, 1997) . A liberal 
lateral fasciotomy will usually decompress a 
thigh compartment syndrome. 



ch23.qxd 4/16/04 3:27 PM Page 447 



23 • COMPARTMENT SYNDROMES 



447 



Arm/Hand 

The muscles of the arm, forearm, and hand 
are also grouped into compartments but are 
no t as defined by tight investing fascia as those 
of the calf (Doyle, 1998). The arm has ante- 
rior and posterior compartments. The ante- 
rior compartment contains the biceps muscle, 
the brachial vessels, and the median, ulnar, 
and musculocutaneous nerves. The posterior 
compartment contains the triceps muscle and 
the radial nerve. The forearm has three com- 
partments the volar, dorsal, and the "mobile 
wad." The volar compartment contains the 
flexor and pronator muscles, the radial and 



ulnar arteries, and the median and ulnar 
nerves. The dorsal compartment contains the 
extensor muscles. The "mobile wad" is closely 
associated with the dorsal compartment and 
contains the radial nerve. The hand has 
four compartments: the central, thenar, 
hypothenar, and interossei. The thick reti- 
naculum cutis and the carpal tunnel serve as 
a venous obstruction at the wrist for com- 
partment syndromes of the forearm and 
hand. Fasciotomies of the forearm, usually 
carried out with "zig-zag" and "straight" inci- 
sions, are carried across the carpal tunnel onto 
the hand to achieve a complete decompres- 
sion (Fig. 23-1). 



Ulnar a. 




Ulnar n. 



Flexor carpi ulnaris m. 



■ FIGURE 23-1 

Elective incisions that can be used for approach to the radial and ulnar arteries. A, An S-type 
incision starting along the course of the distal brachial artery, carried throughout the antecubital 
fossa and continued down on the forearm will give excellent exposure of the proximal ulnar and 
radial arteries as well as the origin of the common interosseous artery (A). An extension of this 
incision (B) along the course of the radial artery can be used for exposure to the wrist level. A 
separate incision can be used over the course of the ulnar artery (C). B, This drawing demonstrates 
exposure of the ulnar neurovascular bundle within the deep muscle layers, which have been split 
proximally. ■ 



ch23.qxd 4/16/04 3:27 PM Page 448 



448 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



Abdomen 

The abdominal viscera are encircled with peri- 
toneum and the intra-abdominal contents are 
contained within the endoabdominal fascia, 
named in various locations as the transversalis 
fascia or Gerota's fascia. This investing fascia 
is contiguous with the esophageal hiatus and 
abdominal outlets at the groins. Numerous 
layers of muscles exist outside the endoab- 
dominal fascia. Both the retroperitoneal and 
intraperitoneal organs are within this fascia. 
Swelling, gaseous distention, tissue edema, 
and hemorrhage are contained within this 
fascia. Although having great capacity to 
contain large quantities of fluid, tissue edema 
combined with large volumes of fluid increases 
abdominal pressure. 



Spinal Cord 

The spinal cord is surrounded by thick dura 
and is contained within a bony encasement. 
It is supplied by radicular arteries from the 
thoracic and abdominal aorta. A single radic- 
ular artery from the segmental arteries divides 
into anterior and posterior radicular arteries. 
The anterior radicular artery feeds a single 
anterior spinal artery, and the posterior radic- 
ular artery feeds paired posterior spinal 
arteries. The anterior spinal artery is more 
rudimentary and may even be interrupted, 
thus explaining the more common anterior 
spinal artery syndrome. Nine paired segment 
arteries arise in the chest, although the 
number may ranges from three to twelve. On 
occasion, one of the segmental arteries off of 
the aorta is much larger than the others and 
has been called the artery of Adamkiewicz. 
This variation is not consistent. Any condition 
from swelling of the spinal cord to pericord 
hematomas can contribute to a spinal column 
compartment syndrome. 



Pericardium 

Contained within the pericardial sac are the 
heart, ascending aorta, intrathoracic inferior 
vena cava, superior vena cava, pulmonary 
artery, right and left main pulmonary arter- 



ies, azygous vein, lymphatic channels, and peri- 
cardial vessels. Any condition that results in 
increasing fluid within the pericardial sac may 
contribute to hemopericardium and the devel- 
opment of pericardial compartment syn- 
drome. Concomitant injury to the anterior 
pericardium and internal mammary arteries 
can also produce hemopericardium. Iatro- 
genic causes of hemopericardium, such as 
puncture of the heart or vessels during peri- 
cardiocentesis or trocar chest tube insertion, 
have been described. 



DIAGNOSIS 

The symptoms of deep muscle pain, pain on 
passive motion, muscle weakness or paralysis, 
hyperesthesia, and tense muscle compart- 
ments have been well described and repeated 
to generations of surgery residents (Matsen, 
Windquist, and Krugmire, 1980; Perry, 1988; 
Velmahos and Toutouzas, 2002) . Recognition 
of the symptom constellation should prompt 
immediate measurement of compartment 
pressure, using any of the several accurate 
devices available. If accurate measurements 
cannot be performed, or if the results are con- 
flicting, a clinical diagnosis of compartment 
syndrome should lead to strong consideration 
for compartment decompression. Once diag- 
nosis is made, immediate release of pressure 
is indicated. 

Abdominal compartment syndrome should 
be suspected in the patient with a tense, 
distended abdomen within a few hours of 
laparotomy for trauma or massive bleeding. 
Visceral swelling or continued bleeding 
push abdominal compliance beyond its 
limits. Oliguria that does not respond to fluid 
boluses or frequent ventilator alarms should 
prompt immediate measurement of the 
intra-abdominal pressure. This can be accom- 
plished easily at the bedside by measuring the 
bladder pressure through a Foley catheter 
(Burch and colleagues, 1996; Ivatury, 
Sugerman, and Peiztman, 2001). 



ch23.qxd 4/16/04 3:27 PM Page 449 



23 • COMPARTMENT SYNDROMES 



449 



Extremity Compartment 
Measurements 

There are several techniques for measuring 
extremity compartment pressures (Matsen 
and colleagues, 1976; Perron, Brady, and 
Keats, 2001; Hargens and colleagues, 1977). 
There are two variations of the catheter tech- 
nique, the wick and the slit catheters. The 
catheters are inserted into the muscle through 
large-bore needles and then connected to a 
pressure transducer or manometer via saline- 
filled tubing. Because insertion and connec- 
tion of the catheters are cumbersome, 
measuring several compartment pressures is 
difficult. The new electronic transducer- 
tipped catheter is promising but shares many 
of the shortcomings with the other catheter 
techniques, such as need for tubes, catheter 
kinking, and poor placement beneath the 
fascia (Willy, Gerngross, and Sterk, 1999) . 
Commercial devices for measuring compart- 
ment pressures are readily available at the 
bedside and are easier to use. 

Manufactured pressure monitors such as 
the Stryker (Stryker Instruments, Kalamazoo, 
Michigan) and Ace (Ace Medical Company, 
Los Angeles, California) instruments employ 
modifications of the needle technique and 
measure pressure directly through a needle 
inserted into the muscle compartment. These 
self-contained units require no assembly, 
making multiple measurements at different 
sites and times easier. 

Regardless of the device used, multiple mea- 
surements should be taken at various sites in 
the muscle and in different compartments. 
Pressure is not uniformly distributed through- 
out each compartment, and measurements 
can be highly variable. In the calf, the ante- 
rior and deep posterior compartments, at 
least, should be measured. The highest mea- 
surement in each compartment should be 
used for clinical decisions. 



Noninvasive Assessment of 
Compartment Compromise 

The persistent trend in medicine toward non- 
invasive diagnosis and treatment extends to 



compartment syndrome. Several techniques 
that have clinical utility in other settings have 
been tried here, including near-infrared spec- 
troscopy (NIRS) , have been studied (Garr and 
colleagues, 1999; Giannotti and colleagues, 
2000; Gentilello and colleagues, 2001). NIRS 
measures muscle perfusion, not pressure, 
and can reliably diagnose ischemic tissue. 
Oxyhemoglobin saturation of less than 60% 
correlates with muscle compromise of 
compartment syndrome. Champions for its 
use argue that it directly identifies ischemic 
tissue rather than compartment pressure, 
which is a proxy for tissue compromise. If clin- 
icians monitor for tissue ischemia rather than 
a rise in pressure, unnecessary fasciotomies 
might be prevented. Conversely, skeptics 
argue that waiting until ischemia is manifest 
may delay surgery. Also, the probe's range is 
limited to 2 cm or less below the skin surface. 
Therefore, it may miss deep muscle ischemia. 
Of the noninvasive tests discussed, NIRS 
holds the most promise. It reliably identifies 
ischemic tissue and can provide continuous 
measurements. The latter makes it particularly 
attractive for use in the operating room and 
intensive care unit, where serial physical 
examinations are difficult on unconscious 
and or multi-injured patients. Continuous 
monitoring may identify development of 
compartment syndrome while surgeons are 
occupied with other injuries. Reliable 
measurements and safe thresholds for oper- 
ation may reduce the unnecessary prophy- 
lactic fasciotomies. Studies of these questions 
continue. 

Continuous compartment pressure or NIRS 
monitoring may influence the decision to 
refrain from fasciotomy. If the surgeon has 
continuous reliable monitoring, an operation 
should not be performed unless pressure 
or tissue perfusion reaches the threshold. 
However, the risk of compartment syndrome 
must be recognized and frequent or contin- 
uous measurements must be undertaken, 
always keeping in focus the clear indications 
for fasciotomy and the consequences of failing 
to act expeditiously. 

Although it has been suggested, digital pulse 
oximetry is not sensitive in diagnosing com- 
partment syndrome and muscle ischemia. It 
relies on pulsatile arterial flow to the distal 



ch23.qxd 4/16/04 3:27 PM Page 450 



450 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



digit to accurately measure the hemoglobin 
oxygen saturation. Because the arterial blood 
measured in the toe or finger bypasses the 
muscle compartments, measuring the former 
gives little useful information of the latter 
(Mars and Hadley, 1994). 

Scintigraphy using technetium-99 methoxy- 
isobutyl isonitrile ( 99m Tc-MIBI) has been used 
to diagnose chronic exertional compartment 
syndrome (Edwards and colleagues, 1999; 
Owens and colleagues, 1999). The study 
requires a stable, ambulating patient, a trip 
to the nuclear medicine department, and a 
subsequent study the next day with the patient 
at rest. With these limitations, this study 
cannot diagnose acute compartment syn- 
drome in time to save the limb. We found no 
reports that it has been studied in acute com- 
partment syndromes. 



Laboratory Evaluation 

There are no laboratory tests that will predict 
or diagnose early compartment syndrome. 
Serum creatinine phosphokinase, a marker 
for muscle cell injury, is a finding in late or 
missed compartment syndrome (Robbs and 
Baker, 1979; Moore and Friedman, 1989). 
Postoperative levels may be useful in moni- 
toring response to treatment. 

Similarly, myoglobinuria is a marker for 
muscle injury. It often occurs with crush or 
electrical injuries, which often lead to com- 
partment syndrome. The presence of myo- 
globinuria in such patients does not per se 
diagnose compartment syndrome. The muscle 
injury may follow from direct trauma rather 
than ischemia secondary to elevated com- 
partment pressures. Therefore, myoglobin- 
uria has little value in diagnosing acute early 
compartment syndrome. 



Pathophysiology 

Restoration of oxygenated blood flow to an 
ischemic limb often worsens the initial cellu- 
lar damage. The reperfused tissue suffers from 
the initial ischemia and from free radical tox- 
icity. If microvascular flow slows or stops, then 
ischemia recurs and free radicals accumulate, 



compounding the injury. This sequence of 
events produces reperfusion injury, which 
is a common etiology of compartment 
syndrome. 

The cellular damage and capillary leak 
result from oxygen free radical and neutrophil 
activity. Hypoxanthine accumulates as a 
product of dephosphorylated adenosine and 
is converted to urate in the presence of xan- 
thine oxidase and oxygen. The enzyme xan- 
thine oxidase also catalyzes the reduction of 
molecular oxygen to superoxide and hydro- 
gen peroxide. These radicals contribute to 
increased microvascular permeability. Super- 
oxide can also generate the hydroxyl radical 
in the presence of Fe 3+ , which is reduced to 
Fe 2+ . The hydroxyl radical is highly cytotoxic 
through lipid peroxidation of the cell 
membrane. Neutrophils adhere to damaged 
microvascular endothelium and release 
superoxide radicals and proteases, con- 
tributing further to reperfusion injury 
(Granger, 1988) . 



TREATMENT OF EXTREMITY 
COMPARTMENT SYNDROME 



Surgery has and continues to be the mainstay 
of treatment of compartment syndromes. 
Releasing the pressure through generous 
fascial incisions restores microvascular flow 
and rescues the threatened tissue. Nonoper- 
ative therapies received minimal theoretical 
initial enthusiasm and support; however, to 
date, none have demonstrated adequate 
efficacy. Choices in operative treatment are 
choices of incision and wound closure. The 
necessity of fasciotomy for diagnosed 
compartment syndrome remains unassail- 
able. Indications for prophylactic fasciotomies, 
however, have been questioned (Field and 
colleagues, 1994; Velmahos and colleagues, 
1997; Velmahos and Toutouzas, 2002). 

Prophylactic fasciotomies in the calves have 
been advocated for combined popliteal artery 
and vein injuries and for ischemic times of 
more than 6 hours. Advocates argue that while 
waiting for compartment pressures to reach 
threshold for precise diagnosis may lead to 



ch23.qxd 4/16/04 3:27 PM Page 451 



23 • COMPARTMENT SYNDROMES 



451 



severe dysfunction or need for an amputation; 
therefore, Hofmeister and Shin (1998) rec- 
ommend liberal fasciotomies, especially in the 
anesthetized or comatose patient. 

Hofmeister and Shin (1998) recommend 
prophylactic fasciotomy of all muscle com- 
partments of the arm after replantation, 
because replantation requires 5 to 10 hours 
to accomplish, and the already compromised 
muscle relies on tenuous arterial and 
venous anastomosis. Fasciotomy, therefore, 
should be performed before compartment 
syndrome develops (Hofmeister and Shin, 
1998) . Under these circumstances, fasciotomy 
is prudent. 



Technique of Decompression 

The four compartments of the lower leg can 
be decompressed through a single lateral 



incision (Fig. 23-2) or through lateral and 
medial incisions (Fig. 23-3) . The two-incision 
technique is more common because it is 
technically easier to reach the posterior com- 
partments through the medial incision. 
Fibulectomy has been described but aban- 
doned because easier and less morbid oper- 
ations accomplish adequate decompression 
(Mubarak and Owen, 1977; Gulli and Tem- 
pleton, 1994) . Care must be taken at the upper 
end of a lateral calf fasciotomy incision to avoid 
injury to the peroneal nerve. Likewise, care 
must be taken to not incise or damage the 
long saphenous vein while making a medial 
calf fasciotomy incision. 

Less invasive methods have been attempted. 
Surgical textbooks of the 1960s showed draw- 
ings of small skin incisions, and using long 
scissors, a continuous medial and lateral 
fascial incisions would be made. As the fascial 
compartments then had increased swelling, 



l-unsecltd p'C? : '■■• . : rtt ■■■■ I 




Tibia 




poster to* 
■ : .-" .• •■ partn enl '■ ■ ' " " d 1 1 i I 



■ i • 



m FIGURE 23-2 

In selected patients, a fasciotomy by means of the subperiosteal fibulectomy technique may have 
merit in obtaining adequate decompression of all four major compartments of the leg. The 
completed fibulectomy/fasciotomy is shown. A, A cross section at the midcalf level, showing 
(arrows)tbe direction to be followed for four-compartment decompression. 6, The area 
decompressed. (From Ernst CB, Kauder HJ: Fibulectomy-fasciotomy. An important adjunct in the 
management of lower extremity arterial trauma. J Trauma 1971 ;1 1[3]:365-380.) ■ 



ch23.qxd 4/16/04 3:27 PM Page 452 



452 



V • SPECIAL PROBLEMS AND COMPLICATIONS 




Saphenous 
vein 



Anterior 
compartment 



Lateral 
compartment 



Deep posterior 
compartment 




Superficial posterior 
compartment 



Lateral 

■ FIGURE 23-3 

Drawing depicting medial and lateral calf incisions to decompress four of the fascial compartments 
of the lower leg. Specifically, note the proximity of the superficial peroneal nerve and saphenous 
vein, which must be protected. (Redrawn from Baylor College of Medicine, 1987.) ■ 



the skin became an investing constriction. 
Ota and colleagues (1999) described endo- 
scopic release of the anterior leg compartment 
using an arthroscope and a transparent 
outer tube for chronic compartment syn- 
drome in an athlete. The patient enjoyed relief 
of symptoms postoperatively, and the com- 
partment pressures diminished (Ota and col- 
leagues, 1999) . Other authors have been less 
enthusiastic about endoscopic fasciotomies. 
Havig, Leversedge, and Seiler (1999) com- 
pared endoscopic and open forearm 
fasciotomies in cadavers. They found the 
endoscopic procedure reduced compartment 
pressures, but not as dramatically as the open 
procedure, and cautioned against using the 
endoscopic forearm fasciotomy in the clini- 
cal setting. 

After diagnosing compartment syndrome 
and performing fasciotomy, the surgeon faces 
a large problematic wound. Primary closure 



is usually impossible because of exuberant 
muscle swelling. Delayed primary closure or 
later skin grafting is the most common method 
of wound closure. 

Advocates of liberal fasciotomies tend to dis- 
count the morbidity of the scars. Conversely, 
other experts hold that complications from 
fasciotomies, including prophylactic ones, can 
be significant (Field and colleagues, 1994; 
Velmahos and colleagues, 1997; Fitzgerald 
and colleagues, 2000) . Wound complications 
include ulcers, skin tethering to the muscle, 
paresthesias, pruritus, muscle herniation, and 
disfigurement. Fitzgerald and colleagues 
(2000) report that unsightly scars resulted in 
life changes for many patients and recom- 
mend primary closure of the wounds when- 
ever possible. 

Delayed primary closure of extremity 
wounds offers the benefit of a smaller scar but 
is usually labor intensive. This method involves 



ch23.qxd 4/16/04 3:27 PM Page 453 



23 • COMPARTMENT SYNDROMES 



453 



some daily manipulation of sutures, wires, or 
elastic bands. Steri-Strips (3M Surgical Prod- 
ucts, St. Paul, Minnesota) have been used for 
gradual approximation of skin edges, closing 
the wound in 5 to 8 days (Harrah and col- 
leagues, 2000) . Chiverton and Redden (2000) 
used subcuticular polypropylene sutures to 
achieve skin closure. Harris (1993) described 
using rubber vessel loops stretched between 
skin staples in shoelace fashion. One historic 
technique involved using interrupted wires 
stretched between skin staples, but this tech- 
nique has been abandoned because of the 
difficulty endured by physician, patient, and 
nurses. The technique required adding 
tension daily by twisting of 20 to 30 interrupted 
wires spanning the incisions. The theory was 
attractive, but the practice was arduous. 

Wound closure with split-thickness skin 
grafts is accomplished in 5 to 7 days after the 
fasciotomy. This method requires little bedside 
wound manipulation and achieves closure of 
large wound. It requires an additional general 
anesthetic for the patient and produces a sig- 
nificant scar. Skin grafting, however, is a main- 
stay in this setting because of its simplicity and 
coverage of large wound areas. 

Because of the morbidity of fasciotomy, 
medical treatments have been researched in 
animals. The results are equivocal. Most are 
used to ameliorate the damage from oxygen 
free radicals (Hofmeister and Shin, 1998) . 
They include deferoxamine to chelate iron, 
xanthine oxidase inhibitors, such as allop- 
urinol, to block production of hypoxan thine, 
and superoxide dismutase, an enzyme to cat- 
alyze the superoxide radical to hydrogen per- 
oxide. These antioxidants have been studied 
in many animal models, but not in humans. 
Currently, such nonprocedural therapies are 
not recommended. 

In the abdomen, primary closure of the 
fascia is usually impossible, and sometimes, a 
skin-only closure can be accomplished. The 
most common forms of closure after laparo- 
tomy for abdominal compartment syndrome 
involve some form of temporary prosthesis 
such as a "Bogota bag" or vacuum pack 
(Burch and colleagues, 1996; Ivatury, Suger- 
man, and Peiztman, 2001). These prostheses 
maintain protection of the visceral while 



allowing loss of domain and effectively increas- 
ing the volume of the abdominal cavity. 
Removal of the prosthesis may be accom- 
plished when swelling recedes. If delayed 
primary closure cannot be performed, skin 
grafting or component separation can cover 
the viscera. For the most severe forms of 
abdominal compartment syndrome in 
patients with multisystem trauma and pro- 
longed intensive care unit stays, secondary 
reconstruction of the abdominal wall, using 
prosthetic material sewn to the fascia, may 
be accomplished several months later, often 
longer than 12 months. 



SUMMARY 



Compartment syndrome, if not detected early, 
can result in loss of limb, organ function, and 
even life. Effective treatment relies on early 
diagnosis through clinical examination and 
bedside measurements of compartment pres- 
sures. Measurements are accomplished using 
one of several commercially available devices. 
NIRS may have benefit as a noninvasive 
harbinger of muscle compromise. Although 
research has mapped the complex reactions 
in reperfusion injury, it has not produced a 
means for prevention or effective medical 
treatment. 

Once diagnosis is made, the surgeon must 
perform expeditious decompression. Avariety 
of incisions have been described. In the lower 
leg, median and lateral longitudinal incisions 
are m ost comm only used . In the forearm , volar 
and radial incisions are preferred. For the 
abdomen, a midline laparotomy accomplishes 
decompression. Pericardiotomy relieves 
compartment syndrome of the pericardium. 

Prophylactic fasciotomy for high-risk 
patients is common. With newer, more reliable 
methods of tissue perfusion and compartment 
pressure measurements, prophylactic fas- 
ciotomy may be performed less commonly. 
Obviously, unnecessary fasciotomy should be 
avoided if possible. However, if muscle, organ, 
or limb loss is the alternative, decompression 
of the compartment is always indicated. 



ch23.qxd 4/16/04 3:27 PM Page 454 



454 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



REFERENCES 

Burch JM, Moore EE, Moore FA, Franciose R: The 
abdominal compartment syndrome in complex 
and challenging problems in trauma surgery. 
Surg Clin North Am 1996;76(4):883-843. 

Chiverton N, Redden JF: A new technique for 
delayed primary closure of fasciotomy wounds. 
Injury 2000;31 (l):21-24. 

Clemente C: Anatomy, a Regional Atlas of the 
Human Body, 2nd ed. Baltimore: Urban & 
Schwarzenberg, 1981. 

Cywes S, LouwJH: Phlegmasia cerulea dolens: suc- 
cessful treatmentby relieving fasciotomy. Surgery 
1962;51:169-172. 

Dennis C: Disaster following femoral vein ligation 
for thrombophlebitis; relief by fasciotomy; clin- 
ical case of renal impairment following crush 
injury. Surgery 1945;17:264-269. 

Doyle J: Anatomy of the upper extremity muscle 
compartments. Hand Clin 1998;14:343-364. 

Edwards PD, Miles KA, Owens SJ, et al: A new non- 
invasive test for detection of compartment syn- 
dromes. Nuclear Med Commun 1999;20(3): 
215-218. 

Field CK, SenkowskyJ, Hollier LH, et al: Fasciotomy 
in vascular trauma: is it too much, too often? Am 
Surg 1994;60(6):409-411. 

Fitzgerald AM, Gaston P, Quaba A, McQueen MM: 
Long term sequelae of fasciotomy wounds. Br J 
Plast Surg 2000;53:690-693. 

GarrJL, Gentilello LM, Cole PA, etal: Monitoring 
for compartmental syndrome using near-infrared 
spectroscopy: a noninvasive, continuous, trans- 
cutaneous monitoring technique. J Trauma 
Injury Infect Crit Care 1999;46(4):613-618. 

Gentilello LM, Sanzone A, Wang L, et al: Near- 
infrared spectroscopy versus compartment pres- 
sure for the diagnosis of lower extremity 
compartmental syndrome using electromyog- 
raphy-determined measurements of neuromus- 
cular function. J Trauma Injury Infect Crit Care 
2001;51(l):l-9. 

Giannotti G, Cohn SM, Brown M, et al: Utility of 
near-infrared spectroscopy in the diagnosis 
of lower extremity compartment syndrome. J 
Trauma Injury Infect Crit Care 2000;48(3) :396- 
401. 

Gilman AG, Goodman LS , Murad F: Goodman and 
Gilman's The Pharmacological Basis of Thera- 
peutics, 8th ed. New York: Macmillan, 1989. 

Granger DN: Role of xanthine oxidase and gran- 
ulocytes in ischemia-reperfusion injury. Am J 
Physiol 1988;246:H1269-H1275. 



Gulli B, Templeton D: Compartment syndrome of 
the lower extremity. Orthoped Clin North Am 
1994;25(4):677-684. 

Hargens AR, Mubarak SJ, Owen CA, et al: Inter- 
stitial fluid pressure in muscle and compartment 
syndromes in man. Microvasc Res 1977;14:1-10. 

Harrah J, Gates R, Carl J, Harrah JD: A simpler, 
less expensive technique for delayed primary 
closure of fasciotomies. AmJ Surg 2000;180(1) : 
55-57. 

Harris I: Gradual closure of fasciotomy wounds 
vising a vessel loop shoelace. Injury 1993;24(8) : 
565-567. 

Havig MT, Leversedge FJ, Seller JG 3rd: Forearm 
compartment pressure: an in vitro analysis of 
open and endoscopic assisted fasciotomy. J Hand 
Surg Am 1999;24(6):1289-1297. 

Hill SL, BianchiJ: The gluteal compartment syn- 
drome. Am Surg 1997;9:823-826. 

Hofmeister EP, Shin AY: The role of prophylactic 
fasciotomy and medical treatment in limb 
ischemia and revascularization, in compartment 
syndrome and Volkmann's ischemic contracture. 
Hand Clin 1998;14(3):457-465. 

Ivatury RR, Sugerman HJ, Peiztman AB: Abdomi- 
nal compartment syndrome: recognition and 
management. In: Advances in surgery, vol. 35. 
St. Louis, MO: Mosby, 2001:251-269. 

Lafforgue P, Janand-Delenne B, Lassman-Vague V, 
et al: Painful swelling of the thigh in a diabetic 
patient: Diabetic muscle infarction. Diabetes 
Metab 1999;25(3):255-260. 

Mars M, Hadley GP: Failure of pulse oximetry in 
the assessment of raised limb intracompart- 
mental pressure. Injury 1994;25(6):379-381. 

Matsen FA, Mayo KA, Sheridan GW, Krugmire RB: 
Monitoring of intramuscular pressure. Surgery 
1976;79(6):702-709. 

Matsen FA, Windquist RA, Krugmire RB: Diagno- 
sis and management of compartmental syn- 
dromes. J Bone Joint Surg Am 1980;62:286. 

McGee DL, Dalsey WC: The mangled extremity; 
compartment syndrome and amputations, in soft 
tissue emergencies. Emerg Med Clin North Am 
1992;10(4):783-800. 

Moore RE III, Friedman RJ: Current concepts in 
pathophysiology and diagnosis of compartment 
syndromes. J Emerg Med 1989;7:657-662. 

Mubarak SJ, Owen CA: Double-incision fasciotomy 
of the leg for decompression in compartment 
syndromes. J Bone Joint Surg 1977;59A:184- 
187. 



ch23.qxd 4/16/04 3:27 PM Page 455 



23 • COMPARTMENT SYNDROMES 



455 



Ota Y, Senda M, Hashizume H, Inoue H: Chronic 
compartment syndrome of the lower leg: a new 
diagnostic method using near-infrared spec- 
troscopy and a new endoscopic fasciotomy. 
Arthroscopy 1999;15(4) :439-443. 

Owens S, Edwards P, Miles K, et al: Chronic com- 
partment syndrome affecting the lower limb: 
MIBI perfusion imaging as an alternative to pres- 
sure monitoring: two case reports. Br J Sport 
Medl999;33(l):49-51. 

Perron AD, Brady WJ, Keats TE: Orthopedic pit- 
falls in the ED: acute compartment syndrome. 
Am J Emerg Med 2001;19(5):413-416. 

Perry MO: Compartment syndromes and re per- 
fusion injury. Vase Trauma Surg Clin North Am 
1988;68(4):853-864. 

Robbs JV, Baker LW: Late revascularization of the 
lower limb following acute arterial occlusion. Br 
J Surg 1979;78:490-493. 

Russel GV, Kregor PJ, Jarre t CA, Zlowodowski M: 
Complicated femoral shaft fractures, in treat- 
ment of complex fractures. Orthop Clin North 
Am2002;33(l):l-17. 

Rutgers PH, van der Harst E, Koumans RKJ: Sur- 
gical implications of drug induced rhabdomy- 
olysis. Br J Surg 1991;78:490-492. 

Schwartz JTJr, Brumback RJ, Lakatos R, et al: Acute 
compartment syndrome of the thigh, a spectrum 
of injury.JBone Joint Surg Am 1989;71:392-400. 



Silberstein L, Britton KE, Marsh FP, et al: An unex- 
pected cause of muscle pain in diabetes. Ann 
Rheum Dis 2001;60(4):310-312. 

Simmons DJ: Compartment syndrome complicat- 
ing metastatic malignant melanoma. Br J Plast 
Surg 2000;53(3):255-257. 

Singhal P, Horowitz B, Quinones QC, et al: Acute 
renal failure following cocaine abuse. Nephron 
1989;52:76-78. 

Smith AL, Laing PW: Spontaneous tibial com- 
partment syndrome in type I diabetes mellitus. 
Diabetes Med 1999;16(2):168-169. 

Velmahos GC, Theodorou D, Demetriades D, et 
al: Complications and nonclosure rates of fas- 
ciotomy for trauma and related risk factors. World 
J Surg 1997;21:247-253. 

Velmahos GC, Toutouzas KG: Vascular trauma and 
compartment syndromes, in vascular trauma: 
complex and challenging injuries, part II. Surg 
Clin North Am 2002;82(1):1 25-141. 

Willy C, Gerngross H, SterkJ: Measurement of in tra- 
compartmental pressure with use of a new elec- 
tronic transducer-tipped catheter system.J Bone 
Joint Surg Am 1999;81 (2) : 158-1 68. 

Wood KE, Reedy JS, Pozniak MA, Coursin DB: 
Phlegmasia cerulean dolens with compartment 
syndrome: a complication of femoral vein cau- 
terization. Crit Care Med 2000;28 (5) :1 626-1 630. 



ch24.qxd 4/16/04 3:25 PM Page 457 




Historic Review of 
Arteriovenous Fistulas and 
Traumatic False Aneurysms 

NORMAN M. RICH 



ARTERIOVENOUS FISTULA 

History 

Incidence 

Etiology 

Pathophysiology 

Clinical Pathology 

Clinical Features 

Diagnostic Considerations 

Surgical Treatment 

Spontaneous Cure 

Results 

Follow-up 

TRAUMATIC FALSE ANEURYSMS 

History 

Incidence 

Etiology 

Clinical Pathology 

Clinical Features 

Diagnostic Considerations 

Surgical Treatment 

Spontaneous Cure 

Follow-up 



457 



ch24.qxd 4/16/04 3:25PM Page 458 



458 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



ARTERIOVENOUS FISTULA 



If it should be found by experience, that a 
large artery, when wounded, may be healed 
up by this kind of suture, without becoming 
impervious, it would be an important 
discovery in surgery. It would make the 
operation for the Aneurysm still more 
successful in the arm, when the main trunk 
is wounded; and by this method, perhaps, 
we might be able to cure the wounds of 
some arteries that would otherwise require 
amputation, or be altogether incurable. 
Lambert, 1762; quoted in Hallowell, 1762 

It is generally accepted that the first successful 
arterial repair was performed by Hallowell in 
1759. His comments emphasize his realization 
that repair of false aneurysms and arteriove- 
nous fistulas (AVFs) could be valuable. The 
diagnosis, pathophysiology, and surgical man- 
agement of AVFs and false aneurysms have 
stimulated the intellectual curiosity and chal- 
lenged the technical abilities of surgeons for 
more than 200 years. These lesions are often 
found in association, and they are often dis- 
cussed together, despite the variable aspects 
that exist. Appropriate emphasis is given 
where indicated. 

Because of the outstanding contributions 
of Matas, Halsted, Reid, Holman, Elkin, 
Shumacker, Hughes, and others and a 
plethora of reports from three major armed 
conflicts in this century, considerable docu- 
mentation exists regarding principles of diag- 
nosis and management of AVFs and false 
aneurysms. During the Korean Conflict, 
Hughes, Jahnke, and Spencer documented 
that arterial repair could be successful, even 
in a combat zone. Consequently, more vascular 
repairs were done at the time of initial wound- 
ing, with a resultant decrease in the number 
of AVFs and false aneurysms that required later 
repair. With the rapid progress that was made 
in vascular surgery in the 10 years preceding 
the increased American military involvement 
in Southeast Asia in 1965, hundreds of well- 
trained young surgeons from both military 
and civilian training programs were available 



and eager to perform vascular repairs 
during the fighting in the Republic of South 
Vietnam. 

With the establishment of the Vietnam Vas- 
cular Registry at Walter Reed Army Medical 
Center in 1966, an effort was made to docu- 
ment as accurately as possible all vascular 
injuries that occurred among American casu- 
alties in Southeast Asia and to provide long- 
term follow-up of these casualties. The initial 
analysis was important in providing guide- 
lines for determining the ultimate success 
or failure following various types of repairs. 
It was believed that there would be relatively 
few AVFs and false aneurysms, compared with 
other recent wars. Nevertheless, it was re- 
cognized that a number of factors, such as 
multiple wounds and other more serious 
problems, might lead to delayed recognition 
of both AVFs and false aneurysms. In later 
follow-up from the registry, it was shown that 
there were more AVFs and false aneurysms 
than initially anticipated. The registry report 
provided an analysis of information gathered 
over 9 years for nearly 7500 records of 
American casualties, showing that there were 
558 AVFs and false aneurysms among 509 
combat casualties (Rich, Hobson, and Collins, 
1975) (Fig. 24-1). 

History 

Hunter (1757, 1762) provided documentation 
more than 200 years ago that the heart 
enlarged in a patient with an AVF and that 
the arterial dilation occurred proximal to an 
arteriovenous communication (Table 24-1). 
Norris (1843) noted the recurrence of phys- 
ical findings associated with an arteriovenous 
aneurysm 10 days after ligation of the artery 
above and below the fistula. Nicoladoni (1875) 
andBranham (1890) described slowing of the 
heart with pressure occlusion of an arteri- 
ovenous communication, and their names are 
often associated with this physical finding (the 
Nicoladoni-Branham sign) . Annandale (1875) 
described the successful management of a 
popliteal AVF by the ligature of the popliteal 
artery and vein. Eisenbrey (1913) described 
pathologic changes associated with arteri- 
ovenous aneurysms of the superficial femoral 



ch24.qxd 4/16/04 3:25 PM Page 459 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



459 




■ FIGURE 24-1 

Multiple fragments from various exploding 
devices were responsible for the majority 
(87.3%) of arteriovenous fistulas and false 
aneurysms in this study. The subtraction study of 
an arch angiogram helped confirm the clinical 
impression of an arteriovenous communication in 
the patient's right neck at the level of the 
common carotid bifurcation. Excision of the 
fistula with ligation of the external jugular vein 
was performed at Walter Reed Army Medical 
Center in 1971 . (From Rich NM, Hobson RW II, 
Collins GJ Jr: Traumatic arteriovenous fistulas 
and false aneurysms: A review of 558 lesions. 
Surgery 1975;78:817-828.) ■ 



TABLE 24-1 

ACHIEVEMENTS AND UNDERSTANDING IN TREATING ARTERIOVENOUS FISTULAS: 
REPRESENTATIVE HISTORICAL NOTES 



Author (Yr) 

Hunter (1757) 



Norris(1843) 
Breshet(1833) 

Nicoladoni (1875) 

Branham (1890) 

Stewart (1913) 

Gunderman (1915) 

Reid (1920) 

Nanu and colleagues (1922) 

Franz 

Holman (1937) 



Contribution 

Recognized an abnormal communication between an artery and vein. 

Described the associated thrill and bruit. 

Eliminated the thrill and bruit by pressure over the proximal artery or site 

of communication. 
Noted tortuosity and dilation of the artery proximal to the fistula. 
Cured an arteriovenous fistula by double arterial ligation. 
Described two patients in whom ligation of the artery proximal to the 

arteriovenous communication was followed by gangrene. 
The first to demonstrate the remarkable slowing of the pulse rate by 

compression of the artery proximal to the arteriovenous fistula. 
Emphasized the slowing of the pulse rate by obliterating a large 

acquired arteriovenous fistula (Branham-Nicoladoni sign). 
Noted that the heart diminished in size within 10 days after elimination 

of the arteriovenous fistula. 
The first to mention an increase in blood pressure on obliteration of an 

acquired arteriovenous fistula. 
Presented experimental evidence of cardiac enlargement in the 

presence of an arteriovenous fistula. 
Accurately described the effect on the blood pressure of closure of 

the arteriovenous fistula. 
Observed an increase in skin temperature and an increase in extremity 

growth in the presence of a femoral fistula of 18 months' duration in a 

12-year-old boy. 
Clarified many of the anatomic and hemodynamic variations seen with 

arteriovenous fistulas. 



ch24.qxd 4/16/04 3:25 PM Page 460 



460 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



vessels (Fig. 24-2). Holman (1937), in his 
classic monograph, described the patho- 
physiology associated with abnormal com- 
munications between arterial and venous 
circulations. Holman (1940, 1962) has also 
provided reviews of the pathophysiology of 
AVFs. Osier (1893, 1905) made a number of 
early observations on AVFs. His respect for 
these lesions is exemplified by a quotation 
from an article written in 1905, "The great 
danger of operating is in the gangrene which 
is apt to follow." 

Halsted made numerous contributions in 
the field of vascular surgery, including the 



management of AVFs. He referred to case pre- 
sentation by Bernheim in 1916, when the latter 
used an interposition autogenous saphenous 
vein graft as a replacement for a popliteal 
repair (Halsted, 1916). He noted the impor- 
tant contributions of Carrel and specifically 
stated that the operation of Lexer, which Bern- 
heim also was advocating, was the "the ideal 
operation." Reid, in two important contribu- 
tions (1920, 1925), described abnormal arte- 
riovenous communications. Using the vast 
World War II experience, Elkin ( 1945) , Elkin 
and DeBakey (1955), and Shumacker (1946, 
1950) documented a number of important 



■ FIGURE 24-2 

An arteriovenous communication with 
extensive vascular alterations. (From 
EisenbreyAB. JAMA 1913;61:2155- 
2157.) ■ 




Sat ov\U* 



ch24.qxd 4/16/04 3:25 PM Page 461 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



461 



findings. Shumacker (1946) outlined the sur- 
gical approach to various AVFs and false 
aneurysms. He recognized the important 
work by Matas (1901, 1908), who made sig- 
nificant contributions to the present man- 
agement of both AVFs and false aneurysms. 
The management of 215 AVFs and false 
aneurysms during the Korean Conflict was 
reported by Hughes andjahnke (1958). 



Incidence 



The lessons learned in Korea, the advances 
made in the techniques of vascular surgery, 
the increased numbers of surgeons trained 
in vascular techniques, plus rapid 
evacuation, new instruments and antibiotics 
have resulted in practically all arterial 
injuries occurring in Vietnam being repaired 
primarily with a high degree of success, so 
that only rarely do patients develop an 
arteriovenous fistula or false aneurysm. 



It is difficult to determine the true incidence 
of AVFs. Some series combine congenital with 
traumatic lesions. False aneurysms may or may 
not be included. Some reports of arterial 
lesions include AVFs and others do not. Often 
the diagnosis is not made until years later. As 
an example, AVFs are still diagnosed at this 
time amongWorld War II veterans, more than 
30 years after their original injury. 

Encouraged by Halsted, Callander (1920) 
made a literature review of 447 AVFs to 1914, 
including some from World War I. In the 
earliest reports of management of combat- 
incurred AVFs, Soubbotitch (1913) reported 
a insignificant percentage of vascular injuries: 
77 injuries to large blood vessels among 
20,000 wounded. The numerous separate 
reports by Elkin, Shumacker, Freeman, and 
others from their vast experience during 
World War II are included in a final bound 
report (Elkin and DeBakey, 1955). A total of 
593AVFswere treated; however, no incidence 
was given for these lesions among World War 
II combat casualties. In the Korean Conflict, 
202 patients were treated for 215 AVFs and 
false aneurysms, with notation made for inci- 
dence among all combat casualties (Hughes 
andjahnke, 1958). 

The only statistic from the Vietnam 
experience of any value was an incidence 
of approximately 7% of AVFs and false 
aneurysms among nearly 7500 American casu- 
al tiesin Southeast Asia who suffered some type 
of vascular trauma. When Heaton and col- 
leagues (1966) evaluated the initial military 
surgical practices of the U.S. Army in Vietnam, 
they recorded the following: 



The factors mentioned certainly played a 
significant role in limiting the number of 
AVFs and false aneurysms. With time, however, 
an increasing number of AVFs and false 
aneurysms were recorded. In many cases, 
these occurred in patients sustaining multi- 
ple small fragment wounds over a large 
portion of the body, which made it impracti- 
cal to explore every artery in which a vascu- 
lar injury might be present. 

Hewitt and Collins (1969) reported a 10% 
incidence of AVFs among 60 patients with arte- 
rial injuries treated between December 1966 
and October 1967, at the Eighteenth Surgi- 
cal Hospital and during November 1967 at 
the Seventy-first Evacuation Hospital in 
Vietnam. Five of the six lesions were acute 
AVFs, which were noted on admission of the 
patients to the hospital within 1 to 6 hours 
after injury. 

Civilian reports of vascular trauma have 
increased in the past 40 years, and some 
include reviews of experience in managing 
AVFs. Patman, Poulos, and Shires (1964) 
included six patients with AVFs among their 
256 patients with civilian arterial injuries, an 
incidence of 2.3%. Drapanas and colleagues 
(1970) stated that because the immediate 
repair of all acute arterial injuries is advocated, 
the development of serious delayed compli- 
cations, including AVFs and false aneurysms, 
should largely be prevented. They found that 
chronic AVFs and false aneurysms declined 
noticeably during the last period of their study, 
between 1958 and 1969 at Charity Hospital in 
New Orleans (Fig. 24-3) . Hewitt, Smith, and 
Drapanas (1973) reported a 6.8% incidence, 
with 14 cases of acute AVFs among 206 
patients with acute arterial injuries treated 



ch24.qxd 4/16/04 3:25 PM Page 462 



462 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



[2 12- 

5 iO- 
1*- 


v . 


u. 61 

2 2- 
0- 


1 1 1 | A i A X X 



58 60 62 64 66 68 70 
YEAR 

■ FIGURE 24-3 

The number of patients with chronic 
arteriovenous fistulas and false aneurysms 
admitted to Charity Hospital in New Orleans on 
the Tulane Service between 1958 and 1969. 
There has been a notable decline in the 
incidence of these vascular injuries with 
delayed recognition. (From Drapanas T, Hewitt 
RL, Weichert RF, Smith AD: Civilian vascular 
injuries: A critical appraisal of three decades of 
management. Ann Surg 1970;172:351-360.) ■ 



on the Tulane University Surgical Service 
(Fig. 24-4). 

The incidence of AVFs compared to that of 
false aneurysms has varied from one series 
to another. Shumacker and Carter (1946) 
studied 364 AVFs and false aneurysms in 351 
individuals. There were 245 AVFs and 119 
aneurysms, with 206 and 82, respectively, 
operated upon at one of the three Vascular 
Centers, Mayo General Hospital, established 
by the Army Surgeon General during World 
War II (Fig. 24-5; Table 24-2). In the 1964 
series of Patman, Poulos, and Shires from 
Dallas, there were 17 patients who developed 
late complications, but only five AVFs were 
reported, compared with 12 false aneurysms. 
Thus, in their series, the false aneurysms out- 
numbered the AVFs by 2 : 1, a ratio opposite 
to that reported by Hughes andjahnke (1958) 
from the Korean experience. 

Seel ey and colleagues (1952) reported that 
AVFs occurred in at least twice as often as false 



CAROTID- JUGULAR/(2 



I 1 SUBCLAVIAN 

\ 

2) AXILLARY 




EXTERNAL ILIAC ( I 



2) COMMON FEMORAL 



POPLITEAL 



■ FIGURE 24-4 

Distribution of acute arteriovenous fistulas in 14 
of 206 patients with acute civilian arterial 
injuries in New Orleans: an incidence of 6.8%. 
(From Hewitt RL, Smith AD, Drapanas T: Acute 
traumatic arteriovenous fistulas. J Trauma 
1973;13:901-906.) ■ 



aneurysms in 106 cases seen at Walter Reed 
General Hospital. Most of the patients sus- 
tained their injury in the earlier part of the 
Korean Conflict. The incidence was nearly 
equal in the Vietnam experience (Rich, 1975) , 
although there were fewer AVFs than false 
aneurysms (Table 24-3). AVFs and false 
aneurysms are often found together in var- 
ious anatomic configurations (Figs. 24-6 and 
24-7). Shumacker and Wayson (1950) out- 
lined the development of AVFs, showing that 
pulsating hematomas may present initially, 
with well-formed saccular aneurysms devel- 
oping subsequently. Notes made at the time 



ch24.qxd 4/16/04 3:25 PM Page 463 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 463 



ARTERIOVENOUS FISTULAS 



INNOMINATE, 
CAROTID, 
VERTEBRAL, 
ft BRANCHES 

SUBCLAVIAN, 

AXILLARY, 

BRACHIAL, 

RADIAL, 

ULNAR, 

* BRANCHES 



EXTERNAL ILIAC, 
HYPOGASTRIC, 
ft BRANCHES 



27 (11%) 



47 (19.255) 



5 (250 



ARTERIAL ANEURYSMS 



9 (7.6J5) 



COMMON FEMORAL, 

FEMORAL, 

PROFUNDA, 78 (51. 8#) 

ft BRANCHES 



POPLITEAL, 
ft BRANCHES 



46 (18.4J?) 



ANTERIOR TIBIAL, 
POSTERIOR TIBIAL, 
PERONEAL, 4J (17.651) 
ft BRANCHES 




64 (46.470 



2 (1.75!) 



21 (17.6J5) 



21 (17.655) 



12 (10. 150 



■ FIGURE 24-5 

General distribution of arteriovenous fistulas and false aneurysms in a study from the Mayo General 
Hospital during World War II. (From Shumacker HB Jr, Carter KL: Arteriovenous fistulas and false 
aneurysms in military personnel. Surgery 1946;20:9-25.) ■ 



TABLE 24-2 

COMPARISON OF INCIDENCE OF ARTERIAL ANEURYSMS AND ARTERIOVENOUS 
FISTULAS IN THE MAIN PERIPHERAL ARTERIES: MAYO GENERAL HOSPITAL, 
WORLD WAR II 



Involved Artery 


Arteriovenous Fistulas 




No. 


% 


Subclavian 


10 


4.1 


Axillary 


12 


4.9 


Brachial 


13 


5.3 


Common femoral and femoral 


66 


26.9 


Popliteal 


42 


17.1 



Arterial Aneurysm 

No. % 



5 
15 
28 
17 
21 



4.2 

12.6 
23.5 
14.3 
17.6 



From Shumacker HB Jr, Carter KL: Arteriovenous fistulas and false aneurysms in military personnel. Surgery 1946;20:9-25. 



ch24.qxd 4/16/04 3:25 PM Page 464 



464 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



TABLE 24-3 

ARTERIOVENOUS FISTULAS AND 
FALSE ANEURYSMS: VIETNAM 
VASCULAR REGISTRY 



Lesions 


No. 


» 


False aneurysms 
Arteriovenous fistulas 
Total 


296 
262 
558 


53.1 

46.9 

100.0 



From Rich NM, Hobson RW II, Collins GJ Jr: Traumatic 
arteriovenous fistulas and false aneurysms: A review of 
558 lesions. Surgery 1975;78:817-828. 







CD A 



(fill 







■ FIGURE 24-6 

This diagrammatic representation of various types of arteriovenous fistulas and associated 
aneurysms evolved from a study of 195 cases of arteriovenous fistulas. There was an associated 
aneurysm in 60% of the arteriovenous fistulas. A, artery; S, sac; V, vein. (From Shumacker HB Jr, 
Wayson EE: Spontaneous cure of aneurysms and arteriovenous fistulas, with some notes on 
intravascular thrombosis. Am J Surg 1950;79:532-544.) ■ 



ch24.qxd 4/16/04 3:25 PM Page 465 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



465 




■ FIGURE 24-7 

Femoral angiogram demonstrating an 
arteriovenous fistula at the level of the right 
common femoral arterial bifurcation. There is an 
associated false aneurysm (arrow). 
Arteriorrhaphy of the origin of the profunda 
femoris artery and venorrhaphy of the common 
femoral vein were successfully accomplished 
at Walter Reed Army Medical Center in 1970. 
(From Rich NM, Hobson RW II, Collins GJ Jr: 
Traumatic arteriovenous fistulas and false 
aneurysms: A review of 558 lesions. Surgery 
1975;78:817-828.) ■ 

of operation and on examination of the 
excised specimen permitted an analysis of the 
presence or absence of an aneurysms in 195 
cases of AVFs. There was no associated 
aneurysms in 78 cases, or 40%. The 60% 
majority had one or more aneurysm. Multi- 
ple lesions may also exist in various anatomic 
sites (Table 24-4). 

A wide variation exists in the regional dis- 
tribution of AVFs. This may include specific 
arteries, as well as regional areas. During a 
15-year period from 1947 through 1962, 50 
patients with AVFs were admitted to the 
Baylor University College of Medicine- 
affiliated hospitals in Houston. The greatest 



TABLE 24-4 

ARTERIOVENOUS FISTULAS AND 
FALSE ANEURYSMS; MULTIPLE 
LESIONS AT VARIOUS ANATOMIC 
SITES: VIETNAM VASCULAR REGISTRY 



Patients 


Lesions 




Total 


468 


1 




468 


35 


2 




70 


4 


3 




12 


2 


4 




8 


509 






558 



From Rich NM, Hobson RW II, Collins GJ Jr: Traumatic 
arteriovenous fistulas and false aneurysms: a review of 
558 lesions. Surgery 1975;78:817-828. 



TABLE 24-5 

LOCATION OF ARTERIOVENOUS 
ANEURYSMS: BAYLOR UNIVERSITY 
COLLEGE OF MEDICINE AFFILIATED 
HOSPITALS 

Location 



Popliteal 

Femoral 

Brachial 

Common carotid 

Radial 

Subclavian 

External carotid 

Internal carotid 

Posterior tibial 

Temporal 

Aortic arch 

Internal iliac 

External iliac 

Occipital 

Internal maxillary 

Thyrocervical 

Uterine 

Peroneal 

Medial circumflex femoral 

Total 



50 



From Beall AC Jr, Harrington BO, Crawford ES, DeBakey ME: 
Surgical management of traumatic arteriovenous aneurysms. 
Am J Surg 1963;106:610-618. 



number of these lesions were found in the 
extremities, with the lower extremities being 
more commonly involved than the upper 
(Table 24-5). Vollmar and Krumhaar (1968) 
found that nearly 50% of the AVFs in their 



ch24.qxd 4/16/04 3:25 PM Page 466 



466 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



series were localized in the lower extremities 
(Fig. 24-8). Next in frequency were fistulas 
of the upper extremities and shoulders (27%) , 
head and neck (22.5%), and trunk (2%). 
Table 24-6 outlines representative World War 
II statistics concerning predominantly lower 
extremity injuries, specifically those involving 
the femoral and popliteal vessels. Involve- 
ment of major (Table 24-7) and minor (Table 
24-8) vessels was outlined from the Korean 
experience by Hughes and Jahnke (1958). The 
Vietnam data centered around lower extrem- 
ity involvement (Table 24-9) , with the super- 
ficial femoral and popliteal arteries being most 
commonly injured (Table 24-10). 



There are hundreds of reports of specific 
or unusual AVFs. Complete analysis is beyond 
the scope of this review. Creech, Gantt, and 
Wren (1965) presented a series of traumatic 
AVFs at unusual sites, including the superior 
gluteal, hepatic-portal, coronary, and vertebral 
vessels. Conn and colleagues (1971) reported 
challenging arterial injuries, including an aor- 
tocaval fistula, an iliac AVF, and a mesenteric 
AVF in eight patients. 

Other representative reports include the fol- 
lowing (additional information can be found 
in specific chapters): Hunt and colleagues 
(1971) reported their experience in manag- 
ing five AVFs of major vessels in the abdomen. 



LOCALIZATION OF 200 TRAUMATIC ARTERIOVENOUS FISTULAE 
( Surg. Clin of the Univ. of Heidelberg, 1939 - 1967 ). 

Carotis interna(8\^ f \ J^f) Carotis- Sin. cavern. 

(9) Carotis externa 



Thyreoideosup 
Carotis commun 

Transversa scapul. 
Anonyma C~2\- 

Axillans 

Cubitahs(2_ 

Ulnans\3) 



Popliteal) 



\T) Vertebra lis 

~\f) Thyreoidea inf. 

J7)Subclavia 
i)Pulmonalis 

2T)Brachialis 



T) Aorta 
f)lliacacomm 




MjFemoralis 



(^Tibialis post. 



m FIGURE 24-8 

In the cases seen at Heidelberg University, nearly 50% of the arteriovenous fistulas were found in 
the lower extremities. (From Vollmar J, Krumhaar D: In: Hiertonn T, Rybeck B, eds. Traumatic arterial 
lesions. Stockholm, Sweden: Research Institute of National Defense, 1968.) ■ 



ch24.qxd 4/16/04 3:25 PM Page 467 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



467 



TABLE 24-6 

DISTRIBUTION OF FALSE ANEURYSMS AND ARTERIOVENOUS FISTULAS: MAYO 
GENERAL HOSPITAL 





Arteriovenous Fistulas: 


No. of Cases 


Arterial 


Aneurysms: 


No. of Cases 




Operation 






Operation at 








at Mayo 






Mayo 






Involved 


General 


Operation 


"Spontaneous 


General 


Operation 


"Spontaneous 


Artery 


Hospital 


Elsewhere 


Cure" 


Hospital 


Elsewhere 


Cure" 


Aorta 


1 












Innominate 








1 






Internal carotid 


3 






2 






External carotid 


3 












Common 


6 




1 


1 


1 


2 


carotid 














Vertebral 


4 












Lingual 


1 












Occipital 


1 












Cirsoid, nose, 


2 












ear 














Superior 


2 






1 






temporal 














Transverse 


1 


1 










cervical 














Deep cervical 








1 






Internal 


1 












mammary 














Subclavian 


6 


3 


1 


5 






Axillary 


12 






13 


2 




Branch axillary 


4 






2 






Brachial 


11 


1 


1 


22 


5 


1 


Radial 


1 


1 




2 






Ulnar 


4 


2 




2 






External iliac 




1 




1 






Hypogastric 


1 












Superior gluteal 


2 






1 






Obturator 


1 












Common 


3 


2 








1 


femoral 














Femoral 


47 


13 


1 


6 


9 


1 


Profunda 


6 






2 






femoris 














Branch 


2 


2 


1 


2 






profunda 














Popliteal 


41 


1 




14 


6 


1 


Geniculate 


4 












Posterior tibial 


21 


5 




1 


4 


2 


Anterior tibial 


5 


1 




2 


1 




Peroneal 


5 


1 






1 




Branches in 


5 






1 






calf 














Total 


206 


34 


5 


82 


29 


8 



From Shumacker HB Jr, Carter KL: Arteriovenous fistulas and arterial aneurysms in military personnel. Surgery 1946;20:9-25. 



ch24.qxd 4/16/04 3:25 PM Page 468 



468 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



TABLE 24-7 

LOCATION OF TOTAL MAJOR VESSEL LESIONS: ARTERIOVENOUS FISTULAS; 
KOREAN EXPERIENCE 



Vessel 

Common carotid 

Internal carotid 

Subclavian 

Axillary 

Brachial 

Iliac 

Common femoral 

Superior femoral 

Popliteal 

Total 



Arteriovenous Fistulas 

7 

3 

6 

9 
10 

3 

7 
24 
_22 
91 



False Aneurysms 

2 

2 

11 

9 

1 

1 

7 
10 
43 



Total 

9 

3 

8 

20 

19 

4 

8 

31 

32 

134 



From Hughes CW, Jahnke EJ Jr: The surgery of traumatic arteriovenous fistulas and aneurysms: A five-year followup study of 
215 lesions. Ann Surg 1958;148:790-797. 



TABLE 24-8 

LOCATION OF ARTERIOVENOUS FISTULAS: TOTAL MINOR VESSEL LESIONS 
TREATED; KOREAN EXPERIENCE 



Vessel 




Lesions 




Treatment 






Arteriovenous 
Fistulas 


False 
Aneurysms 


Ligation 


Spontaneous 

Closure Anastomosis 


Total 



Occipital 


1 


1 


2 


Supraorbital 


— 


1 


1 


Superior temporal 


2 


1 


3 


Vertebral 


3 


— 


3 


Superior thyroid 


— 


1 


— 


Inferior thyroid 


2 


— 


2 


Thoracoacromial 


2 


— 


2 


Thoracodorsal 


2 


1 


3 


Posterior humeral circumflex 


1 


1 


2 


Subscapular 


1 


— 


— 


Profunda brachii 


— 


1 


1 


Radial 


2 


4 


5 


Ulnar 


2 


2 


4 


Posterior interosseous 


1 


— 


1 


Anterior interosseous 


2 


— 


2 


Digital 


— 


1 


1 


Profunda femoris 


10 


— 


10 


Muscular branch femoral 


— 


1 


1 


Circumflex femoral, lateral 


1 


1 


2 


Inferior genu 


2 


1 


3 


Posterior tibial 


11 


2 


12 


Peroneal 


8 


1 


9 


Anterior tibial 


3 


3 


6 


Dorsalis pedis 


— 


1 


1 


Deep mantar 


_1_ 


— 


J_ 


Total 


57 


24 


77 



2 
1 
3 
3 
1 
2 
2 
3 
2 
1 
1 
6 
4 
1 
2 
1 

10 
1 
2 
3 

13 
9 
6 
1 

J_ 

81 



From Hughes CW, Jahnke EJ Jr: The surgery of traumatic arteriovenous fistulas and aneurysms: A five-year followup study of 
215 lesions. Ann Surg 1958;148:790-797. 



ch24.qxd 4/16/04 3:25 PM Page 469 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 469 



TABLE 24-9 

ARTERIOVENOUS FISTULAS AND 
FALSE ANEURYSMS: ANATOMIC 
LOCATION; VIETNAM VASCULAR 
REGISTRY 

Location No. 



Head/neck 


42 


7.5 


Upper extremity 


134 


24.0 


Thorax 


17 


3.1 


Abdomen 


22 


3.9 


Lower extremity 


343 


61.5 


Total 


558 


100.0 



From Rich NM, Hobson RW II, Collins GJ Jr: Traumatic 
arteriovenous fistulas and false aneurysms: A review of 
558 lesions. Surgery 1975;78:817-828. 



TABLE 24-10 

ARTERIOVENOUS FISTULAS AND FALSE ANEURYSMS: ARTERIAL INJURIES; 
VIETNAM VASCULAR REGISTRY 





Arteriovenous 


False 






Artery 


Fistulas 


Aneurysms 


Total 


O/ 


Common carotid 


6 


5 


11 


2.0 


Internal carotid 


2 


4 


6 


1.1 


External carotid 


2 


3 


5 


0.7 


Vertebral 


6 


2 


8 


1.4 


Subclavian 


1 


7 


8 


1.4 


Axillary 


10 


8 


18 


3.2 


Brachial 


22 


33 


55 


9.9 


Radial 


2 


25 


27 


4.8 


Ulnar 


8 


15 


23 


4.1 


Innominate 


1 


1 


2 


0.4 


Thoracic aorta 





2 


2 


0.4 


Abdominal aorta 





1 


1 


0.2 


Common iliac 


1 


1 


2 


0.4 


External iliac 





6 


6 


1.1 


Internal iliac 





1 


1 


0.2 


Common femoral 


4 


7 


11 


2.0 


Superficial femoral 


57 


31 


88 


15.8 


Deep femoral 


17 


20 


37 


6.6 


Popliteal 


41 


28 


69 


12.4 


Posterior tibial 


30 


33 


63 


11.3 


Anterior tibial 


20 


18 


38 


6.8 


Peroneal 


12 


12 


24 


4.3 


Miscellaneous 


20 


33 


53 


9.5 


Total 


262 


296 


558 


100.0 



From Rich NM, Hobson RW II, Collins GJ Jr: Traumatic arteriovenous fistulas and false aneurysms: A review of 558 lesions. 
Surgery 1975;78:817-828. 



ch24.qxd 4/16/04 3:25 PM Page 470 



470 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



One of the cases was unique in that the authors 
could find no previous report of successful 
repair of a fistula between the aorta, the renal 
vein and the portal vein (Fig. 24-9). They also 
described immediate repair of a mesenteric 
AVF and other fistulas involving the portal 
and renal veins. Dillard, Nelson, and Norman 
(1968) reported one case in which a 29-year- 
old woman was stabbed in the right flank 
and 3 years later was found to have severe 
hypertension. After correction of the renal 
AVF, the patient's blood pressure returned to 
normal. 



Etiology 

Although AVFs may be either acquired or 
congenital, we are essentially concerned with 
those that are acquired by trauma. On the 
other hand, one cannot be knowledgeable 
about acquired AVFs without also under- 
standing the anatomic and pathophysiologic 
aspects of congenital AVFs (Table 24-11). 
Long-standing acquired AVFs must be differ- 
entiated from congenital AVFs, because there 
is a considerable difference in their surgical 
management, as well as the final results. An 




■ FIGURE 24-9 

Abdominal aorta injured by a small-caliber bullet. This angiogram reveals the tip of the catheter in 
the area of injury; the portal vein fills selectively. The additional injury to the renal vein could not be 
shown simultaneously in this unique lesion involving the aorta, the renal vein, and the portal vein. 
(From Hunt TK, Leeds FH, Wanebo HJ, Blaisdell FW: Arteriovenous fistulas of major vessels in the 
abdomen. J Trauma 1971 ;1 1 :483-493.) ■ 



ch24.qxd 4/16/04 3:25 PM Page 471 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



471 



TABLE 24-11 

ARTERIOVENOUS FISTULAS: 10-YEAR 
EXPERIENCE AT THE MAYO CLINIC 



Congenital Acquired 



AV fistulas of the 

extremities 
Aorta-inferior vena 

cava fistulas 
Pulmonary AV fistulas 
Renal AV fistulas 
AV fistulas of the 

portal system 
AV fistulas of the neck 

and face 
Pelvic AV fistulas 
AV fistulas of the chest 

wall 
Total 



80 



47 




11 

1 



139 



17 

7 


6 
1 



5 
2 

42 



AV, arteriovenous. 

Modified from Gomes MMR : Gernatz PE: Arteriovenous 
fistulas: A review and 10-year experience at the Mayo Clinic. 
Mayo Clin Proc 1970;45:81-102. 



acquired AVF can have one, or possibly two, 
communications, whereas the communication 
between the arteries and the veins in the con- 
genital type of AVF may be myriad. 

Usually an AVF results from a simultaneous 
injury of an artery and adjacent vein, which 
permits blood to flow directly from the injured 
artery into the vein. Penetrating injuries are 
usually responsible for these lesions. In mili- 
tary injuries, penetrating missiles are the major 
cause, and in civilian injuries, stab wounds, as 
well as missile wounds, are associated with 
these lesions. The largest series of AVFs have 
been associated with recent combat wounds 
that have occurred during wars in the past 
century (Table 24-12). Both gunshot and 
fragment wounds have created AVFs that have 
been recognized either in the immediate or 
in the acute state or after a delayed period of 
several weeks or months. One of the ironies 
of the combat situation in Vietnam, where 
modern weapons have been employed, is that 
the primitive punji stick has also caused AVFs. 
I saw such an injury of the anterior tibial artery 
and vein at the Second Surgical Hospital in 
1966. 

Vollmar and Krumhaar (1968), based on 
their experience with 200 traumatic AVFs 



TABLE 24-12 

ARTERIOVENOUS FISTULAS AND 
FALSE ANEURYSMS: ETIOLOGY OF 
INJURY; VIETNAM VASCULAR 
REGISTRY 



Wounding Agent 

Fragment 
Bullet 
Blunt 

Punji stick 
Total 



No. 



487 


87.3 


59 


10.6 


7 


1.2 


5 


0.9 


558 


100.0 



From Rich NM, Hobson RW II, Collins GJ Jr: Traumatic 
arteriovenous fistulas and false aneurysms: a review of 558 
lesions. Surgery 1975;78:817-828. 



treated at the Surgical Clinic at the Univer- 
sity of Heidelberg between 1939 and 1967 
(Table 24-13), reported that two world wars 
greatly increased the incidence of traumatic 
AVFs. 

In civilian experience, many AVFs result 
from stab wounds, although they can also be 
caused by bullets. However, these are usually 
low-velocity gunshot wounds. Beall (1963) 
reported that 36 of 50 of AVFs in their 15-year 
study of vascular injuries resulted from 
gunshot wounds (Table 24-14). 

Sako and Varco (1970) reported their expe- 
rience in managing 57 patients with congen- 
ital and acquired AVFs of the extremities, 
abdomen, and chest wall during a 20-year 



TABLE 24-13 

ETIOLOGY OF 200 TRAUMATIC 
ARTERIOVENOUS FISTULAS; 
SURGICAL CLINIC OF THE UNIVERSITY 
OF HEIDELBERG: 1939-1967 



Wounding Agent 



No. 



War projectiles 


177 


88.5 


Fractures 


10 


5.0 


Stab wounds 


7 


3.5 


Iatrogenic trauma 


4 


2.0 


Gunshot wounds (civil) 


2 


1.0 


Total 


200 


100.0 



From Vollmar J, Krumhaar D: In: Traumatic Arterial Lesions. 
Hiertonn T, Rybeck B, eds. Stockholm, Sweden: Research 
Institute of National Defense, 1968. 



ch24.qxd 4/16/04 3:25 PM Page 472 



472 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



TABLE 24-14 

TYPES OF INJURIES RESULTING IN 
ARTERIOVENOUS ANEURYSMS: 
BAYLOR UNIVERSITY COLLEGE OF 
MEDICINE AFFILIATED HOSPITALS 



Type of Injury 



No. 



Gunshot wounds 36 

Stab wounds and lacerations 10 

Shrapnel injuries 3 

Blunt trauma 1 

Total 50 

From Beall AC Jr, Harrington OB, Crawford ES, DeBakey ME: 
Surgical management of traumatic arteriovenous aneurysms. 
Am J Surg 1963;106:610-618. 



period (1949 to 1969). Fewer than 50%, or 
25 patients, had acquired AVFs. The etiology 
of these injuries included small-arms fire in 
nine, penetration with a knife or glass in six, 
a shell fragment or land mine explosion in 
three, multiple puncture for cardiac catheter- 
ization in two, blunt injury of the hand in one, 
pelvic fracture in one, renal needle biopsy 
in one, rupture of an aneurysm in one, and 
following gastrectomy in one. 

Though uncommon, traumatic AVFs have 
been reported after both major and minor 
surgical procedures (Fig. 24-10). The vessels 
that have been involved include the superior 
thyroid (Ranshoff, 1935), renal (Muller and 
Goodwin, 1956), intercostal (Reid and 
McGuire, 1938), uterine (Elkin and Banner, 
1946), and aortocaval (DeBakey, 1958). 
Pridgen and Jacobs (1962) reviewed three 
postoperative AVFs treated in a 3-year period 
at Vanderbilt University. They emphasized the 
necessity for exercising extreme care to avoid 
accidental injury to vessels during any surgi- 
cal procedure. In one of their cases, they also 
emphasized that en masse ligation must be care- 
fully avoided. They felt that the suture liga- 
ture had passed through the right superior 
epigastric artery and vein in one of their 
patients to result in an arteriovenous com- 
munication. AVFs have occurred following 
mass ligature of the renal vessels during 
nephrectomy and of the blood supply to the 
thyroid gland during lobectomy. One case 
report from Walter Reed General Hospital 




■ FIGURE 24-10 

Arteriogram of the aortoiliac vessels 
demonstrating an inferior epigastric artery false 
aneurysm, which occurred as a complication of 
abdominal retention sutures. (From Ello FV, 
Nunn DB: False aneurysm of the inferior 
epigastric artery as a complication of 
abdominal retention sutures. Surgery 
1973;74:460-461.) ■ 



documented the development of an AVF fol- 
lowing subtotal gastric resection (Blackmore 
and Whelan, 1965) (Fig. 24-11). 

Beattie, Oldhan, and Ross (1961) pre- 
sented the case of a 25-year-old man with an 
AVF of the superior thyroid vessels. Approxi- 
mately 18 months earlier, he had a partial thy- 
roidectomy for primary thyrotoxicosis. The 
superior thyroid pedicles were each ligatured 
with one ligature of no. 40 linen thread. 
Approximately 5 months after his partial thy- 
roidectomy, the patient noted swelling in his 
neck and was aware of a "humming" in the 
region of the swelling. After angiographic 
demonstration of the superior thyroid AVF 
between the superior thyroid artery and vein, 



ch24.qxd 4/16/04 3:25 PM Page 473 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



473 




■ FIGURE 24-11 

A, Antegrade aortogram showing larger anomalous artery to the left of the aorta communicating 
with veins in the lower part of the abdomen. B, The specimen in situ showing the artery ending in a 
cul-de-sac communicating with dilated veins. The Kutner dissector has been placed beneath the 
arteriovenous fistula. The transverse colon and mesocolon lie inferior to the fistula. (From Blackwell 
TL, Whelan TJ: Arteriovenous fistulas as a complication of gastrectomy. Am J Surg 1965;109:197- 
200.) ■ 



excision of the remnant of the left lobe of the 
thyroid gland was accomplished. 

There have been unusual forms of AVFs 
reported following essentially every type of sur- 
gical operation and every type diagnostic or 
therapeutic procedure: For example, an 
AVF was reported following removal of an 
intervertebral disk with injury to the iliac 
artery and vein. Another such fistula oc- 
curred following a percutaneous transaxillary 
angiogram performed at Walter Reed General 
Hospital. Lester (1966) described AVFs as a 
complication of selective vertebral angi- 
ography. One of these lesions has also been 
treated at Walter Reed General Hospital. 

White, Talbert, and Haller (1968) stated 
that there was an increasing awareness of 
peripheral arterial injuries in infants and chil- 
dren. One of their patients, a 3-month-old 
female, had a right femoral vein right heart 



catheterization to investigate a small ventric- 
ular septal defect and mild pulmonic steno- 
sis. Over the following 3 years, she developed 
borderline heart failure, with a pulse rate of 
120 and an increase in her heart size. At the 
age of 4.5 years, a thrill was noted over the 
left groin, and the left leg was 2 cm longer than 
the right. The proximal fibula was present on 
the left and not on the right. A large AVF (Fig. 
24-12) was demonstrated between the pro- 
funda femoris artery and profunda femoris 
vein. Arterial blood gases had been measured 
and samples obtained from arterial punctures 
of the right femoral artery. The needle must 
have been inserted in a lateral and downward 
direction, penetrating the femoral vein before 
puncturing the femoral artery for the blood 
samples. A direct AVF was created by the 
needle. After ligation of this fistula and 
without sacrifice of either the artery or the 



ch24.qxd 4/16/04 3:25 PM Page 474 



474 



V • SPECIAL PROBLEMS AND COMPLICATIONS 




■ FIGURE 24-12 

Angiogram demonstrating an arteriovenous 
communication between the right profunda 
femoris artery and the deep femoral vein 
following arterial puncture for blood gas 
analysis. Over the subsequent 3 years, the 
patient was in borderline heart failure, with an 
increased heart rate and increased growth in 
her lower extremity. (From White JJ, Talbert JL, 
Haller JA Jr. Peripheral arterial injuries in infants 
and children. Ann Surg 1968;167:757-766.) ■ 



vein, over the next several months, her pulse 
rate gradually returned to normal and her 
cardiac failure decreased. 

Lord, Ehrenfeld, and Wylie (1968) pre- 
sented the case of a profunda femoris AVF 
caused by passage of a Fogarty arterial catheter. 
At the time of their report, they stated that 
there were two other similar incidences in the 
literature. Subsequent reports include those 
of Rob and Battle (1971) and Gaspard and 
Caspar (1972). 

AVFs occasionally occur with fractures 
(additional details are given in Chapter 5) . 
Harris (1963) reported an AVF following 
closed fracture of the tibia and fibula in a 35- 
year-old man (Fig. 24-13). Vascular injuries 
have occurred with orthopedic procedures 
rather than those involved in the management 
of fractures. Ferguson (1914) presented an 
infant who developed an AVF following an 
osteotomy of the femur genu valgum. 




■ FIGURE 24-13 

A, Closed fracture of the tibia and fibula; the arteriogram shows an arteriovenous fistula of the 
peroneal artery and the arterial phase of filling. B, The arteriogram shows the venous phase of filling 
of the peroneal arteriovenous fistula. C, A postoperative arteriogram following excision of the fistula. 
Note the rapid advance and union of the fracture of the tibia and fibula following excision of the 
fistula. (From Harris JD: A case of arteriovenous fistula following closed fracture of tibia and fibula. 
Br J Surg 1963;50:774-776.) ■ 



ch24.qxd 4/16/04 3:26 PM Page 475 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



475 



Anthopoulos, Johnson, and Spellman 
(1965) reported on the unusual case of 23- 
year-old woman who at age 9 years had sus- 
tained a human bite at the base of the finger. 
The authors believed that the AVF of the fifth 
finger developed as a complication of the 
human bite. There was spontaneous, periodic 
subungual spurting of the arterial blood, as 
well as increased growth, venous distention, 
increased local temperature, and more rapid 
growth of the nail. Surgical excision of 
the aneurysmal sacs and ligation of visible 
communications on two separate occasions 
resulted in relief of symptoms and enabled 
the patient to resume her occupation as a 
typist. 



Pathophysiology 



As a student at the Johns Hopkins Medical 
School in 1917, my curiosity about 
arteriovenous fistulas was aroused and 
repeatedly whetted by Doctor Halsted's 
recurrent expressions of great puzzlement 
at the occasional massive enlargement of 
the heart to the point of cardiac failure and 
at the marked dilatation of the proximal 
artery that could accompany an 
arteriovenous fistula, usually one long 
duration. Equally puzzling was the fact that 
this heart enlargement and arterial dilatation 
occurred with some but not all fistulas. 
Holman, 1971 



There is a sense of anatomic and pathologic 
changes that evolve when an AVF is produced 
(Fig. 24-14). An AVF is an abnormal com- 
munication between the arterial and venous 
systems that creates a shorter circuit in rela- 
tion to the heart by allowing blood to pass from 
the higher peripheral resistance of the arter- 
ial system to the lower peripheral resistance 
of the venous system (Fig. 24-15). The sec- 
ondary circuit, which has a constant tendency 
to divert the arterial blood into the lower 
resistance venous system through the fistula, 
causes a number of hemodynamic distur- 
bances. The effective systemic blood flow is 
reduced, and there is a decreased mean sys- 




■ FIGURE 24-14 

This schema shows the circulation in the 
presence of a right femoral arteriovenous fistula 
establishing a second circuit of blood. A 
progressively increasing volume of blood is 
sequestered in circuit B as long as resistance 
in the fistula circuit is less than resistance in the 
capillary bed in circuit A. (From Holman E: 
Arteriovenous aneurysms: Abnormal 
communication between the arterial and 
venous circulations. New York: Macmillan, 
1937.) ■ 



temic arterial pressure. However, there is an 
increase in the blood volume, total cardiac 
output, stroke volume, heart rate, left arter- 
ial pressure and pulmonary arterial pressure, 
as has been described by Holman (1937, 
1968). Holman also emphasized that the size 
of the AVF, the location of the communica- 
tion in the vascular tree and the distensibility 
of the vascular rim are the factors that deter- 
mine the volume of the blood that is diverted 
through the AVF's border permits progressive 
increase of the blood shunted through the 
secondary circuit of the fistula, with additional 



ch24.qxd 4/16/04 3:26 PH Page 476 



476 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



Artery 



B 




■ FIGURE 24-15 

A, Immediately following the development of an 
arteriovenous fistula, there is shunting of blood 
from the artery through the fistula into the vein, 
from which it returns to the heart. This results in 
a decrease in peripheral vascular resistance, a 
decrease in diastolic blood pressure, and an 
increase in heart rate. The venous pressure 
rises in the involved vein. Peripheral blood flow 
is decreased in the involved artery. B, After 
several weeks, collateral circulation enlarges 
around the fistula because of the decreased 
vascular resistance at the site of the fistula. As 
the collateral circulation develops, the involved 
artery and vein also dilate, increasing the 
amount of blood flowing through the fistula. 
C, After several years, extensive dilation may 
develop about a fistula with marked 
enlargement of collateral circulation. In 
addition, there is enlargement of the artery 
immediately distal to the fistula, through which 
blood flows in a retrograde fashion through the 
fistula toward the heart. The vein may enlarge 
to marked proportions, creating varicosities in 
the extremity. Ultimately such progressive 
dilation after some years may result in 
congestive heart failure from the increased 
cardiac output. (From Spencer FC, Schwartz 
SI: Principles of surgery. New York: McGraw- 
Hill, 1974.) ■ 



increase in the blood volume and dilatation 
of the heart. Lewis (1940) demonstrated that 
the entire circuit gradually dilates to accom- 
modate the increased volume of blood flow; 
this includes dilatation of the cardiac cham- 
bers, the arterial tree proximal to the fistula, 
the proximal vein and vena cava and even the 
AVF itself. Nakano and DeSchryver (1964) 
studied the effects of AVFs on systematic and 
pulmonary circulation and stated that the 
increase in cardiac output was essentially a 
result of the increase in stroke volume, noting 
that the heart rate may change very little. 

Holman (1965) reviewed abnormal arteri- 
ovenous communications with particular ref- 
erence to the delayed development of cardiac 
failure. He emphasized that low resistance in 
the venous system to the shunt of blood at the 
site of the fistula and the decrease in periph- 
eral perfusion distal to the AVF were strong 
stimuli for the development of collateral 
circulation. Holman (1940) documented 
significant structural changes in both the 
arteries and the veins associated with the 
hemodynamic disturbances of an AVF. With 
a small communication, the vein gradually 
assumes the appearance of an artery, and it 
may not be easily distinguished from the artery 
at the end of 6 to 9 months. In contrast, with 
larger fistulas, the vein may become so dis- 
tended that it appears to be a false aneurys- 
mal sac. As has been known since the first 
description by Hunter in 1 757, the artery prox- 
imal to the AVF can be dilated; however, 
Holman (1940) stated that the dilatation of 
the artery can also occur distal to the fistula. 
It is not the initial injury that creates the AVF; 
the arterial walls at the fistula or proximal to 
it may become rigid as a result of deposition 
of fibrous tissue, or the lumen may even 
become stenotic by contraction of surround- 
ing fibrous tissues. 

AVFs may be associated with decreased 
resistance in the peripheral arterial tree. The 
consequent enlargement of superficial ven- 
ous collaterals can be mistaken for changes 
associated with chronic venous insufficiency. 
One Vietnam casualty seen subsequently at 
Walter Reed General Hospital had been 
treated for varicose veins of his left lower 
extremity for 5 years, when in actuality the 
increase in his left thigh and associated 



ch24.qxd 4/16/04 3:26 PH Page 477 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



477 



superficial varicosities were associated with an 
acquired femoral AVE 

According to Petrovsky and Milinov (1967) , 
the structural changes that occur in the walls 
of both the arteries and the veins associated 
with an AVF are called "venization" of ar- 
teries and "arterialization" of veins. The 
alterations in the venous walls are easier to 
understand because they can be caused by an 
abrupt increase in the venous pressure and 
can be a consequence of adaptions. There is 
more difficulty in understanding the changes 
in Petrovsky and Milinov ( 1967) — performed 
experiments, which showed thickening of the 
media of the venous wall due to an increase 
in the amount of muscular and connective 
tissue elements, marked elastosis of all layers 
of the vessel wall, intimal thickening, and an 
increase in the vasa vasorum, which made it 
resemble the wall of an artery. They saw an 
increase in muscular fibers and fibrosis in the 
arterial wall, with a corresponding increase 
of mucopolysaccharides and extracellular 
fibers, elastosis and later dystrophy of the 
elastic fibers, focal necrosis of connective tissue 
elements in the adventitia, and diminution of 
the vasa vasorum. It was felt that a decrease 
in the oxidative process accounted for the 
accumulation of mucopolysaccharides, the 
extracellular fibrosis, and the elastolysis 
(decrease in oxidative process and tissue 
hypoxia, which results from a decreased blood 
supply in the arterial wall) . 

Holman (1940) stated that hemodynamic 
changes caused by AVFs were reversible. 
However, some structural changes may notbe 
reversible, such as dilatation of the proximal 
artery associated with a long-standing AVF, 
which may not regress if aneurysmal deterio- 
ration of the wall has occurred. Also, cardiac 
enlargement associated with long-standing 
AVFs and dilatation may not revert to normal. 

Eisenbrey (1913) emphasized the extensive 
alterations in both the artery and the vein up 
to the bifurcation of the aorta and vena cava 
in a patient with a superficial femoral AVF (see 
Fig. 24-2). The patient complained of short- 
ness of breath and presented symptoms of 
cardiac insufficiency. Eighteen years earlier, 
the patient had been shot in the thigh with 
a small-caliber (probably .22-caliber) rifle 
bullet. Terminal illness allowed necropsy 



examination of the aneurysmal dilatation and 
tortuosity of the artery and vein. 

Subsequent studies have augmented the 
original and monumental contributions of 
Holman. Schenk and colleagues (1957) eval- 
uated the regional hemodynamics of experi- 
mental acute AVFs. Their objective was to use 
the newer electronic methods for pressure and 
flow measurements to investigate the pressure- 
flow changes that occurred immediately 
upon opening an experimental fistula. Figure 
24-16 summarizes the pressure-flow data in 
a representative model. 

Johnson, Peters, and Dart (1967) studied 
the cardiac vein negative pressure in AVFs with 
a plastic model. They demonstrated creation 
of negative pressure in the cardiac vein, the 
result of transformation of energy, and 
explained this by the use of the principles of 
flow through a conduit (Fig. 24-17). 

Johnson and Blythe (1970) evaluated eight 
patients with AVFs created for hemodialysis 
over a period of 3 years. Their study demon- 
strated that peripheral AVFs created for 
hemodialysis in patients with chronic renal 
failure result in a slight increase in cardiac 
output and pulse rate and a decrease in the 
total peripheral resistance. Although these 
alterations in hemodynamics did not lead 
to perceptible cardiac strain, a warning was 



PRESSURE -MM.U 3 




FLOW - CC/MIN. 

PROXIMAL PROXIMAL 
ARTERY VEIN 



305 , 



ms/isl 

/OS)/ 


\90/£0\ 
UTS) \ 


/ / 


\ IT0\ 


DISTAL 


DISTAL 


DISTAL 


01STAL 


ARTERY 


VEIN 


ARTERY 


VEIN 



■ FIGURE 24-16 

Schematic summary of pressure-flow data in a 
representative animal after a large femoral 
arteriovenous fistula was opened. (From 
Schenk WG Jr, Bahn RA, Cordell A, Stephens 
JG: Surg Gynecol Obstet 1957;105:733.) ■ 



ch24.qxd 4/16/04 3:26 PH Page 478 



478 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



CARDIAD 
ARTERY 



CARDIAD 
VEIN 




PERIPHERAL PERIPHERAL 
ARTERY VEIN 

m FIGURE 24-17 

Flow pattern through fistula. In model, pressure 
at 1 was -10 mm Hg, at 2 it was -5 mm Hg, and 
at 3 it was -4 mm Hg, emphasizing the 
negative pressure in the cardiac vein and 
arteriovenous fistula. (From Johnson G Jr, 
Peters RM, Dart CH Jr: A study of cardiac vein 
negative pressure in arteriovenous fistula. Surg 
Gynecol Obstet 1967;124:82-86.) ■ 




■ FIGURE 24-18 

This small fragment wound of the upper right 
thigh created an arteriovenous fistula that was 
not diagnosed initially. The surrounding 
increased density on the roentgenogram was 
caused by an associated pulsating hematoma. 
(From Rich NM: Vascular trauma in Vietnam. 
J Cardiovasc Surg 1970;11:368-377.) ■ 



made that physicians managing these patients 
should be cognizant of this possibility, espe- 
cially in patients on long-term hemodialysis. 



Clinical Pathology 

The capillary circulation is bypassed in an AVF 
when there is a direct communication between 
an artery and a vein. Although this type of 
communication can be a normal function of 
the microcirculation, the AVF becomes patho- 
logic when its size or location causes signifi- 
cant hemodynamic alterations. An AVF may 
be established immediately after a penetrat- 
ing injury in which blood flows directly from 
the injured artery into the vein. On the other 
hand, thrombus may surround the AVF, and 
the communication may not be obvious until 
days or weeks later when the surrounding clot 
becomes liquefied (Fig. 24-18). 

Once atraumatic AVF has been established, 
there is usually little difficulty in its recogni- 
tion. The previous history of trauma, the 
finding of a prominent pulsation and palpa- 
ble thrill, and the presence of an audible 



machinery-like murmur, or any combination 
of these findings should alert one to the pres- 
ence of an AVF. A bruit often appears over 
the sight of arteriovenous communications 
within a matter of hours after the establish- 
ment of the lesion. Other signs and symptoms 
that can develop distal to an AVF include inter- 
mittent claudication, edema (Fig. 24-19), and 
prominent veins (Fig. 24-20) , which are often 
accompanied by bluish discoloration of the 
skin and venous stasis. The last two findings 
result from shunting of the arterial blood into 
the venous system. 

More than 200 years ago, in 1757 William 
Hunter recognized an abnormal communi- 
cation between an artery and a vein and accu- 
rately described the thrill and bruit associated 
with the communication. He noted that he 
can eliminate both the thrill and the bruit by 
pressure over either the proximal artery or 
the site of the communication. He also doc- 
umented his observation of tortuosity and dila- 
tion of the artery proximal to the fistula. 

Nicoladoniin 1875 is generally given credit 
for being the first to demonstrate the remark- 
able fact that the pulse rate could be lowered 



ch24.qxd 4/16/04 3:26 PH Page 479 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



479 




■ FIGURE 24-19 

Edema can be associated with arteriovenous fistulas. The massive swelling of the left lower 
extremity in this Vietnam casualty is obvious. He had a femoral arteriovenous fistula of 5 years' 
duration; however, he had been treated as a patient with varicose veins. (NMR Vietnam Vascular 
Registry #630 1972.) ■ 




■ FIGURE 24-20 

The position of an arteriovenous fistula and pulsating venous lakes 
(circled); note the difference in size of the two lakes. The site of the 
fistula is indicated by a cross. (From Holman EF: Arch Surg 
1923;7:64-82.) ■ 



ch24.qxd 4/16/04 3:26 PH Page 480 



480 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



by compression of the artery proximal to the 
AVE Fifteen years later, in 1890 Branham again 
called attention to the reduction of the pulse 
rate by obliteration of a large acquired AVE 
This phenomenon is frequently referred to 
as the "Branham-Nicoladoni sign." 



oftheheartin the presence of an AVE In 1913, 
Stewart noted that the heart diminished in 
size within 10 days after elimination of the 
fistula. 



Clinical Features 



The most mysterious phenomenon connected 
with the case, one which I have not been 
able to explain myself, or to obtain a 
satisfactory reason for from others, was 
slowing of the heart's beat, when 
compression of the common femoral was 
employed. This began to be noticeable after 
the wound had entirely healed. The patient 
was apparently well, with exception of the 
injured vessel, which necessitated his 
confinement to bed. This symptom became 
more marked until pressure of the artery 
above the wound caused the heart's beat to 
fall from 80 to 35 to 40 per minute, and so 
remain until the pressure was relieved. 
Harris H. Branham, 1890 



While working as a student of Halsted, Reid 
(1920) established that there was enlargement 



If the patient has had a penetrating injury, 
the possibility of an AVF must be recognized; 
however, this may not be immediately obvious. 
As previously noted, if the arteriovenous com- 
munication has surrounding thrombus, the 
classic findings of the thrill and bruit may not 
exist until several days or weeks later. There 
may be little evidence of vascular trauma in 
the way of blood loss or loss of peripheral 
pulses (Fig. 24-21). The patient may or may 
not be aware of a buzzing sensation when his 
fingers are placed over the area of the arteri- 
ovenous communication. One patient in the 
registry had originally been wounded in 
Korea; however, it was nearly 15 years later, 
when he was piloting a helicopter in Vietnam, 
that he noticed a buzzing sensation in his 
popliteal fossa. It may be more unusual for 
the patient to present with one of the com- 
plications of AVF, such as infection within the 




■ FIGURE 24-21 

A small wound may deceive the casual observer as to the extent of underlying vascular pathology. 
(NMR Vietnam Vascular Registry #2513 1971 .) ■ 



ch24.qxd 4/16/04 3:26 PH Page 481 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



481 



vascular system, peripheral embolization, or 
congestive heart failure. 

Errors in diagnosis can exist. Patients with 
AVFs have been treated for years for varicose 
veins (see Fig. 24-19). Venous hypertension 
with resultant varices, peripheral pigmenta- 
tion, and ulceration from venous insuffi- 
ciency can confuse the diagnosis; however, the 
classic findings of a thrill and bruit should be 
carefully sought. 

There may or may not be a soft diffuse mass 
on physical examination. Depending on the 
period of time that the AVF has existed, dilated 
veins may surround the area. A thrill, with its 
maximal component during systole, is usually 
felt very easily on palpation. A "machinery 
murmur" is usually heard easily on ausculta- 
tion, the loudest part of the continuous 
murmur occurring during systole. Detection 
of this classic finding differentiates an 
AVF from an arterial false aneurysm. The 
Nicoladoni-Branham sign, which has been 
previously described, is another significant 
finding if a slowing pulse can be demonstrated 
when the fistula is obliterated by digital com- 
pression. Ironically, this test was not positive 
in many of the Vietnam casualties with AVFs. 
The peripheral resistance increases when the 
fistula is digitally occluded, causing the blood 
pressure to rise, with reflex slowing of the heart 
rate and consequent slowing of the pulse. The 
temporary bradycardia results from a neuro- 
genic reflex mediated through pressure- 
sensitive receptors in the carotid sinuses and 
great vessels. 

With large AVFs and large shunting of 
blood, cardiac enlargement and, more rarely, 
cardiac failure may occur (Fig. 24-22) . Smith 
(1963) found the most serious complication 
of AVF, left ventricular myocardial failure, in 
two of their patients. One of these was a 16- 
year-old male who had been shot in the right 
thigh with a .22-caliber rifle bullet. Nine days 
after the accident, the patient developed a 
gallop rhythm and severe dyspnea. A chest 
roentgenogram revealed a marked enlarge- 
ment of the cardiac shadow. An emergency 
operation was performed to correct a 
common femoral arteriovenous communica- 
tion. The signs of congestive cardiac failure 
regressed in 3 weeks. The authors pointed out 
that there was a regrettable error of omission 



in the immediate exploration of the wound. 
They felt that the rapid development of 
cardiac decompensation, which made surgi- 
cal intervention most urgent, was an unusual 
aspect of the case. 



Diagnostic Considerations 

The history of a penetrating injury and the 
classic physical findings usually establish 
the diagnosis of an AVF. Establishment of the 
diagnosis may be more difficult if the lesion 
is within the thoracic or abdominal cavity. 
Angiography readily demonstrates the rapid 
filling of an adjacent vein and increased col- 
lateral circulation (Fig. 24-23) . Angiographic 
demonstration of most arteriovenous lesions 
is usually not necessary from the diagnostic 
standpoint, but it may be helpful in planning 
the surgical correction. This is particularly true 
if multiple arteriovenous communications 
exist, or if one or more false aneurysms are 
associated with the arteriovenous communi- 
cation (Fig. 24-24) . Bell and Cockshott (1965) 
demonstrated the angiographic features 
found in patients with both acute and chronic 
AVFs. 

Cardiac enlargement may be noted on 
roentgenogram of the chest. Shumacker and 
Stahl (1949) evaluated the cardiac frontal area 
in patients with AVFs to determine the heart 
size before and after operative obliteration of 
the fistula in a large group of patients. They 
studied 185 soldiers with traumatic peripheral 
AVFs of relatively short duration. Cardiac 
enlargement was noted in a large number 
before operative excision of the fistula, and 
the reduction in the heart size occurred in a 
comparable number after operation. These 
authors believe that the location of the fistula, 
the size of the artery involved, the size and 
age of the fistula, and the magnitude of 
the pulse and blood pressure response to 
temporary occlusion of the fistula could be 
correlated with the tendency toward early 
development of cardiac enlargement. These 
studies showed conclusively that demon- 
strable evidence of cardiac enlargement was 
present in approximately 50% of young sub- 
jects with peripheral AVFs of relatively short 
duration; however, few had symptoms of 



ch24.qxd 4/16/04 3:26 PM Page 482 



482 



V • SPECIAL PROBLEMS AND COMPLICATIONS 




■ FIGURE 24-22 

A, Cardiac enlargement may occur with large arteriovenous fistulas and occasionally progress to 
cardiac fistulas and occasionally progress to cardiac failure. B, This patient had an arteriovenous 
fistula 1 .0 x 1.5cm between the right common iliac artery and the left common iliac vein following 
disk surgery. C, The heart returned to normal size limits after closure of the fistula by lateral suture of 
the vein and resection of a small segment of artery followed by end-to-end anastomosis. (From 
Jarstfer BS, Rich NM: The challenge of arteriovenous fistula formation following disk surgery: A 
collective review. J Trauma 1976;16:726-733.) ■ 



ch24.qxd 4/16/04 3:26 PH Page 483 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



483 




■ FIGURE 24-23 

A, Angiography is helpful in identifying the site of communication in an arteriovenous fistula. Left, 
Contrast media descends in the artery to the fistula. Right, The dilated veins are then rapidly 
visualized by passage of contrast media through the fistula. B, From left to right, the distal 
superficial femoral artery is visualized angiographically with rapid filling of the adjacent vein. 
(A, From NMR, Vietnam Vascular Registry #7182; B, from Vietnam Vascular Registry #2760, Walter 
Reed General Hospital.) ■ 



ch24.qxd 4/16/04 3:26 PM Page 484 



484 



V • SPECIAL PROBLEMS AND COMPLICATIONS 




■ FIGURE 24-24 

Multiple false aneurysms and arteriovenous 
fistulas were demonstrated angiographically in 
this Vietnam casualty. The extent of pathologic 
involvement of the posterior tibial and peroneal 
vessels could not be determined clinically. 
(From Vietnam Vascular Registry #2761, Walter 
Reed General Hospital.) ■ 



cardiac strain, and there was essentially no 
evidence of cardiac failure. This fact was 
confirmed by a measurable reduction in car- 
diac size after operation in a comparable per- 
centage. Cardiac failure has been reported in 
a series of 14 patients (Pate and colleagues, 
1965). 



Surgical Treatment 

A literature review and a successful personal 
repair of an AVFs prompted Stewart (1913) 
to write, "With angiorrhaphy the aneurysm 
can be dealt with radically and the vessels 
conservatively, thus effecting cure without 
interrupting the bloodstream and without pro- 
ducing gangrene." 

Despite his interest in repair of AVFs, he 
noted that suture of vessels was not always pos- 



sible. He cited that in a number of instances, 
the surgeon had planned to repair the vessel 
but was forced to abandon the idea because 
of hemorrhage (Delanglade), friability of 
the artery, (Thompson) , the large size of 
the opening (Mignon), dense adhesions 
(Cranwell), or obliteration of the sutured 
vessel (Cestan). 

Shumacker (1948) stated, "It has long been 
recognized that the ideal method of treating 
aneurysms and arteriovenous fistula involving 
important arteries is the extirpation of the 
lesion combined with some procedure which 
permits maintenance or re-establishment of 
the continuity of the affected artery." 

The best time for surgical cure of a trau- 
matic AVF is immediately after the establish- 
ment of the communication. 

There has been a period of profound 
changes in the surgical management of AVFs. 
Surgeons' energies were formally directed 
toward the selection of a time when maximal 
collateral circulation would have developed. 
If the intervention was properly timed and 
if the collateral circulation was adequate, 
ligation of the four component vascular 
trunks without excision of the fistula cured 
the lesion, the extremity remaining viable 
despite the fact that there might be some arte- 
rial and venous insufficiency. The important 
change, which was strongly influenced by the 
experience at Walter Reed General Hospital 
in managing Korean battle casualties, con- 
centrated on dissection of the proximal and 
distal communicating artery and vein with 
repair of defects in both vessels (Seeley and 
colleagues, 1952). 

The initial treatment of AVFs included a 
delay of 2 to 6 months to allow collateral cir- 
culation to develop. It was anticipated that this 
would improve extremity survival after liga- 
tion of the involved artery. However, it should 
be emphasized that some of the earliest 
vascular repairs, including venorrhaphy and 
arteriorrhaphy, of this century involved AVFs. 
At present, division of the fistula with venous, 
as well as arterial, repair is preferred. Exci- 
sion with multiple ligations is accepted for 
smaller vessels not essential to normal circu- 
lation, such as one of the tibial arteries or veins. 
There is currently an interest in treating AVFs 
when they are initially diagnosed. If the lesion 



ch24.qxd 4/16/04 3:26 PH Page 485 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



485 




■ FIGURE 24-25 

Proximal and distal control of both the artery and the vein is important in the successful 
management of arteriovenous fistulas. It was possible to repair the popliteal vein by lateral suture 
and the popliteal artery by an end-to-end anastomosis to correct the arteriovenous fistula. (From 
Rich NM: In: Beebe HG, ed. Complications in vascular surgery. Philadelphia: JB Lippincott, 
1973.) ■ 



is discovered on the third or fourth day after 
an injury, it is usually better to wait until 3 
weeks have passed to allow soft tissue healing 
to occur and edema to subside. 

Elective incisions should be used, as was 
emphasized for arterial repair and for venous 
repair. Adequate exposure of the artery and 
vein proximal and distal to the fistula should 
be accomplished before the fistula is directly 
approached (Fig. 24-25). When these vessels 
are isolated and temporarily occluded, the 
arteriovenous communication can be incised 
and directly isolated (Fig. 24-26). There 
usually is a small lesion between the artery 
and the vein, and the size of the surrounding 
false sac may vary greatly. Nevertheless, a small 
segment of the artery is usually involved, the 
remainder of the artery being freed from the 
false sac during mobilization to perform the 
arterial repair. Lateral venorrhaphy is usually 
possible. Only in large arteries is arterior- 
rhaphy by lateral suture possible. Frequently, 
minimal excision of the damaged artery and 




■ FIGURE 24-26 

Proximal and distal control of both artery and 
vein has been obtained with silk suture loops in 
place. Multiple ligatures with excision of the 
arteriovenous fistula and false aneurysm were 
elected for this distal posterior tibial lesion. 
(From NMR, Vietnam Vascular Registry #1806, 
1969.) ■ 



ch24.qxd 4/16/04 3:26 PH Page 486 



486 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



end-to-end anastomosis are possible. Other- 
wise, segmental replacement with autogenous 
greater saphenous vein is preferred. 

Intra-arterial balloon catheter control of 
hemorrhage and many other useful tech- 
niques used in arterial repair are covered in 
more detail in Chapter 19 and in the discus- 
sions of specific arteries. LeVeen and Cerruti 
(1963) described a method for intra-arterial 
balloon tamponade of blood vessels in the 
surgical management of AVFs (Fig. 24-27). 
Noon (1969) emphasized the use of balloon 
catheters to provide temporary arterial occlu- 
sions for control of hemorrhage. The use of 



multiple balloon catheters to control intra- 
arterial hemorrhage and venous bleeding 
has been successful in the management of 
Vietnam casualties with AVFs at Walter Reed 
Army Medical Center (Fig. 24-28). 



Spontaneous Cure 

Shumacker and Wayson (1950) evaluated 
spontaneous cure of aneurysms and AVFs. 
They studied 122 aneurysms and 245 AVFs. 
Thrombosis appeared to be responsible for 
the obliteration of the lesions. Fibrosis can also 



MEf tiger ft 4, TcrtHou* 
JMt*rr>4.' 




/*46&mtk/jfo&ktJtm 



n j-f v t4 



■ FIGURE 24-27 

A Woodruff catheter was inserted through the open brachial artery and the balloon inflated at the 
region of the arteriovenous fistula. A, Dilated vessels anterior to the fistula. This was a method for 
intra-arterial balloon tamponade of blood vessels in the surgical management of arteriovenous 
fistulas. (From LeVeen HH, Cerruti MM: Surgery of large inaccessible arteriovenous fistulas. Ann 
Surg 1963;158:285-289.) ■ 



ch24.qxd 4/16/04 3:26 PH Page 487 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



487 




■ FIGURE 24-28 

Intraoperative photograph 
showing a Fogarty balloon 
catheter in the distal internal 
carotid artery for intraluminal 
arterial control of hemorrhage 
(small arrow) and a Foley 
balloon catheter inserted into 
the adjacent internal jugular 
vein for temporary control of 
venous hemorrhage during 
repair of a distal 
carotid-internal jugular 
arteriovenous fistula. (From 
NMR, Walter Reed General 
Hospital, 1972.) ■ 



occur as a more gradual process. Because there 
are only five spontaneous cures, only 2% of 
the 245 AVFs (four of five arteriovenous lesions 
that healed spontaneously did so suddenly and 
within three months of the original injury), 
the authors stated, "Satisfactory spontaneous 
cures occurred in our series so infrequently 
as to make consideration of this possibility of 
little or no importance in reaching a decision 
as to the necessity for or the proper time for 
surgical treatment of the lesion." 

Spontaneous closure of the AVFs has been 
a relatively unusual event. Billings, Nasca, and 
Griffin (1973) reported one such instance in 



a 19-year-old Marine who had sustained 
multiple fragment wounds from a land mine 
explosion in August 19 70 while on duty in the 
Republic of Vietnam. In September, a 3-cm 
pulsating mass was noted in the right axilla, 
and there was continuous bruit and thrill over 
the mass. An axillary AVF was demonstrated 
by angiography (Fig. 24-29). Treatment of 
other multiple wounds was carried out, 
and during the first week in November, the 
axillary mass was no longer palpable. A second 
arteriogram demonstrated that there had 
been spontaneous closure of the AVF 
(Fig. 24-30) . Two similar patients — Vietnam 




■ FIGURE 24-29 

Subtraction print demonstrating the early arterial phase of an arteriogram performed via subclavian 
injection. The right axillary artery (-t+») communicates with the axillary vein (— >) through a large 
arteriovenous fistula (+-»). (From Billings KJ, Nasca RJ, Griffin HA: Traumatic arteriovenous fistula 
with spontaneous closure. J Trauma 1973;13:741-743.) ■ 



ch24.qxd 4/16/04 3:26 PH Page 488 



488 



V • SPECIAL PROBLEMS AND COMPLICATIONS 




■ FIGURE 24-30 

A second arteriogram performed 6 weeks later, demonstrating that the arteriovenous 
communication in the axillary vessels (see Fig. 24-29) is no longer present. Elective operative 
closure had not been performed because of a wound infection. The site of "thrombosis" is visible as 
a small, contrast-filled saccule on the inferior surface of the axillary artery. (From Billings KJ, Nasca 
RJ, Griffin HA: Traumatic arteriovenous fistula with spontaneous closure. J Trauma 1973;13:74-1- 
743.) ■ 



casualties — were seen at Walter Reed General 
Hospital. 



Results 

Annandale (1875) reported the successful lig- 
ature of a popliteal artery and vein in the 
treatment of a traumatic popliteal AVE Pick 
(1883) reported the case of a 28-year-old man 
who sustained a gunshot wound of the thigh 
with a resultant AVF of the femoral vessels. 
He stated that the only operative procedure 
that appeared to hold any hope for success 
was ligature of the artery above and below 
the point of communication. According to 
Murphy (1897), Von Zoege-Manteuffel suc- 
cessfully repaired a femoral arteriovenous 
aneurysm by lateral suture of the wall in 1895. 
When the first end-to-end arterial anastomo- 
sis in a human was reported by Murphy (1897) , 
he described his successful treatment in 1896 
of a common femoral AVF. In addition to the 
end-to-end arterial anastomosis following 
resection of the damaged portion of the artery, 
he closed the wound in the vein by lateral 
venorrhaphy. Bickham (1904) suggested 
that Matas endoaneurysmorrhaphy could be 
employed for the intravascular repair of AVFs. 
He also recommended transverse closure of 
the defects in the vascular walls as a practical 



method of preserving the continuity of both 
the injured artery and the injured vein. Matas 
emphasized the reason for failure when partial 
ligation was used in the treatment of AVFs was 
the remaining patency in other vessels not 
ligated (Fig. 24-31). 

Soubbotitch (1913) reported on the mili- 
tary experience in the Serbo-Turkish and 
Serbo-Bulgarian Wars. There were 16 differ- 
ent surgeons who performed ligation of large 
vessels on 41 arteries and 4 veins and partial 
suture on 17 vessels — 8 arteries and 9 veins. 
Circular suture was employed on 15 vessels — 
11 arteries and 4 veins — to bring the total 
number of vessels sutured to 32 (19 arterior- 
rhaphies and 13 venorrhaphies). The 60 
arteries and 17veinsmadeofatotal 77 injuries 
to the larger blood vessels among 20,000 
wounded. Osier (1915) stated that there was 
agreement with a conclusion arrived at by 
Soubbotitch, senior surgeon at the Belgrade 
State Hospital, from his experience in the 
Balkan War, "that arteriovenous aneurysms 
should be operated upon, as they offer small 
prospect of spontaneous cure, although they 
often remain stationary for a long time and 
cause relatively little trouble." 

World War I contributed little significant 
data compared to World War II. Because 
competent vascular surgeons had chosen to 
head three centers for vascular surgery during 



ch24.qxd 4/16/04 3:26 PH Page 489 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 489 
AKTERX 



ART&RY 



E-PF-BRWt 
Y&IH 




EPPfcMST ARTERY 



A.WERBNT VEIN 



V£IH 



■ FIGURE 24-31 

Schematic drawing showing the communications of an arteriovenous fistula and the necessity of not 
only quadruple ligation but also complete excision including all branches. (From Matas R: Military 
surgery of the vascular system. In: Keene's surgery, vol. 7. Philadelphia: WB Saunders, 1921.) ■ 



World War II, a large number of AVFs and 
arterial aneurysms were managed. Elkin and 
Shumacker (1955) outlined the techniques 
of operative treatment of 585 AVFs (Table 
24-15). Arterial repair was used in only 34 
lesions. 



The representative material that follows 
covers a small portion of the World War II 
experience. Freeman and Shumacker (1955) 
outlined various approaches in the manage- 
ment of AVFs. Figure 24-32 shows one of the 
approaches, which involved the following: 



r\ 



r 



5pltt ii»rnum' 



Innominal^ v 




ouoclavian vaii-i 



■ FIGURE 24-32 

Transvenous repair of an arteriovenous fistula involving the left subclavian artery and innominate 
vein. Insert shows the surgical approach to the lesion that was used to manage a World War II 
combat casualty at DeWitt General Hospital in 1945. (From Freeman NE, Shumacker HB Jr, 
DeBakey ME: Vascular surgery. Washington, DC: US Government Printing Office, 1955.) ■ 



ch24.qxd 4/16/04 3:26 PM Page 490 



490 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



TABLE 24-15 

TECHNIQUES OF OPERATIVE TREATMENT IN 585 ARTERIOVENOUS FISTULAS: 
WORLD WAR II EXPERIENCE 







Quadruple 


Ligation Alone (Mass 






Arterial 


Ligation and 


Proximal, Distal, or 


Total 


Location 


Repair 


Excision 


Proximal and 


Distal) 


Cases 


Upper extremity 












Axillary 


— 


32 


— 




32 


Brachial 


— 


29 


— 




29* 


Cervical, transverse 


— 


1 


— 




1* 


Humeral, posterior 


— 


1 


— 




1 


circumflex 












Interosseous, 


— 


1 


— 




1 


common 












Radial 


— 


2 


— 




2 


Scapular, transverse 


— 


2 


— 




2 


Subclavian 


1 


16 


1 




18* 


Ulnar 


— 


9 


— 




9 


Lower extremity 


— 


— 


— 




— 


Calf, to muscles of 


— 


4 


— 




4 


Circumflex, lateral 


— 


1 


— 




1 


Femoral 


16 


124 


1 




141 s 


Geniculate 


— 


5 


— 




5 


Gluteal, inferior 


— 


1 


— 




1 


Gluteal, superior 


— 


3 


— 




3 


Peroneal 


— 


24 


1 




25 


Plantar 


— 


6 


— 




6 


Popliteal 


11 


91 


— 




102 


Profunda femoris 


— 


19 


— 




19 


Profunda branch 


— 


2 


— 




2 


Tibial 


— 


87 


— 




87 


Head and neck 












Carotid 


5 


29 


14 




48* 


Cirsoid 


— 


9 


— 




9 


Lingual 


— 


1 


— 




1 


Occipital 


— 


1 


— 




1 


Temporal, 


— 


3 


2 




5 


superficial 












Vertebral 


— 


8 


5 




13 


Trunk 












Aorta-vena cava 


1 


— 


— 




1 


Hypogastric 


— 


1 


— 




1 


Iliac 


— 


9 


— 




9 


Innominate 


— 


— 


1 




1 


Mammary, internal 


— 


1 


— 




1 


Obturator-iliac vein 


— 


1 


— 




1 


Subscapular 


— 


2 


— 




2 


Thoracoacromial 


— 


1 


— 




1 


Total 


34 


526 


25 




585 



*This total does not include two fistulas: one in which the method of management was not stated and one in which spontaneous 

cure occurred. 

*This total does not include one fistula in which the method of management was not stated. 

*This total does not include one fistula in which spontaneous cure occurred. 

5 This total does not include three fistulas: one in which methods of management were not stated and two in which spontaneous 

cure occurred. 

From Elkin DC, Shumacker HB Jr: In: Vascular Surgery in World War II. Elkin DC, DeBakey ME, eds. Washington, DC: 

Government Printing Office, 1955. 



ch24.qxd 4/16/04 3:26 PH Page 491 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



491 



1. Mass ligation of the fistula 

2. Quadruple ligation and division of the 
main vessels with excision of the fistula 

3. Transvenous closure of the arterial opening 

4. Repair of the opening in both the artery 
and vein 



Shumacker (1948) stressed the importance 
of maintaining arterial continuity in the 
repair of aneurysms and AVFs. In his early 
experience, he performed only four repara- 
tive procedures, with 2.9% of 138 cases in- 
volving the innominate, common carotid, 
extracranial internal carotid, subclavian, axil- 



lary, brachial, iliac, common femoral, femoral, 
and popliteal arteries. In later experience, he 
repaired 52.6% of the arteries: 30 of 57 cases. 
This included lateral arteriorrhaphy, end-to- 
end anastomosis, and vein graft repair (Table 
24-16) . The types of autogenous interposition 
venous grafts used range from the saphenous 
to a branch of the femoral. Figures 24-33 and 
24-34 reveal patency of the venous grafts and 
no dilatation of the grafts in the early follow- 
up period of 7 to 10 weeks. 

Shumacker (1948) also used oscillometry 
to evaluate the patency of arterial repair (Table 
24-17). The results of oscillometry were 
good in those cases in which arterial repairs 




■ FIGURE 24-33 

This arteriogram taken 10 weeks after repair of 
a fistula between the femoral and profunda 
femoral arteries and the femoral vein, with 
resection and end-to-end anastomosis of the 
profunda femoral artery proximally to the 
superficial femoral artery distally, shows no 
narrowing at the suture line after 70% Diodrast 
was injected into the common femoral artery. 
(From Shumacker HB Jr: Problems of 
maintaining continuity of artery in surgery of 
aneurysms and arteriovenous fistulae; notes on 
development and clinical application of 
methods of arterial suture. Ann Surg 
1948;127:207-230.) ■ 



ch24.qxd 4/16/04 3:26 PH Page 492 



492 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



O 



Q. 
CO 

z 
< 

cr 



LU 

> 
> 

CD 

Q 
LU 

cr 
o 

H 
CO 
LU 

cr 

CO 

< 

> 
cr 

LU 

H 

cr 
< 

LL 

o 

> 



o 
o 

X 

o 

•i 

■<* 

CM 



CO 
LU 



oa w 
<! < 
H O 









3 




(0 




0) 




tc 


i- a. 




O 3 


aT 


§1 


4-< 

c 
o 


££ 


S 


«#— 


i-" 


o 


"S 


0) 

o 


C5 


k. 




3 


c 


o 


u 


<fl 


> 


c 







(- ^ > 


> 


E 


H- 


o 


o 




£ 


*. 


4-1 


co 


O) 


ki 


c 


C5 


<u 




_l 






>. 


<u 


E 


> 


o 


I 


5 


k. 


CD 


<u 


.c 


Q. 

o 




4-< 

Q. 


k. 
0. 


E 




(/> 


H- 




o 




c 


c 


o 


o 

"<0 


(B 


0) 


O 


_l 


o 




_l 






c 




o 




m 




0) 




_l 




»*- 




o 




0) 




Q. 




£■ 


c 




o 




■<o 




<u 


^^ 


_l 


CO 


«l— 


£ 


o 


4-1 

c 


c 


o 


o 


2 


*3 




re 




k. 




3 




D 




*- 


4-< 


o 


c 


0) 


o 


O) 


re 


< 


Q. 


01 




<A 




(0 


d 


O 


z 



o 



co •>*■ i- ^- 



r 


n 






a 
c 

CO 


E 

CD 


JZ 

a. 

m 


CO 

F 


LU 




aj 


C/J 



CO p 

en c 

CD 



O o 
O .5= 
D) O 





"a 






TJ 




o 


~a 


"a 


E 




T5 


E 




Q. 


E 










CO 






"co 


















u 
E 


o 
E 


Lc 


U 

E 


Q. J= 


O " 

F -f= 


Si 


j_ 




LL 




LL 


CL 




CD 

LL 


CD 





>- 




,_ 


>, 


i_ 


>i 


, 











































en 




a) 




















-> 


CO ^ 


■> 


rrt 


CC 


m 


CC 


<fl 


<L 


< 


< 


U) 




aj 




C/J 



c 9 "* 

CD w CT) 

II B 

"J E c 

< LL < 



ch24.qxd 4/16/04 3:26 PH Page 493 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



493 



< 

D. 
LU 

<r 

_l 

< 

cc 

LU 

h- 

cc 
< 

cc 

LU 



< 

CO 
LU 

Q 

Z> 
H 
CO 

o 

cc 

u _j 
M ° 
H O 







14_ 








£ 




o 

<D 


_c 


CO 




Ul 




CD 

eg 


CD 
C 


CO 


-Q 






c 


^ 


CO 


E 




a> 


03 


^ 


_j 


*— 




o 





c 




re 


E 
< 


55 


CC 


o 

O 






Q. 






<i) 


*_ 










E 


o 










o 
























~ 








a> 





o 








c 


CD 


<fl 
O 








T3 
CO 
CD 

CC 


CO 

ID 
> 
< 



(0 

o 

* I 

£» 
« o 

E LL 

O i_ 

= o 
o 

<A 

o 



c 

< 

** -■_■ 
& I 

t5 * 
o 

<A 
O 



(0 

a) 
m 

as 
O 



re 

Q. 

a> 
cc 

£■ 
a> 

r 
< 



re 

Q. 

a> 
cc 

•^ 
o 

0) 

a. 
,>. 



o ,- CO 

C 13 CO .E 

CO 1= — I 

O C 



CO CD 

£ > 
cc < 



o ,- CO 

au - cB 

a e s-i 

C 13 CO .E 

CO ~ —I 

O C 

55 cc o 



o h- 
o CD 



CD oo 

CD i- 



o un 



in 


o t- t- 


O O 


m 


en 


en 


O) 


r^ ^t- lo 


in in 


■* 


CO 


t- 



- CD 


co in co 


in 




CO 


ro 


5 h- 


co t- c\j 


■* -*■ 


CO 


■* 


o 



ro 


in 


CO r~- CO 


o r- 


N- 


m 


n- 


0) 


r-~ 


CO CO CO 


CD CD 


UJ 


r^- 


i— 



CD CD 
C CD 

-a 2 


CO 


_ 


lOO ID 


O T- 


CO 
<D > 

cc < 


CM 


in 


CNJ t- C\J 


CO CO 



CD O 
C\j -<- 



m *$■ cm cm ^t -i- 'vf t- ^r lo 



co 


CO 




> 

ra 


>i 


CO 


ca 


CO 


CO 

(11 


u 
F 


"5. 





JO 


o 


u 

F 


U 

F 


~Cl 


ai 


1 1 


1 






id 


en 


1 1 


j_ 


D_ 


CO 


< 


ill 


LL 


LL 


LL 



— .t= a. 



n 2 " 
D- CM 



JZ. 


m 










- 




J> 










i 




i/i 


r 





ch24.qxd 4/16/04 3:26 PM Page 494 



494 



V • SPECIAL PROBLEMS AND COMPLICATIONS 




■ FIGURE 24-34 

Left, Arteriogram, taken 7 weeks after repair of 
an arteriovenous fistula between the superficial 
femoral artery and vein, with interposition of a 
segment of a large branch of the femoral vein 
2cm in length, reveals that the venous insert 
and the artery have relative diameters about 
equal to those observed at completion of the 
operation (case 2). Right, Arteriogram showing 
no dilatation of the venous segment of a 2-cm 
piece of saphenous vein used to reconstruct 
the superficial femoral vessels (case 1). There 
was no dilatation at the completion of the 
anastomosis, and no dilatation was seen on 
this arteriogram performed 10 weeks later. 
(From Shumacker HB Jr: Problems of 
maintaining continuity of artery in surgery of 
aneurysms and arteriovenous fistulae; notes 
on development and clinical application of 
methods of arterial suture. Ann Surg 
1948;127:207-230.) ■ 



remained patent and poor in those in which 
arterial repair failed due to thrombosis. 

Hughes and Jahnke (1958) performed 
end-to-end anastomosis in the majority of AVFs 
(61/134) from the Korean Conflict (Table 
24-18). As a result of the Korean Conflict, 
more than 200 patients with false aneurysms 
and AVFs, 133 of the injuries involving major 
vessels, were seen atWalter Reed General Hos- 
pital. The lesions were excised, with repara- 
tive or reconstructive surgery of the major 
vessel, without loss of a single limb. Treatment 



of minor vessel lesions has previously been 
outlined in Table 24-8. Repair of major veins 
was performed whenever possible to prevent 
venous insufficiency. This venous repair was 
possible in about 30% of major veins involved 
in fistula formation. Cardiac dilatation was 
common with large fistulas; however, only two 
patients showed cardiac failure. 

Rich, Hobson, and Collins (1975) reported 
the experience from Vietnam. Of 558 lesions 
identified in 509 patients, there was almost 
an equal number of AVFs (262 AVFs) and false 
aneurysms (296 false aneurysms). As might 
be anticipated by the number of American 
troops committed to Southeast Asia in that 
year, the largest number of lesions resulted 
from wounds in 1968 (Table 24-19). There 
was also a relatively large number of similar 
wounds in 1967 and 1969. The time from 
injury to recognition of the lesion was arbi- 
trarily divided into four categories: immedi- 
ate, early, delayed, and remote. The largest 
number of lesions was recognized in the early 
period of 1 to 30 days: 273, or 48.9% (Table 
24-20). Nearly an equal number was diag- 
nosed in the delayed period between 1 and 6 
months. In the remote group, all but 7 of the 
35 patients had recognition and treatment of 
their lesions in less than 2 years. Only two had 
recognition and treatment of their lesions 
after more than 5 years following the initial 
injury, and both were treated in less than 6 
years. Nearly an equal number of lesions were 
treated in the intermediate hospitals in Japan 
and similar Far West locations as were treated 
in the continental United States (Table 
24-21). Several hundred surgeons were 
involved in these repairs. Approximately one 
fifth of these operations were performed at 
Walter Reed Army Medical Center. 

Table 24-22 outlines the method of treat- 
ment used for the various arterial and venous 
injuries. Arterial ligation was used in 290 
lesions, or 52.0%. Compelling problems often 
caused this method to be used over the favored 
and desired arterial repair. Infection, associ- 
ated injuries, poor general condition of the 
patient, and involvement of smaller caliber 
arteries were considered. The overall mortality 
rate for the 509 patients was 1.8%, or 7 deaths 
(Table 24-23) . Even considering this low mor- 
tality rate, only two deaths could be directly 



ch24.qxd 4/16/04 3:26 PH Page 495 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



495 



TABLE 24-18 

TOTAL OPERATIONS FOR MAJOR VESSEL LESIONS: MILITARY SERIES FROM 
KOREAN CONFLICT 





Ligation 


















and 




Vein 


Artery 


Lateral 


Division of 


Spontaneous 




Vessel 


Excision 


Anastomosis 


Graft 


Graft 


Repair 


Fistula 


Closure 


Total 

HHHH 


Common 


_ 


6 








_ 


1 


2 




carotid 


















nternal 


2 


— 


— 


— 


— 


— 


— 


3 


carotid 


















Subclavian 


3 


2 


— 




1 


1 


— 


8 


Axillary 


4 


8 


2 




2 


— 


3 


20 


Brachial 


6 


9 


1 




1 


1 


— 


19 


Iliac 


— 


3 


— 




— 


— 


— 


4 


Common 


— 


3 


2 




— 


2 


— 


8 


femoral 


















Superficial 


6 


14 


9 


— 


— 


2 


— 


31 


femoral 


















Popliteal 


_9 


16 


_3 


1 


— 


_2 


1 


32 


Total 


30 


61 


17 


6 


4 


10 


6 


134 



From Hughes CW, Jahnke EJ Jr: The surgery of traumatic arteriovenous fistulas and aneurysms: A five-year followup study of 
215 lesions. Ann Surg 1958;148:790-797. 



TABLE 24-19 

ARTERIOVENOUS FISTULAS AND 
FALSE ANEURYSMS BY YEAR: 
VIETNAM VASCULAR REGISTRY 



Year 

1963 
1964 
1965 
1966 
1967 
1968 
1969 
1970 
1971 
1972 
Total 



No. 

1 



11 

44 

116 

249 

124 

5 

7 

1 

558 



0.2 

0.0 

2.0 

7.9 

20.8 

44.6 

22.2 

0.9 

1.2 

0.2 

100.0 



From Rich NM, Hobson RW II, Collins GJ Jr: Traumatic 
arteriovenous fistulas and false aneurysms: A review of 
558 lesions. Surgery 1975;78:817-828. 



attributed to the vascular problem. The mor- 
bidity rate of 6.8% included 35 complications: 
hemorrhage in 14, thrombosis in 12, stenosis 
in 2, and persistent, immediately adjacent, or 
recurrent AVFs requiring additional opera- 
tions in 7. 



TABLE 24-20 

ARTERIOVENOUS FISTULAS AND 
FALSE ANEURYSMS, BY TIME OF 
DIAGNOSIS: VIETNAM VASCULAR 
REGISTRY 



Time 

Immediate (24hr) 
Early (1-30 days) 
Delayed (1-6 mo) 
Remote (>6mo) 
Total 



No. 

22 
273 
228 

35 

558 



3.9 
48.9 
40.9 

6.3 
100.0 



From Rich NM, Hobson RW II, Collins GJ Jr: Traumatic 
arteriovenous fistulas and false aneurysms: A review of 
558 lesions. Surgery 1975;78:817-828. 



Experience in the civilian hospitals is 
increasing. Hershey (1961) encountered a 
technical complication. The artery proximal 
to the AVF had dilated and become fragile; it 
was crushed by clamp, and a hematoma devel- 
oped (Fig. 24-35). 

Beall (1968) repaired 8 of 50 AVFs within 
24 hours of injury; an additional 17 were 
repaired within 24 hours to 3 months follow- 
ing injury. However, there was a delayed repair 



ch24.qxd 4/16/04 3:26 PH Page 496 



496 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



TABLE 24-21 

ARTERIOVENOUS FISTULAS AND 
FALSE ANEURYSMS, BY HOSPITAL 
LOCATION FOR REPAIR: VIETNAM 
VASCULAR REGISTRY 



Location 


No. 


of Repairs 


% 


Vietnam 




57 


10.2 


Japan, etc. 




238 


42.7 


CONUS 




251 


45.0 


No repair 




12 


2.1 


Total 




558 


100.0 



From Rich NM, Hobson RW II, Collins GJ Jr: Traumatic 
arteriovenous fistulas and false aneurysms: a review of 
558 lesions. Surgery 1975;78:817-828. 



TABLE 24-23 

ARTERIOVENOUS FISTULAS AND 
ANEURYSMS, MORTALITY AND 
MORBIDITY RATES: VIETNAM 
VASCULAR REGISTRY 



No. 



Deaths 

Morbidity 
Amputations 
Complications 



35 



1.8 

1.7 

6.3 



From Rich NM, Hobson RW II, Collins GJ Jr: Traumatic 
arteriovenous fistulas and false aneurysms: a review of 
558 lesions. Surgery 1975;78:817-828. 



TABLE 24-22 

ARTERIOVENOUS FISTULAS AND 
FALSE ANEURYSMS, BY METHOD OF 
MANAGEMENT: VIETNAM VASCULAR 
REGISTRY 



Type 


No. 


% 


Arterial 






Ligation 


290 


52.0 


End-to-end anastomosis 


143 


25.6 


Vein graft 


57 


10.2 


Lateral suture 


40 


7.2 


Prosthesis 


2 


0.3 


Miscellaneous 


26 


4.7 


Total 


558 


100.0 


Venous 






Ligation 


138 


52.7 


Suture 


79 


30.1 


Miscellaneous 


45 


17.2 


Total 


262 


100.0 



From Rich NM, Hobson RW II, Collins GJ Jr: Traumatic 
arteriovenous fistulas and false aneurysms: a review of 
558 lesions. Surgery 1975;78:817-828. 



of more than 3 months following injury for 
23, or nearly 50%, of these lesions. No repair 
was performed for two of the AVFs. Excision 
and repair was used for 27 lesions and liga- 
tion and excision for 17 lesions. No deaths 
were reported. There were no amputations 
required. Not counting two patients lost in 
follow-up who had no treatment, 42 were 



asymptomatic. Six patients were sympto- 
matic after their original definitive surgical 
procedure, and three required subsequent 
operations. 

In the civilian series of 61 arterial injuries 
reported by Smith, Foran, and Caspar (1963), 
approximately two thirds of the 33 chronic or 
late lesions were AVFs. They mentioned 
that the time interval from original injury to 
treatment varied considerably from a few days 
to 29 years, with most patients, 57%, being 
treated after 1 year. Of the six patients with 
AVFs reported by Patman, Poulos, and Shires 
(1964), five did not have initial explorations 
of the area. The remaining patient did have 
initial exploration; however, the AVFs were 
not diagnosed until 4 hours after injury. The 
authors stressed that this development demon- 
strated the rapidity with which an AVF can 
develop. The common and superficial femoral 
arteries were involved in AVFs. The remain- 
ing four fistulas were equally divided among 
the smaller radial and posterior tibial arter- 
ies. There were no deaths, amputations, or 
other significant complications in any of the 
patients. 

Dillard, Nelson, and Norman (1968) 
reported a number of AVFs including (1) a 
29-year-old female who was stabbed in the right 
flank and 3 years later was found to have severe 
hypertension; after correction of the renal 
AVF, the patient's blood pressure returned to 
normal; (2) a patient with severe leg ulcers 
that healed only after correction of an AVF in 



ch24.qxd 4/16/04 3:26 PH Page 497 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



497 




3cm 




■ FIGURE 24-35 

A, Artist's sketch of a superficial femoral arteriovenous fistula. B, End-to-end anastomosis after 
excision of the fistula. An intraluminal hematoma developed at the site of a Blalock clamp. C, Sketch 
of the vein graft after excision of the hematoma, showing size discrepancies. D, Postoperative 
arteriogram showing (arrows,) the site of anastomosis. (From Hershey FB: Secondary repair of 
arterial injuries. Am Surg 1961 ;27:33-41 .) ■ 



the same extremity between the common 
femoral artery and vein; and (3) a patient with 
an AVF between the popliteal artery and vein, 
which resulted in amputation. Two of the 
nine AVFs reported by Dillard, Nelson, and 
Norman (1968) involved high-output failure. 
One of these fistulas occurred between the 
subcapsular artery and the axillary vein, and 
the other between the right iliac artery and 
the left common iliac vein. 

Sako and Varco (1970) reported corrective 
procedures in 25 patients with acquired AVFs. 
Excisions of the fistula with arterial and 
venous repair were performed in more than 
50%, or 16 lesions. Quadruple ligation was 
used in six and multiple ligation in two, and 
included in the arterial repairs were 13 
primary anastomoses, 3 autologous venous 
grafts, and 1 homograft. All of the acquired 
fistulas were cured by the surgical procedures 
described without a death. 

Gaspard and Caspar (1972) reported two 
patients who developed AVF after Fogarty 
catheter thrombectomy in the lower extrem- 
ity. They emphasized that neither of their 
patients required immediate operation for 
limb salvage or had an operation performed 
subsequently. They cited the report by Rob 



and Battle (1971) in which correction of the 
AVF 26 days after the use of the Fogarty 
catheter was mandatory because the distal 
extremity was in jeopardy. 

Hewitt, Smith, and Drapanas (1973) advo- 
cated immediate repair of acute AVFs. This 
was possible in 13 of the 14 patients in their 
series, and they reported satisfactory results 
in all repairs, including resection with end- 
to-end anastomosis in 6, saphenous vein graft 
in 2, saphenous vein patch graft in 2, and 
lateral suture repair in 3, with ligation being 
required only for one distal internal carotid 
artery. 

In addition to the anticipated complications 
of cardiac enlargement, cardiac failure, endo- 
carditis, and proximal arterial aneurysm 
formation, unusual complications have been 
reported. Rhodes, Cox, and Silver (1973) 
reported a case of a 53-year-old male with a 
10-day history of bruising easily, hematoma, 
and bleeding from his tongue. The patient 
was involved in a shooting accident 17 years 
previously and had acquired an AVF between 
the left subclavian artery and vein as a result. 
The authors attributed the local sustained 
intravascular coagulation that caused a man's 
symptoms to turbulence from the fistula and 



ch24.qxd 4/16/04 3:26 PH Page 498 



498 



V • SPECIAL PROBLEMS AND COMPLICATIONS 




■ FIGURE 24-36 

A, Arteriography after excision and anastomosis of the superficial femoral artery shows an excellent 
lumen. A vein graft inserted into the popliteal artery is demonstrated by angiography approximately 
6 months after operation. B, Examination of this patient 5.5 years after operation showed the vein 
graft to be functioning perfectly without clinical evidence of dilatation. C, Arteriography was used to 
demonstrated an arterial homograft that replaced the common femoral artery. These angiograms 
were part of the follow-up of Korean casualties who had repair of arteriovenous fistulas. (From 
Hughes CW, Jahnke EJ Jr: The surgery of traumatic arteriovenous fistulas and aneurysms: A five- 
year follow up study of 215 lesions. Ann Surg 1958;148:790-797.) ■ 



stasis from the aneurysm. The coagulopathy 
and bleeding responded to surgical elimina- 
tion of the fistula and aneurysm. The authors 
felt that this was the first report of a con- 
sumption coagulopathy resulting from an AVF 
and false aneurysm. 



Follow-up 

Hughes and Jahnke (1958) included a 5-year 
follow-up of 148 lesions treated during the 
Korean Conflict, with satisfactory results 
being obtained in most of the patients 
(Fig. 24-36). 

The Vietnam Vascular Registry continues 
to follow patients included in the report by 
Rich (1975). More than one fourth — 149 
patients or 29.3% — have been evaluated in the 
vascular clinic at Walter Reed Medical Center. 
Many of these patients can be expected to live 
50 years or more (Fig. 24-37). 



SakoandVarco (1970) reported long-term 
follow-up of 14 of 25 patients with acquired 
AVFs who were cured of their lesions for 5 to 
16 years. Seven additional patients were fol- 
lowed for more than 2 years and were all cured. 
One patient in this group had a portion of 
the anterior tibial artery repaired after exci- 
sion of the fistula, but the artery was occluded 
within the first year. Two were lost to follow- 
up after the first year, and one had quadru- 
ple ligation of the subclavian AVF. When last 
seen, he had symptoms indicating some 
ischemia of the arm. The other patient lost 
to follow-up had quadruple ligation of the 
gluteal AVF. The remaining two patients 
who had an aneurysmal dilatation in the prox- 
imal artery excised and replaced with pros- 
thetic graft were well 8 and 1 1 years after the 
operation (Fig. 24-38) . The other two patients 
with aneurysmal dilatation of the artery 
proximal to the fistula had not yet had these 
corrected. 



ch24.qxd 4/16/04 3:26 PH Page 499 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



499 




■ FIGURE 24-37 

This angiogram corroborated the clinical 
impression of a left femoral arteriovenous fistula. 
Additional assistance, however, was provided to 
establish that the communication involved a 
muscular branch of the superficial femoral artery 
and the superficial femoral vein. Note the 
development collaterals. Also note the proximal 
arterial dilatation of the superficial femoral artery 
in this former soldier who had been wounded 5 
years before this study. (From Rich NM: In: 
Beebe HG, ed. Complications in vascular 
surgery. Philadelphia: JB Lippincott, 1973.) ■ 




■ FIGURE 24-38 

In this operative photograph, aneurysmal dilatation of the superficial femoral artery (A), the 
narrowed segment (B) where the artery traversed Hunter's canal, and a popliteal aneurysm (C) are 
demonstrated. This patient had closure of an arteriovenous fistula of 21 years' duration, which 
involved the anterior tibial vessels. Fourteen years after the fistula closure, multiple aneurysms of 
femoropopliteal arteries developed. (From Sako Y, Varco RL: Arteriovenous fistula: Results of 
management of congenital and acquired forms, blood flow measurements, and observations on 
proximal arterial degeneration. Surgery 1970;67:40-61.) ■ 



ch24.qxd 4/16/04 3:26 PM Page 500 



500 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



TRAUMATIC FALSE 
ANEURYSMS 



He was bled at his own desire by a bleeder 
who had performed the same operation for 
him, and generally in the same arm, some 
30 or 40 times. Bleeding from an orifice was 
done by firm compression, and on the day 
following finding the bandage tight, he 
removed it, and found the orifice to be 
completely closed. A short time after this, a 
small pulsating swelling was observed by 
him at this point which slowly increased till a 
day or two previous to my seeing him when 
after some exertion with his arm he 
observed a very considerable augmentation 
of its size. 
Norris, 1843 



History 

Since antiquity, the management of false 
aneurysms has been closely allied to vascular 
surgery. It has been repeatedly recorded 
that Antyllus in the second century treated 
an arterial aneurysm by ligature above and 
below the lesion, with incision of the an- 
eurysm and extraction of the clot. Schwartz 
(1958) reported that Antyllus treated small 
peripheral traumatic aneurysms by ligating 
both ends and puncturing the center; however, 
he advised against this practice in large 
aneurysms. Figure 24-39 shows some of the 
early methods of treatment of aneurysms. 
Hunter electively ligated the femoral artery 
proximal to a popliteal aneurysm in 1786 to 
reduce blood loss during subsequent attempts 
at excision of the aneurysm. Pick (1873) pro- 
vided an interesting and detailed account of 
his management of a large femoral false 
aneurysm by digital compression, which had 



Arxtyllua 



Ar\al 







■ FIGURE 24-39 

Various types of operations employed for the treatment of aneurysms before the introduction of 
Matas' endoaneurysmorrhaphy in 1888. (From Elkin DC: Traumatic aneurysms; Matas operation- 
57 years after. Surg Gynecol Obstet 1946;82:1-12.) ■ 



ch24.qxd 4/16/04 3:26 PH Page 501 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



501 




■ FIGURE 24-40 

A, Matas used a contemporary compressor applied 
to the femoral artery at Hunter's canal to test the 
collateral circulation in lesion such as this popliteal 
aneurysm. B, In World War II, Elkin found the Matas 
compressor to be an inexpensive and easily 
constructed instrument that could compress various 
arteries to determine the development of collateral 
circulation. (A, From Matas R: Keene's surgery, vol. 7. 
Philadelphia: WB Saunders, 1921; B, from Elkin DC: 
Vascular injuries of warfare. Ann Surg 1944;1 20:284- 
310.) ■ 



disastrous final results. This digital compres- 
sion directly over the pulsating mass was 
applied fairly continuously initially and then 
for a considerable period of the waking hours 
until 4 days later when the area became so 
tender that the compression had to be dis- 
continued. Not only did this initiate throm- 
bus formation in the false aneurysm, but it 
also became evident in less than 1 week that 
the distal pulses could not be felt over either 
the anterior or the posterior tibial artery. 
Gangrene developed approximately 3 weeks 
after the initiation of the digital compression, 
an amputation was performed at the hip 
level. The patient had a stormy postoperative 
course for approximately 3 hours before he 
died. 

Matas (1888) described an endoaneurys- 
morrhaphy operation, a method of intrasac- 
cular suture, for the treatment of a brachial 
arterial aneurysm. Within a few years, Matas 
(1903) also recommended restoration of cir- 
culation through the damaged artery as the 
ideal treatment for arterial aneurysms. He 
developed a compressor (Fig. 24-40) to test 



the development of collateral circulation 
before performing his endoaneurysmor- 
rhaphy. His approach to widely open the 
aneurysm and to suture the communications 
into the artery (Fig. 24-41) was the standard 
treatment, with minimal modification, for 
more than 50 years, a period that included 
World War II. 

Despite the acceptance of the Matas endo- 
aneurysmorrhaphy during World War I and 
World War II, interest in preserving arterial 
continuity was maintained. Lexer (1907) was 
the first to use a segment of saphenous vein 
as an interposition graft in an arterial defect 
caused by excision of a traumatic axillary 
aneurysm. Some of the problems associated 
with arterial repair have been detailed in 
Chapter 1 . Individual series of successful arte- 
rial repairs have been reported. Soubbotitch 
(1913) used suture repair, as has previously 
been described in Chapter 1. Elkin (1946) 
emphasized that all of the previous approaches 
outlined by Antyllus, Anel, Hunter, and 
Brasdor and Wardrop were frequently fol- 
lowed by infection, hemorrhage, gangrene, 



ch24.qxd 4/16/04 3:26 PH Page 502 



502 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



or failure to cure the false aneurysm. Only 
the Matas procedure avoided these compli- 
cations during the World War II experience. 
The following methods of managing arterial 
aneurysms were outlined by Freeman and 
Shum acker (1955): 

1. Endoaneurysmorrhaphy of Matas 

2. Measures designed to produce clot in the 
aneurysmal sac or to induce formation of 
fibrous tissue about it to prevent further 
expansion and possible rupture 

3. Obliteration of the sac by closure of the 
offending vessel 

4. Extirpation of the aneurysm-bearing is 
portion of the artery 

5. Extirpation of the lesion, combined with 
some procedure to permit maintenance or 
to reestablish continuity of the affected 
artery. 

The extensive World War II experience is 
documented in detail by Elkin, by Shumacker, 
and by DeBakey and Elkin (1955). 

Since the Korean Conflict in which arter- 
ial repair was emphasized, vascular recon- 



struction has become the procedure of choice 
in restoring arterial continuity in the repair 
of false aneurysms, in both the military and 
the civilian situation. Hughes and Jahnke 
(1958) reviewed the Korean experience and 
provided a 5-year follow-up. A similar exten- 
sive review has been completed recently for 
the Vietnam experience (Rich, 1975). 

As might be anticipated, a smaller number 
of false aneurysms have been documented in 
civilian experience than in recent military 
experience. Patman, Poulos, and Shires 
(1964) reported 12 patients who developed 
false aneurysms in their series of 256 patients 
with civilian arterial injuries in Dallas, an inci- 
dence of 4.7%. None of these patients had 
an initial exploration. Among the major 
vessels that developed false aneurysms were 
the aorta (1), subclavian (1), axillary (2), 
superficial femoral (1), and popliteal (1). 
There were also three radial artery false 
aneurysms and single false aneurysms of the 
profunda femoris, anterior tibial, and poste- 
rior tibial arteries. The ratio of false aneurysms 
to AVFs was 2:1 in their series, which was the 
opposite of the ratio reported by Hughes and 
Jahnke (1958) from the Korean experience. 



■ FIGURE 24-41 

This diagram illustrates the obliterative 
endoaneurysmorrhaphy of Matas. Although 
Matas also believed in the reconstructive 
endoaneurysmorrhaphy, he elected to use the 
obliterative technique in this case in 1917 
involving a gunshot wound of the superficial 
femoral artery because collateral circulation had 
been established and the obliterative suture 
could be applied with safety to the limb. A 
continuous intrasaccular silk suture obliterated 
the orifices of the communication with the main 
artery, both proximally and distally. (From Matas 
R: Keene's surgery, vol. 7. Philadelphia: WB 
Saunders, 1921.) ■ 




ch24.qxd 4/16/04 3:26 PM Page 503 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



503 



Incidence 

With the increased interest in primary arter- 
ial repair of injured arteries during the past 
25 years, many anticipated that there would 
be a resultant decrease in false aneurysms. This 
was particularly true in Vietnam (Rich 1975) . 
However, considering the various etiologic 
factors, remaining diagnostic problems, and 
priorities of managing a patient with multi- 
ple life-threatening injuries, it should be 
obvious that the treatment of false aneurysms 
remains an important aspect of vascular 
surgery. 

Similar to the varying incidence of arterial 
injuries in the injured patient in general and 
of AVFs, there is considerable disparity in the 
reported incidence of false aneurysms, in both 
civilian and military experience. One expla- 
nation for this is at times false aneurysms are 
included in series of arterial trauma and at 
other times they are not. Also, some series do 
not distinguish AVFs from false aneurysms in 
combined reports. 

Shumacker and Carter (1946) compared 
the incidence of arterial aneurysms and AVFs 
in large caliber peripheral arteries in their 
World War II experience (see Table 24-2) . 
Brachial false aneurysms were more prevalent 
than brachial AVFs, and the converse was true 
with femoral AVFs and false aneurysms. 
Hughes and Jahnke (1958) found that there 
were approximately twice as many AVFs as false 
aneurysms in the Korean experience. When 
major vessel lesions were considered (see 
Table 24-7), they also noted fewer femoral 
false aneurysms than AVFs. Rich (1975) 
reported a somewhat different experience in 
Vietnam, where there were slightly more false 
aneurysms than AVFs (see Table 24-3). 

In the relatively small series of arterial 
injuries reported by Dillard, Nelson, and 
Norman (1968), false aneurysms (nine 
injuries) were more common than AVFs 
(seven injuries) in their civilian experience 
in St. Louis. 



Etiology 

Penetrating injuries are usually responsible for 
a false aneurysm, or traumatic aneurysm, 



which is produced by a tangential laceration 
through all three layers of the wall of an artery. 
In the military experience, fragments from 
various exploding devices and bullets account 
for the penetrating missile wounds (Fig. 
24-42). In Civilian experience, stab wounds, 
in addition to low-velocity bullet wounds, are 
often associated with false aneurysms. 

The increased use of fragmenting missiles 
in combat parallels the relatively high inci- 
dence of the development of these aneurysms 
in a number of wars, particularly before the 
advent of vascular repair. Hughes (1954) 
noted that 85% of the vascular wounds in 
Korea resulted from fragmenting missiles, 
with only 15% being from bullets. Rich 
(1975) found that a similar percentage, about 
87%, of fragment wounds were responsible 
for 558 false aneurysms and AVFs (see Table 
24-12). 

Diagnostic and therapeutic procedures can 
result in false aneurysms if the placement of 













m 1 







■ FIGURE 24-42 

Representative of military wounds associated 
with false aneurysms is this large fragment 
wound of the popliteal fossa with a large false 
aneurysm of the popliteal artery demonstrated 
angiographically. (From Vietnam Vascular 
Registry #2967, NMR, Walter Reed General 
Hospital.) ■ 



ch24.qxd 4/16/04 3:26 PM Page 504 



504 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



needles and catheters injures the arteries. The 
first lesions that were successfully treated by 
Lambert (1759) and Norris (1843) resulted 
from bloodletting. At Walter Reed General 
Hospital, four false aneurysms developed 
following catheterization for angiographic 
procedures (Rich, 1975). Postoperative false 
aneurysms, other than anastomotic false 
aneurysms, have been associated with many 
operations. Smith (1963) cited development 
of a false aneurysm of the common femoral 
artery following an inguinal herniorrhaphy. 
There was sudden profuse arterial bleeding 
in the course of the herniorrhaphy, and 
hemostasis was eventually secured by multi- 
ple silk sutures. Approximately 2 weeks after 
the operation, a pulsatile, firm, and tender 
mass measuring 8 by 6 by 4 cm was palpated 
in the area of the left inguinal ligament. 
Despite tile fact, there was a purulent exudate 
surrounding the area of the 1-cm tear in the 
common femoral artery where a number of 
sutures had been placed in the defect. It was 
elected to excise the traumatized area and 
perform an end-to-end anastomosis. Five days 
after this second procedure, severe hemor- 
rhage occurred and it was necessary to ligate 
the common femoral artery. Nevertheless, via- 
bility of the extremity persisted. 

The fact that a false aneurysm can develop 
following the operative removal of a herni- 
ated nucleus pulposus was documented by 
Seeley (1954). Seeley mentioned treating a 
20-year-old patient with a right common iliac 
artery aneurysm who had been operated on 
the L4-5 intervertebral space 1 month before 
his admission at Walter Reed General Hospi- 
tal. Six weeks following the initial disk oper- 
ation, a second operation was performed and 
an enormous false sac was found surround- 
ing a right common iliac artery defect (Fig. 
24-43). It was necessary to restore arterial 
continuity by inserting a 2-cm homologous 
arterial graft. Subsequent complications asso- 
ciated with disruption of the graft necessitated 
ligation of the right common iliac artery and 
vein. Fortunately, viability of the extremity was 
maintained. 

Fractures can be associated with false 
aneurysm formation. Cameron, Laird, and 
Carroll (1972) presented an interesting review 
of 10 cases of false aneurysms complicating 




■ FIGURE 24-43 

An enormous false aneurysm from a defect in 
the right common iliac artery was operated on 
at Walter Reed General Hospital 6 weeks after 
the initial disk operation. The right ureter was 
displaced laterally by the mass. The segment 
of the artery with the posterior defect was 
excised, and the hypogastric artery ligated. A 
2-cm homologous arterial graft was used to 
bridge the defect. (From Seeley SF, Hughes 
CW, Jahnke EJ Jr: Major vessel damage in 
lumbar disc operation. Surgery 1954;35:421- 
429.) ■ 



closed fractures in a variety of anatomic loca- 
tions (Table 24-24) . Singh and Gorman 
(1972) emphasized that the formation of a 
false aneurysm as a result of a closed trauma 
to the lower extremity was unusual. They pre- 
sented a case of a 51-year-old man who sus- 
tained a closed fracture at the junction of the 
middle and distal thirds of the tibia and fibula 
when his leg was caught by an encircling boat 
cable in a ship-building yard in 1966. Initially, 
a closed reduction of the fracture was 
performed, with immobilization of the limb 
in a long leg cast. This was replaced by a 
walking cast, which was kept on for 6 months 
before it was determined that the fracture was 
healed. At that time, the patient noted super- 
ficial varicosities. The examining physician 
stated that his extremity had the typical post- 
phlebitic syndrome appearance, except that 



ch24.qxd 4/16/04 3:26 PM Page 505 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



505 



TABLE 24-24 

REPORTED CASES OF FALSE ANEURYSMS COMPLICATING CLOSED FRACTURES 



Author 



Robson 



Year 



1957 



Crellin 




1963 


Meyer and Slager 


1964 


Dameron 


1964 


Staheli 




1967 


Smith 




1963 


Stein 




1958 


Bassett 


and Houck 


1964 


Bassett 


and Silver 


1966 


Harrow 




1970 



Artery Involved 



Fourth lumbar artery 

Anterior tibial artery 
Profunda femoral artery 
Profunda femoral artery 
Popliteal artery 
One false aneurysm with closed 

fracture in 61 arterial injuries; 

site not stated 
Anterior tibial artery 
Profunda femoral artery 
Thoracic aorta 
Right internal iliac artery 



Fracture 



Fractured spinous processes and 

traumatic spondylolisthesis 
Fracture upper third tibia 
Subtrochanteric osteotomy 
Screwn blade plate 
Fracture distal femoral shaft 



Fracture of tibial plateau 

Blade plate for subtrochanteric osteotomy 

Eleventh dorsal vertebra 

Pelvis 



Modified from Cameron HS, Laird JJ, Carroll SE: False aneurysms complicating closed fractures. J Trauma 1972;12:67-74. 



there was a pulsatile mass with a bruit located 
over the posteromedial aspect of the distal 
tibia. A femoral arteriogram revealed a large 
false aneurysm and an AVF of the distal part 
of the posterior tibial artery and accompany- 
ing veins (Fig. 24-44). It was possible to 
perform a lateral repair of the posterior tibial 
artery with interruption of the venous com- 
ponent. Six months later, the patient was 
asymptomatic with no extremity edema. 

Blunt trauma without an associated fracture 
can also result in a false aneurysm. Lai, 
Hoffman, and Adamkiewicz (1966) presented 
an unusual case of dissecting aneurysm of 
the cervical carotid artery in a 21-year-old 
male following a hyperextension neck injury 
sustained in an automobile accident. The 
patient presented at the Johns Hopkins Hos- 
pital with a chief complaint of pain of the left 
side of his head and neck 6 months after the 
car he was driving collided with a truck. A firm, 
tender, 4-by-4cm mass high in the left cervi- 
cal area was obvious, and a bruit was heard 
over the mass. A left carotid angiogram 
revealed considerable lateral displacement 
of the internal carotid artery, and the mass 
promptly filled with contrast media. A 4-by-6 
cm dissecting aneurysm of the internal carotid 
artery was found at the time of exploration, 
with the hypoglossal nerve, the vagus nerve, 
and the spinal accessory nerve all being 
displaced by the aneurysmal sac. Because 



the superior portion of the aneurysmal sac 
approached the base of the skull, it was 
necessary to ligate the internal and external 
carotid arteries. The patient had an unevent- 
ful postoperative recovery with no abnormal 
neurologic findings other than the cranial 
nerve deficits present before surgery. 



Clinical Pathology 

A false aneurysm, or traumatic aneurysm, is 
caused by trauma that lacerates or ruptures 
all three layers of the wall of an artery. Arter- 
ial flow through the artery is usually main- 
tained, and the extravasated blood through 
the laceration is contained by surrounding 
tissues to become a pulsating hematoma and 
subsequently an encapsulated false aneurysm. 
The hematoma that is formed compresses and 
seals the point of injury. Within days to weeks 
later, the thrombus gradually liquefies. False 
aneurysms are distinguished from true 
aneurysms. Whether the true aneurysm is 
congenital in origin, arteriosclerotic, mycotic, 
syphilitic, or caused by unusual systemic dis- 
eases such as polyarteritis nodosa, the true 
aneurysm has a sac composed of one or more 
layers of the artery rather than a rupture 
through all of the walls of the artery, as occurs 
in the traumatic false aneurysm. Indirect 
or blunt trauma can actually cause a true 



ch24.qxd 4/16/04 3:26 PM Page 506 



506 



V • SPECIAL PROBLEMS AND COMPLICATIONS 




■ FIGURE 24-44 

A, A large false aneurysm of the posterior tibial artery and a posterior tibial arteriovenous fistula 
were demonstrated angiographically in a 51 -year-old man who sustained a closed fracture at the 
junction of the middle and distal thirds of the tibia and fibula. B, The large posterior tibial false 
aneurysm was demonstrated at the time of surgical exploration. (From Singh I, Gorman JF: 
Vascular injuries in closed fractures near junction of middle and lower thirds of the tibia. J Trauma 
1972;12:592-598.) ■ 



aneurysm. True traumatic aneurysms caused 
by blunt, nonpenetrating trauma form a small 
group compared to traumatic false aneurysms. 
Blunt trauma causes a confusion of the arte- 
rial wall, with the damaged arterial segment 
progressively dilating and forming a true 



aneurysm. Early recognition and treatment 
are rarely possible because the injury will 
usually not be apparent until a true aneurysm 
develops to a significant size. Only pathologic 
evaluation may differentiate a traumatic true 
aneurysm from a traumatic false aneurysm. 



ch24.qxd 4/16/04 3:26 PH Page 507 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



507 



An unrepaired laceration of an artery with 
an inevitable periarterial hematoma usually 
has partial liquefaction of the latter, and a com- 
munication is established between the artery 
and the hematoma. Apseudocapsule of con- 
nective tissue forms gradually, and the pul- 
sating hematoma becomes a false aneurysmal 
sac. The lesion will usually continue to expand, 
often causing pressure symptoms. One of the 
most easily recognizable results of pressure is 
a neuropathy, such as the easily recognizable 
neurologic deficits that develop in the hand 
from pressure on the median nerve by a false 
aneurysm (Fig. 24-45). False aneurysms may 
eventually rupture. The potential for exsan- 
guinating hemorrhage endangers not only the 
limb but also the patient's life. If there is an 
associated infection, the threat of rupture is 
even greater (Fig. 24-46) . 

The size, configuration, and location of 
false aneurysms can vary greatly. The false 
aneurysm can be one single sac (Fig. 24-47) 
or it can be bilobed (Fig. 24-48). The distri- 
bution of 82 false aneurysms treated in World 
War II shows that the brachial artery was 
involved most often, followed by the popliteal 



artery (see Table 24-6) . The anatomic region 
most often involved with 558 false aneurysms 
and AVFs in Vietnam casualties was the lower 
extremity (see Table 24-9) . The most common 
involved arteries were the posterior tibial and 
brachial, followed closely by the superficial 
femoral and popliteal arteries (see Table 
24-10). Multiple lesions can exist. Table 
24-4 shows that 41 out of the 509 Vietnam 
casualties had two or more lesions, for a total 
of 90 separate lesions (Fig. 24-49). 

Expanding false aneurysms can cause 
neurologic changes due to direct pressure on 
major nerves (Fig. 24-50). Shumacker and 
Carter (1946) emphasized the high frequency 
of false aneurysms of upper extremity major 
arteries with associated nerve lesions that 
required operations (Table 24-25). 

Usual pathologic changes can occur with 
false aneurysms. Distal embolization of a 
thrombus (Fig. 24-51) from a false aneurysm 
(Fig. 24-52) is unusual, but the potential 
threatwith possible disastrous sequelae always 
exists. Sachtello, Ernst, and Griffen (1974) 
described the case of one patient with a false 
subclavian aneurysm who had distal embolism 




■ FIGURE 24-45 

Pressure from a false aneurysm can compress an adjacent nerve. Fairly rapid expansion of the 
false aneurysm of the brachial artery caused external compression of both the median and the 
ulnar nerves with resultant neurologic deficit. (From Rich NM: In: Beebe HG, ed. Complications 
in vascular surgery. Philadelphia: JB Lippincott, 1973.) ■ 



ch24.qxd 4/16/04 3:26 PH Page 508 



508 



V • SPECIAL PROBLEMS AND COMPLICATIONS 




■ FIGURE 24-46 

A false aneurysm associated with surrounding infection has an increased potential for rupture and 
exsanguinating hemorrhage. An infected false aneurysm of the superficial femoral artery resulted 
in intermittent hemorrhage through the open wound of the thigh in a Vietnam casualty. (From NMR, 
Vietnam Vascular Registry #837.) ■ 




■ FIGURE 24-47 

A false aneurysm can exist in a large variety of 
sizes and configurations. It may be a single 
sac, as shown in this arteriogram. (From NMR, 
Vietnam Vascular Registry #2590.) ■ 



■ FIGURE 24-48 

Among the variety of configurations of false 
aneurysms is a double or bilobed sac, as 
shown in this arteriogram of the common 
carotid artery. The offending fragment is seen 
adjacent to the carotid artery. (From NMR, 
Vietnam Vascular Registry #826.) ■ 



ch24.qxd 4/16/04 3:26 PH Page 509 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



509 



TABLE 24-25 

PERIPHERAL NERVE LESIONS ASSOCIATED WITH ARTERIAL ANEURYSMS 



Artery Involved 



Nerve Lesion 
Requiring 
Operation 



Nerve Lesion Not 
Requiring 
Operation 



No Nerve 
Lesion 



Brachial 

Axillary 

Subclavian 

Popliteal 

Femoral 

Others 

Total 



No. 



% 



No. 



% 



No. 



24 


85.7 


1 


3.6 


3 


10.7 


11 


73.3 


1 


6.7 


3 


20.0 


3 


60.0 


1 


20.0 


1 


20.0 


2 


9.5 


6 


28.6 


13 


61.9 


1 


6.2 


3 


18.8 


12 


75.0 


_7 


13.0 


13 


24.0 


34 


63.0 


48 


40.3 


25 


21.0 


66 


38.7 



Modified from Shumacker HB Jr, Carter KL: Arteriovenous fistulas and arterial aneurysms in military personnel. Surgery 
1946;20:9-25. 




■ FIGURE 24-50 

There is a groove made by the median nerve 
in the excised axillary false aneurysm. Direct 
pressure on the median nerve had resulted in a 
neuropathy. (From Elkin DC: Vascular injuries of 
warfare. Ann Surg 1944;120:284-310.) ■ 



■ FIGURE 24-49 

Multiple lesions can occur, as evidenced 
by this Vietnam casualty who had a false 
aneurysm of the anterior tibial artery, which was 
obvious, and a false aneurysm of the distal 
popliteal artery, which was diagnosed only by 
angiography. (From NMR, Vietnam Vascular 
Registry #51 89.) ■ 



ch24.qxd 4/16/04 3:26 PH Page 510 



510 



V • SPECIAL PROBLEMS AND COMPLICATIONS 




■ FIGURE 24-51 

Mural thrombus may embolize from either a 
false aneurysm or an arteriovenous fistula. This 
angiogram demonstrates an embolus from a 
proximal popliteal artery false aneurysm to the 
distal popliteal and proximal posterior tibial 
arteries. (From Rich NM: In: Beebe HG, ed. 
Complications in vascular surgery. 
Philadelphia: JB Lippincott, 1973.) ■ 



from a thrombus within a false aneurysm. They 
managed the problem by resection of the 
clavicle, resection of the subclavian false 
aneurysm with vein graft replacement and 
brachial arterial embolectomy. Pulses were 
restored. Rhodes, Cox, and Silver (1973) 
reported the unusual complication of con- 
sumption coagulopathy, which developed in 
a patient with a false aneurysm and an AVE 



Clinical Features 

The most obvious clinical finding with a false 
aneurysm is a mass that is usually pulsatile. 
There is often evidence of a penetrating 
wound (Fig. 24-53) . The mass may or may 
not be painful. On examination, the borders 
of the mass can be ill defined because the false 
aneurysm is beneath the deep fascia. Depend- 
ing on the amount of thrombus within the 
false aneurysm, the mass may or may not be 
pulsatile. There is often an associated systolic 
bruit over the mass, and there can be con- 
siderable radiation of the bruit into the 
surrounding anatomy. 

Gradual enlargement of the false aneurysm 
may occur (Fig. 24-54), with the development 



■ FIGURE 24-52 

Except for 14% of the lesions that were 
associated with external hemorrhage into open 
wounds, there were rare preoperative 
complications associated with arteriovenous 
fistulas and false aneurysms in this series. One 
of these was embolization of thrombus from 
popliteal arterial false aneurysm, identified in 
the arteriogram with its adjacent wounding 
fragment. In 1968 at Walter Reed Army Medical 
Center, the false aneurysm was resected, the 
thrombus was removed with a Fogarty catheter, 
and arterial continuity was reestablished by 
end-to-end anastomosis. (From Rich NM, 
Hobson RW II, Collins GJ Jr: Traumatic 
arteriovenous fistulas and false aneurysms: A 
review of 558 lesions. Surgery 1975;78:817- 
828.) ■ 




ch24.qxd 4/16/04 3:26 PH Page 511 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



511 




■ FIGURE 24-53 

The diagnosis of a false aneurysm may be obvious with the physical finding of a pulsating mass. 
There is usually evidence of a penetrating wound. (From NMR, Vietnam Vascular Registry #3273. 




■ FIGURE 24- 

Enlargement of a false aneurysm may be gradual, or there may be rapid expansion of a mass. The 
size of the mass may also be quite variable, as in this large false aneurysm of the profunda femoris 
artery. The mass may be painful, and there may be warmth and tenderness on examination. (From 
NMR, Vietnam Vascular Registry #3159.) ■ 



ch24.qxd 4/16/04 3:26 PH Page 512 



512 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



of a firm, warm, tender area. Confusion with 
an abscess has occurred in the differential 
diagnosis. A stable false aneurysm of longer 
duration can also be confused with a cyst or 
neoplasm. 

If the false aneurysm is associated with an 
AVF, a continuous bruit and thrill over the 
sight of injury may also be present. As previ- 
ously noted, pressure on adjacent nerves may 
result in neurologic deficits, and the first 
symptom or physical finding may result from 
neuropathy. Distal pulses are usually intact and 
considered to be normal on examination. 



Diagnostic Considerations 

Diagnosis is usually made by physical exami- 
nation of a pulsatile mass. Roentgenograms 
in the anteroposterior and lateral views might 
identify an offending metallic foreign body 
in the anatomical location of an artery. 



Nevertheless, angiography may be necessary 
to establish the diagnosis (Fig. 24-55). The 
size of the false aneurysm may be misleadingly 
small because of the amount of laminated clot 
filling the sac (Fig. 24-56) . Angiography may 
delineate a clinically unsuspected adjacent 
AVF (Fig. 24-57) or multiple vascular lesions, 
as demonstrated in Fig. 24-49. 

Angiography may be necessary to make the 
diagnosis of the false aneurysms in arteries 
that are not easily acceptable to physical exam- 
ination, such as those within the chest and 
abdomen (Fig. 24-58). 

Newer investigative techniques, such as 
sonography, can also be valuable in deter- 
mining the size and location of false 
aneurysms. This was emphasized by Bole, 
Purdy, and Munda (1976) (Fig. 24-59). The 
diagnostic value of sonography for both true 
aneurysms and false aneurysms has been 
demonstrated with increasing utilization of 
this modality at Walter Reed General Hospital. 




■ FIGURE 24-55 

Preoperative angiography is helpful in confirming clinical impressions, outlining the site of the 
vascular defect, and ruling out additional adjacent vascular injuries. When not preoperatively 
available or practical, angiograms can be obtained easily. This one demonstrates a popliteal arterial 
false aneurysm seen at Walter Reed Army Medical Center in 1969 before resection and end-to-end 
anastomosis. Similar angiograms in the operating room immediately after repair have helped 
establish the status of vascular repair. (From Rich NM, Hobson RW II, Collins GJ Jr: Traumatic 
arteriovenous fistulas and false aneurysms: A review of 558 lesions. Surgery 1975;78:817-828.) ■ 



ch24.qxd 4/16/04 3:26 PM Page 513 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



513 




■ FIGURE 24-56 

This series of film in an angiogram of the right brachial artery demonstrates early filling of the false 
aneurysm adjacent to the offending fragment (left); the obvious false aneurysm, which was 
angiographically much smaller than the large palpable mass because of the laminated clot that 
filled the false aneurysm sac (middle); and residual contrast in the false aneurysm sac (right). (From 
NMR, Vietnam Vascular Registry #3159.) ■ 




■ FIGURE 24-57 

A large false aneurysm, such as this one demonstrated 
angiographically, can cause local arterial compression. 
In this Vietnam casualty, the large false aneurysm of the 
popliteal artery compressed the artery sufficiently to 
nearly obliterated the associated arteriovenous fistula. 
There was no associated classic "machinery-type" bruit, 
and the arteriovenous fistula was diagnosed 
angiographically. Also, the patient had weak pedal 
pulses because of the compression of the popliteal 
artery by the large false aneurysm, in contrast to what 
has previously been described in most patients who 
have intact distal pulses. (From NMR, Vietnam Vascular 
Registry #2513159.) ■ 



ch24.qxd 4/16/04 3:26 PM Page 514 



514 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



■ FIGURE 24-58 

Compression and lateral displacement of the 
abdominal aorta toward the patient's left side are 
demonstrated in this subtraction study of an 
angiogram of the aorta. The offending fragment 
from an M26 grenade caused a large false 
aneurysm of the aorta, which was repaired by 
lateral suture technique with interrupted sutures 
at Walter Reed Army Medical Center in 1967. 
(From Rich NM, Hobson RW II, Collins GJ Jr: 
Traumatic arteriovenous fistulas and false 
aneurysms: a review of 558 lesions. Surgery 
1975;78:817-828.) ■ 





■ FIGURE 24-59 

Ultrasonic tomography of the upper abdomen in a transverse plane showing the pseudoaneurysm 
(open arrow) in a patient with nonpulsatile diffuse mass. Thrombus echoes and irregular contour of 
the aneurysm are noted (solid arrows). Vertebral body (V) and the left kidney (K) are also visualized. 
(From Bole PV, Purdy RT, Munda R, et al: Traumatic pseudoaneurysms: A review of 32 cases. J 
Trauma 1976;16:63-70.) ■ 



ch24.qxd 4/16/04 3:26 PH Page 515 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 515 




Left Brachial 

SupProfyj 

. Liqaiu re 



Prof 




//Radial 
■Recur r. 



R ec r/;A\Recurr.\\ 
Ulnar Radial 

■ FIGURE 24-60 

The original photograph and drawing shown in the report of Dr. Matas, Philadelphia Medical News, 
October 27, 1988, when he proposed his endoaneurysmorrhaphy approach to the management of 
false aneurysms. (From Elkin DC: Traumatic aneurysm; Matas operation — 57 years after. Surg 
Gynecol Obstet 1946;82:1-12.) ■ 



Surgical Treatment 

The report of the first operation performed 
by Matas in 1 888 was presented again by Elkin 
(1946) to emphasize that the Matas operation 
has stood the test of time for 5 7 years and had 
had a profound impact on the surgery of blood 
vessels (Fig. 24-60) . 

On April 6, 1988, I operated on a young 
male Negro for a very large traumatic (mul- 
tiple gunshot) aneurysm of the brachial 
artery, extending from the armpit to the elbow, 
which opened my eyes to the possibilities of 
an entirely new method of conservative treat- 
ment, which was to revolutionize my previous 
notions of aneurysmal surgery. In this case, 
successive ligation of the main artery on the 
proximal and distal poles of the aneurysm had 
been followed by relapse, and it seemed to 
me that I had no other alternative but to 
extirpate the sac. When I exposed the sac and 
emptied its contents, the failure of the liga- 
tions to control the circulation was easily 
explained by the appearance in the bottom 
of the sac of the large orifices corresponding 
to the collateral branches, which opened into 
the sac in the segment of the artery included 



in between the ligatures (Fig. 24-61). It was 
evident that it was these collateral orifices that 
fed the sac despite the ligatures that had been 
placed at each one of its poles. I, at first, 
intended to secure these collaterals by excis- 
ing the sac, but the branches of the brachial 
plexus of nerves were so densely incorporated 
in its walls that I could have not dissected them 
and detached them, without serious damage, 
thereby paralyzing the arm. It occurred to me 
then that the easiest way out of this awkward 
dilemma was to steal the orifices of all the 
bleeding collaterals by suturing them as we 
would an intestinal wound, leaving the sac 
attached and undisturbed in the wound. This 
procedure was at once put into effect and the 
hemostasis was so perfect and satisfactory that 
it seemed to me strange that no one should 
have thought of so simple an expedient 
before. 

Matas used the intrasaccular suture (see 
Figs. 24-41 and 24-61); however, he was also 
interested in reconstructive endoaneury- 
smorrhaphy (Fig. 24-62). 

In the management of a false aneurysm, 
repair of the arterial defect is usually the goal 
that should be sought. An elective incision 



ch24.qxd 4/16/04 3:26 PH Page 516 



516 



V • SPECIAL PROBLEMS AND COMPLICATIONS 




■ FIGURE 24-6 1 

The Matas obliterative aneurysmorrhaphy is 
demonstrated in this intrasaccular suture 
ligature of a ruptured popliteal aneurysm. The 
dotted line shows the area of extravasation 
filled with clot. Arterial reconstruction was 
considered impractical in this specific case, 
and the distal and proximal popliteal arteries 
were obliterated with encircling sutures. 
Collateral circulation was adequate to maintain 
extremity viability. (From Matas R: Surgery, vol. 
7. Philadelphia: WB Saunders, 1921.) ■ 



should be made thatwill allow adequate expo- 
sure for proximal and distal control. Exam- 
ples were afforded by the extensive World War 
II experience. Shumacker (1946) described 
in detail the incisions that could be success- 
fully employed in the surgical approach to 
aneurysms, especially those in antecubital 
(Fig. 24-63) and popliteal (Fig. 24-64) fossae. 
His report was based on his extensive 
experience in managing false aneurysms in 
hundreds of American combat casualties. 
Specifically, these incisions were devised to 
replace longitudinal incisions across the 
popliteal and antecubital creases, which were 
often associated with heavy scars or keloids, 



contracture, or ulceration. Although resection 
of the false aneurysm is often recommended, 
an alternative plan that is presently employed 
has several advantages. The laminated clot 
within the false aneurysm should be evacuated 
after temporary proximal and distal control 
is obtained with vascular clamps, but the major- 
ity of the sac can usually be left in place. This 
will shorten the length of the operative pro- 
cedure and decrease the possibility of damage 
to associated structures, such as tearing of the 
popliteal vein, which has become closely 
adherent and attenuated to an adjacent 
popliteal arterial false aneurysm. If the false 
aneurysm is inadvertently entered before 
obtaining proximal and distal control, digital 
control will usually suffice as an expedient 
measure. Unnecessary resection of normal 
artery can also be avoided if careful dissec- 
tion of both the proximal and the distal artery 
toward the side of the defect is carried out. 
In this manner, a more limited resection of 
artery will be necessary. 

Occasionally, lateral suture of a punctuate 
wound of an artery is possible, without con- 
striction of the arterial lumen. However, 
limited arterial resection and end-to-end anas- 
tomosis is usually the procedure of choice. If 
it is necessary to use an arterial replacement, 
an autogenous vein graft is usually preferred. 
This graft should be placed in tissue as normal 
as possible, which is often difficult because of 
considerable inflammation and cicatrix. If 
there is extensive scarring, it might be possi- 
ble to place a graft in an extra-anatomic area 
of adjacent tissues in a position away from the 
usual course of the major vessel. In the case 
of noncritical arteries, such as the radial artery 
or distal posterior tibial artery, ligation is 
usually satisfactory. This is particularly impor- 
tant if there is an infected false aneurysm. 
However, arterial repair is preferred even in 
small caliber arteries. 

Surgical correction of a false aneurysm 
should be performed as soon as possible after 
the diagnosis is made to prevent the compli- 
cations of rupture or rapid expansion with 
resultant pressure on adjacent nerves. Imme- 
diate surgery should be advocated if neuro- 
logic symptoms develop. 

Additional information related to specific 
arterial false aneurysms, such as those of the 



ch24.qxd 4/16/04 3:26 PH Page 517 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 517 




■ FIGURE 24-62 

In addition to the obliterative aneurysmorrhaphy for the treatment of the false aneurysms, Matas 
encouraged selective reconstructive endoaneurysmorrhaphy, as was used in this repair of a false 
aneurysm of the brachial artery. (From Matas R: Surgery, vol. 7. Philadelphia: WB Saunders, 
1921.) ■ 



ch24.qxd 4/16/04 3:26 PH Page 518 



518 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



■ FIGURE 24-63 

Incisions used in exposure of vessels in the antecubital 
fossa. A, The usual incision for exposure of brachial 
vessels in antecubital space. B, Incision used when the 
lesion is suspected in proximal portion of ulnar vessels. 
C, Incision used when brachial vessels are involved just 
proximal to antecubital creased. D, Incision used for 
exploration of distal end of brachial or proximal end of 
radial vessels. (From Shumacker HB Jr: Incisions in 
surgery of aneurysms, with special reference to 
exploration in antecubital and popliteal fossae. Ann Surg 
1946;124:586-598.) ■ 





■ FIGURE 24-64 

Skin incisions used in exploring the popliteal vessels in the surgical approach to false aneurysms in 
World War II. A, Incision used when the lesion exists in the midpopliteal space. B, Incision used 
when the lesion is higher in the popliteal fossa. C and D, Incisions used for exploring the distal 
popliteal vessels. E, A modified incision useful when associated nerve lesion require exploration. 
(From Shumacker HB Jr: Incisions in surgery of aneurysms, with special reference to exploration in 
antecubital and popliteal fossae. Ann Surg 1946;124:586-598.) ■ 



subclavian artery, can be found in specific 
chapters. Details of arterial repair and specific 
techniques, such as intraluminal control of 
hemorrhage with a balloon catheter, are to 
be found in Chapter 19. 

Elkin (1946) reported the results of oper- 
ating on 106 false aneurysms at the Ashford 
General Hospital Vascular Center in White 



Sulfur Springs, West Virginia, in a 30-month 
period. The Matas procedure was employed 
in 61 of the operations, and some other type 
of operation, usually complete excision of a 
small sac, was employed in the remaining 45 
cases. There were no deaths in his series, no 
recurrence of the false aneurysm, and no 
incidence of gangrene. Table 24-26 shows 



ch24.qxd 4/16/04 3:26 PH Page 519 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



519 



TABLE 24-26 

LOCATION OF INJURY AND NUMBER 
OF PATIENTS TREATED BY MATAS 
ENDOANEURYSMORRHAPHY: 
ASHFORD GENERAL HOSPITAL 
VASCULAR CENTER, WHITE SULFUR 
SPRINGS, WORLD WAR II EXPERIENCE 



Artery Involved 

Axillary 

Brachial 

Femoral 

Iliac 

Peroneal 

Popliteal 

Profunda femoris 

Radial 

Superior gluteal 

Tibial, anterior 

Tibial, posterior 

Ulnar 

Total 



Cases 

5 

14 
11 

2 

1 

7 

3 

4 

1 

3 

8 
_2 
61 



From Elkins DC: Traumatic aneurysm; Matas operation — 57 
years after. Surg Gynecol Obstet 1946;82:1-12. 



the location and number of injuries 
treated by endoaneurysmorrhaphy. Elkin and 
Shumacker (1955) outlined the operative 
treatment of 209 arterial aneurysms (Table 
24-27) . Nearly an equal number were treated 
by endoaneurysmorrhaphy as by excision. 
Shumacker (1948) reported successful results 
with vein graft repair of arteries that had false 
aneurysms (see Table 24-16). 

De Takats and Pirani (1954) stated that 
Herlyn, a pupil of Stich in Gottingen, reported 
he performed 164 ligatures and 230 recon- 
structive operations in World War II. This 
emphasized that the trend in German was 
surgery, and Herlyn felt that the artery should 
never be ligated for traumatic aneurysm 
unless it was small or the patient's life was in 
danger. 

Hughes and Jahnke (1958) reported on 215 
false aneurysms and AVFs treated at Walter 
Reed Army Medical Center in the early 1950s, 
mainly in casualties from the Korean Conflict. 
The various operations used in managing 43 
false aneurysms and 91 AVFs in large vessels 
are reviewed in Table 24-18. Similar data con- 



cerning smaller caliber arteries are given in 
Table 24-8. 

Rich, Hobson, and Collins (1975) reported 
the experience from the Vietnam War. There 
were 296 false aneurysms among 558 AVFs and 
false aneurysms identified in 509 patients. 
As might be anticipated by the number of 
American troops committed in Southeast 
Asia, the largest number of lesions resulted 
from wounds during 1968, with a relatively 
large number of lesions in 1967 and 1969. The 
time from injury to recognition of the AVFs 
and false aneurysms was arbitrarily considered 
to be immediate if recognized within the first 
24 hours, early if recognized between the 
second and thirtieth day, delayed if recognized 
between the second through the sixth month, 
and remote if recognized after 6 months. In 
this study, the largest number of lesions (273 
or 48.9%) was recognized in the early period 
(see Table 24-20) . A nearly equal number of 
lesions, 228, was recognized in the delayed 
period. In the immediate group, there were 
22 acute lesions operated on in Vietnam. In 
the remote group, all but 7 of the 35 patients 
had recognition and treatment of their lesions 
in less than 2 years. Only two had recognition 
and treatment of their lesions more than 5 
years following the initial injury, and both were 
less than 6 years. Nearly an equal number of 
operations were performed in the inter- 
mediate hospitals, mainly in Japan, and in 
hospitals in the continental United States (see 
Table 24-21). Several hundred surgeons were 
involved in performing the repairs. Table 
24-22 outlines the methods of treatment used 
for the various repairs. Arterial ligation was 
used in 290 lesions, 52%. The overall mor- 
bidity rate for the 509 patients was 1.8% (seven 
deaths) (see Table 24-23). Only two of these 
deaths could be directly attributed to the vas- 
cular problem. One patient died from a rup- 
tured external iliac arterial false aneurysm. 
The overall morbidity included five compli- 
cations, for a morbidity rate of 6.3%. Hem- 
orrhage occurred in 14, thrombosis in 12, and 
stenosis in 2. 

The numerous reports of the management 
of false aneurysms from civilian experience 
range from individual case reports to reports 
of 20 to 30 lesions. However, the civilian expe- 
rience has not been as extensive as the warfare 



ch24.qxd 4/16/04 3:26 PH Page 520 



520 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



TABLE 24-27 

TECHNIQUES OF OPERATIVE TREATMENT IN 209 ARTERIAL ANEURYSMS: 
WORLD WAR II EXPERIENCE 



Location 

Upper extremity 
Axillary 
Brachial 
Radial 
Subclavian 
Ulnar 

Lower extremity: 
Femoral 
Gastrocnemius, 

muscle branch 
Peroneal 
Popliteal 

Profunda femoris 
Profunda branch 
Tibial, anterior 
Tibial, posterior 

Head and neck: 
Carotid 

Cervical, deep 
Temporal, superficial 

Trunk: 

Gluteal, superior 
Iliac 

Innominate 
Thoracic, lateral 
Thoracoacromial 

Total 



Endoaneurysmorrhaphy 

10 
16 

7 

2 

3 

14 



2 

19 

6 

2 
5 
7 



99 





Proximal 


End-to-End 


Total No 


Excision 


Ligation 


Anastomosis 


Cases 


24 


1 




35 


30 


— 


1 


47* 


5 


— 


— 


12 


10 


1 


— 


13+ 


4 


— 


— 


7 


4 
1 


— 


— 


18* 

1 


2 


— 


— 


2 

215 

6 

2 
5 

15' 


8 


— 


— 


5 


8 


— 


13' 
1 
2 


2 


— 


— 


1 


1 


= 


2 

4 
1 


1 

1 


I 


z 


1 
1 



98 



11 



209 



*This total does not include one aneurysm in which cure occurred spontaneously. 

+ This total does not include two aneurysms in which methods of management were not stated. 

*This total does not include two aneurysms in which cure occurred spontaneously. 

§ This total does not include three aneurysms: two in which methods of management were not stated and one in which 

spontaneous cure occurred. 

'This total does not include two aneurysms in which cure occurred spontaneously. 

From Elkin DC, Shumacker HB Jr: In: Vascular Surgery in World War II. Elkin DC, DeBakey ME, eds. Washington, DC: US 

Government Printing Office, 1955. 



experience in this century. Examples of the 
civilian experience are the reports of Lloyd 
(1957), Baird and Doran (1964), Engelman, 
Clements, and Herrmann (1969), and Bole, 
Purdy, and Munda (1976). Particularly note- 
worthy is the report of the management of 23 
traumatic false aneurysm in 23 patients treated 
in New York City over a 5-year period starting 
in 1968. Table 24-28 outlines the method of 



management of these false aneurysms, with 
lateral suture repair, resection and end-to-end 
anastomosis, and ligation being used almost 
equally. These authors reported no mortality, 
no recurrence, and no distal edema or arte- 
rial insufficiency. They did have two patients 
who continued to have pain, and three wound 
infections occurred. 



ch24.qxd 4/16/04 3:26 PH Page 521 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



521 



TABLE 24-28 

TYPE OF REPAIR: CIVILIAN 
EXPERIENCE IN NEW YORK CITY 

Resection and end-to-end anastomosis 8 

Resection and graft replacement 1 

Lateral repair 7 

Ligation 5 

Spontaneous closure 1 

Refused treatment 1 

Total 23 

From Bole PV, Munda R, Purdy RT, et al: Traumatic 
pseudoaneurysms: a review of 32 cases. J Trauma 
1976;16:63-70. 



Spontaneous Cure 

Shumacker and Wayson (1950) evaluated 
spontaneous cure of false aneurysms and 
AVFs. They studied 122 aneurysms and 245 
AVFs. They felt that thrombosis was respon- 
sible for the obliteration of these lesions. 
Because there were only eight satisfactory 
spontaneous cures of false aneurysm — 6.6% 
of 22 lesions — these authors though that there 
was little merit in awaiting the possibility of 
this occurrence. This was only slightly better 
than the 2% spontaneous cure for AVFs. 
Although some case reports have been 
documented, recent experience has not 
witnessed a change in the low incidence of 
spontaneous cure of false aneurysms. This is 
undoubtedly also affected by early surgical 
intervention for the mass majority of false 
aneurysms. 



Follow-up 

Hughes andjahnke (1958) provided a 5-year 
follow-up study of 250 AVFs and false 
aneurysms treated at Walter Reed Army 
Medical Center. Most of the patients had been 
injured during the Korean Conflict. This long- 
term follow-up was one of the first and one of 
the few extensive follow-up studies to be con- 
ducted. This study emphasized the difficulty 
in evaluating vascular trauma in combat casu- 
alties because of the many associated injuries. 
Neither of the two deaths in this follow-up 



study were related to vascular problems 
following repair of false aneurysms. Residual 
pain, coldness, and claudication in the 
involved extremity were noted; however, no 
distinction was made between those patients 
treated for AVFs and those treated for false 
aneurysms. In the follow-up of the Vietnam 
casualties through the Vietnam Vascular 
Registry, more than one fourth — 29.3% — or 
149 of the 509 patients with AVFs and false 
aneurysms have been evaluated at Walter 
Reed Army Medical Center. In the long-term 
follow-up, which extends to 10 years for many 
patients, additional problems and sympto- 
matic residuals have been limited. Unfortu- 
nately, some of these patients have been lost 
to the long-term follow-up effort because of 
untimely deaths. One patient was killed in 
subsequent action during a second tour in 
Vietnam, and another patient died in an 
automobile accident. In the civilian reports, 
there are very limited data regarding long- 
term follow-up of patients who have false 
aneurysms. While few wanted to be reminded 
of the unfortunate Vietnam experience for 
more than 25 years in the United States, as 
we enter the twenty-first century, there is a 
realization that data in the Vietnam Vascular 
Registry are of value. A concerted effort is 
being developed to expand the long-term 
follow-up, which will be helpful to collective 
experiences in the civilian community and in 
responding to military interventions. 

REFERENCES 

Albuquerque FCJavedan SP, McDougall CG: Endo- 
vascular management of penetrating vertebral 
artery injuries. J Trauma 2002;53(3) :574-580. 

Annandale T: Traumatic popliteal arteriovenous 
aneurysm treated successfully by ligature of the 
popliteal artery and vein. Lancet 1875;1:568. 

Anthopoulos LP, Johnson JB, Spellman M: 
Arteriovenous Fistula in multiple saccular 
arterial aneurysms of a finger, following child- 
hood human bite. Angiology 1965;16:89. 

Baird RJ, Doran ML: The false aneurysm. Can Med 
AssocJ 1964;91:281-284. 

Basse ttFH, HouckWS: False aneurysm of profunda 
femoris artery after subtrochanteric osteotomy 
and nail-plate fixation. J Bone Joint Surg 1964; 
46A:583-585. 



ch24.qxd 4/16/04 3:26 PH Page 522 



522 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



Basse tt FH, Silver D: Arterial injury associated with 
fractures. Arch Surg 1966;92:13-19. 

Beattie EJJr, Oldham JB, RossJA: Superior thyroid 
arteriovenous aneurysm. Br J Surg 1961 ;48:456- 
457. 

Bell D, Cockshott WP: Angiography of traumatic 
arterio-venousfistulae. Clin Radiol 1965;16:241- 

247. 

Bickham WS: Arteriovenous aneurysm. Ann Surg 
1904;39:767-775. 

Billings KJ, Nasca RJ, Griffin HA: Traumatic arte- 
riovenous fistula with spontaneous closure. J 
Trauma 1973;13:741-743. 

Blackmore TL, Whelan TJ: Arteriovenous fistula 
as a complication of gastrectomy. AmJ Surg 1 965; 
109:197. 

Bole PV, Munda R, Purdy RT: Traumatic pseudo- 
aneurysms: A review of 32 cases. J Trauma 
1976;16:63-70. 

Bole PV, Purdy RT, Munda RT, et al: Civilian arterial 
injuries. Ann Surg 1976;183:13-23. 

Branham HH: Aneurysmal varix of the femoral 
artery and vein following a gunshot wound. Int 
JSurgl890;3:250. 

Callander CL: Study of arteriovenous fistula with 
an analysis of 447 cases. Ann Surg 1920;19:428- 
459. 

Cameron HS, Laird Jj, Carroll SE: False aneurysms 
complicating closed fractures. J Trauma 1972; 
12:67-74. 

Conn JH, Hardy JD, Chavez CM, Fain WR: Chal- 
lenging arterial injuries. J Trauma 197l;ll:167- 
177. 

Creech O Jr, Gantt J, Wren H: Traumatic arteri- 
ovenous fistula at unusual sites. Ann Surg 
1965;161:908-920. 

Dameron TB Jr: False aneurysm of femoral pro- 
fundus artery resulting from internal-fixation 
device (screw) J Bone Joint Surg 1964;46A:577-580. 

De Takats G, Pirani C: Aneurysms: General con- 
siderations. Angiology 1954;5:173-208. 

Dillard BM, Nelson DL, Norman HG: Review of 85 
major traumatic arterial injuries. Surgery 1968; 
63:391-395. 

DrapanasT, Hewitt RL, WeichertRF III, Smith AD: 
Civilian vascular injuries: A critical appraisal of 
three decades of management. Ann Surg 1970; 
172:351-360. 

Eisenbrey AB: Arteriovenous aneurysm of the sup- 
erficial femoral vessels.JAMA 1913;61:2155-21 57. 

Elkin DC: Arteriovenous aneurysm. Surg Gynecol 
Obstet 1945;80:217-224. 



Elkin DC: Traumatic aneurysm: the Matas 

Operation 57 years after. Surg Gynecol Obstet 

1946;82:1-12. 
Elkin DC, Banner EA: Arteriovenous aneurysms 

following surgical operations. JAMA 1946;131: 

1117-1119. 

Elkin DC, DeBakey ME: Vascular surgery in World 
War II. Washington, DC, US Government 
Printing Office, 1955. 

Engleman RM, Clements JM, Herrmann JB: Stab 
wounds and traumatic false aneurysms in the 
extremities. J Trauma 1969;9:77-87. 

Ferguson WM: Arteriovenous aneurysm following 
osteotomy for genu valgum. Lancet 1914;1:532. 

Freeman NE, Shumacker HB Jr: Vascular surgery. 
In Elkin DC, DeBakey ME (eds): Washington, 
DC, US Government Printing Office, 1955. 

Gaspard DJ, Caspar MR: Arteriovenous fistula after 
Fogarty catheter thrombectomy. Arch Surg 
1972;105:90-92. 

Hallowell. Extract of a letter from Mr. Lambert, 
surgeon at Newcastle upon Tyne, to Dr. Hunter, 
giving an account of new method of treating an 
aneurysm. Med Obser Inq 1762;30:360. 

Halsted W: Discussion in Bernheim, BM. Bull Johns 
Hopkins Hosp 1916;27:93. 

Harris JD: A case of arteriovenous fistula following 
closed fracture of tibia and fibula. Br J Surg 
1963;50:774-776. 

Heaton LD, Hughes CW, Rosegay H, et al: Military 
surgical practices of the United States Army in 
Vietnam. In Current Problems in Surgery. 
Chicago, Year Book Medical Publishers, 1966. 

Hershey FB. Secondary repair of arterial injuries. 
Am Surg 1961;27:33-41. 

Hewitt RL, Collins DJ: Acute arteriovenous 
fistulas in war injuries. Ann Surg 1969;169:447- 
449. 

Hewitt RL, Smith AD, Drapanas T: Acute traumatic 
arteriovenous fistulas. J Trauma 1973;13:901-906. 

Holman E: Arteriovenous Aneurysms: Abnormal 
Communication between the Arterial and 
Venous Circulations. New York, Macmillan, 1937. 

Holman E: Clinical and experimental observations 
on arteriovenous fistulae. Ann Surg 1940;112: 
840-875. 

Holman E: Contributions to cardiovascular physi- 
ology gleaned from clinical and experimental 
observations of abnormal arteriovenous com- 
munications. J Cardiovasc Surg 1962;3:48-63. 

Holman E: Abnormal arteriovenous communica- 
tions. Great variability of effects with particular 



ch24.qxd 4/16/04 3:26 PM Page 523 



24 • ARTERIOVENOUS FISTULAS AND TRAUMATIC FALSE ANEURYSMS 



523 



reference to delayed development of cardiac 
failure. Circulation 1965;32:1001-1009. 

Holman E: Abnormal Arteriovenous Communi- 
cations: Peripheral and Intracardiac, Acquired 
and Congenital, 2nd ed. Springfield, 111, Charles 
C Thomas, 1968. 

Holman EF: Physiology of an arteriovenous fistula. 
Arch Surg 1971;7:64-82. 

Hughes CW: Acute vascular trauma in Korean War 
casualties: An analysis of 180 cases. Surg Gynec 
Obstet 1954;99:91-100. 

Hughes CW,Jahnke EJJr: The surgery of traumatic 
arteriovenous fistulas and aneurysms: A five-year 
follow up study of 215 lesions. Surg Clin North 
Am 1958;148:790-797. 

Hunt TK, Leeds FH, Wanebo HJ, Blaisdell FW: 
Arteriovenous fistulas of major vessels of the 
abdomen. J Trauma 1971 ;1 1 :483-493. 

Hunter W: The history of an aneurysm of the aorta, 
with some remarks on aneurysms in general. Med 
Obs Soc Phys Lond 1757;1:323. 

Hunter W: Further observations upon a particu- 
lar species of aneurysm. Med Obs Soc Phys Lond 
1762;2:390. 

Johnson G Jr, Blythe WB: Hemodynamic effects of 
arteriovenous shunts used for hemodialysis. 
Ann Surg 1970;171:715-723. 

Johnson GJr, Peters RM, Dart CH Jr: A study of 
cardiac vein negative pressure in arterio- 
venous fistula. Surg Gynecol Obstet 1967;1 24:82- 
86. 

Lai MD, Hoffman HB, Adamkiewicz JJ: Dissecting 
aneurysm of internal carotid artery after non- 
penetrating neck injury. Acta Radiol Diag 
1966;5:290-295. 

Lester J: Arteriovenous fistula after percutaneous 
vertebral angiography. Acta Radiol Diag 
1966;5:337-340. 

LeVeen H, Cerruti M: Surgery of large inaccessible 
arteriovenous fistulas. Ann Surg 1963;158:285- 
289. 

Lewis T: The adjustment of blood flow to the 
affected limb in arteriovenous fistula. Clin Sci 
1940;4:277-285. 

Lexer E: Die ideale operation des arteriellen und 
des arteriell-venosen aneurysma. Arch Klin Chir 
1907;83:459-477. 

Lord R, Ehrenfeld W, Wylie EJ: Arterial injury from 
the Fogarty catheter. Med J Aust 1968;2:70-71. 

Matas R: Traumatic aneurysm of the left 
brachial artery — Incision and partial excision of 
sac: Recovery. Phil Med News 1888;53:462-466. 



Matas R: Traumatic arteriovenous aneurysms of the 
subclavian vessels, with an analytical study of 
fifteen reported cases, including one operated. 
Trans Am Surg Assoc 1901;19:237. 

Matas R: An operation for radical cure of aneurysm 
based on arteriography. Ann Surg 1903;37:161- 
196. 

Matas R: Recent advances in the technique of 
thoracotomy and pericardiotomy for wounds of 
the heart. South Med J 1908;1:75-81. 

Meyer TJ, Slager R: False aneurysm following 
subtrochanteric osteotomy. J Bone Joint Surg 
1964;46:581-582. 

Muller WJ, Goodwin W: Renal arteriovenous fistula 
following nephrectomy. Ann Surg 1956;144:240- 
244. 

Murphy J: Resection of arteries and veins injured 
in continuity end-to-end suture. Exp Clin Res 
Med Rec 1897;51:73-104. 

NakanoJ, DeSchryver C: Effects of arteriovenous 
fistula on systemic and pulmonary circulations. 
AmJ Physiol 1964;207:1319. 

Nicoladoni C: Phlebarteriectasie der rechten 
oberen extremitat. Arch Klin Chir 1875;18:252. 

Norris G: Varicose aneurysm at the bend of the 
arm: Ligature of the artery above and below the 
sac; secondary hemorrhages with a return of the 
aneurysm thrill on the tenth day; cure . AmJ Med 

Sci 1843;5:17. 

Osier W: Case of arterio-venous aneurysm of the 
axillary artery and vein of 1 4 years duration . Ann 
Surg 1893;17:37-40. 

Osier W: Report of a case of arteriovenous 
aneurysm of the thigh. Johns Hopkins Hosp Bull 
1905;16:119. 

Osier W: Remarks on arteriovenous aneurysm. 
Lancet 1915;1:949. 

Pate J, Sherman R, Jackson T, Wilson H: Cardiac 
failure following traumatic arteriovenous fistula: 
A report of 14 cases. J Trauma 1965;5:398- 
403. 

Patman R, Poulos E, Shires G: The management 
of civilian arterial injuries. Surg Gynecol Obstet 
1964;118:725-738. 

Petrovsky B, Milinov O: "Arterialization" and 
"venization" ofvessels involved in traumatic arte- 
riovenous fistulae: Aetiology and pathogenesis 
(an experimental study). J Cardiovasc Surg 
1967;8:396-407. 

Pick T: On partial rupture of arteries from exter- 
nal violence. St George's Hosp Rep (Lond) 
1873:6:161. 



ch24.qxd 4/16/04 3:26 PM Page 524 



524 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



Pick T: A clinical lecture of a case of arteriovenous 
aneurysm. Lond Med Times Gaz 1883;2:677. 

Pridgen W, Jacobs J: Postoperative arteriovenous 
fistula. Surgery 1962;51:205. 

Ranshoff J: Arteriovenous aneurysm of the supe- 
rior thyroid artery and vein. Surg Gynecol 
Obstetl935;61:816. 

Reid M: The effect of arteriovenous fistula upon 
the heart and blood vessels: An experiment and 
clinical study. Bull Johns Hopkins Hosp 
1920;31:43-50. 

Reid M: Abnormal arteriovenous communica- 
tions. Acquired and congenital, II. The orgin and 
nature of arteriovenous aneurysms, cirsoid 
aneurysms and simple angiomas. Arch Surg 
(Chicago) 1925;10:996-1009. 

Reid M, McGuire J: Arteriovenous aneurysm. Ann 
Surg 1938;108:643-693. 

Rhodes G, Cox C, Silver D: Arteriovenous fistula 
and false aneurysm as the cause of consumption 
coagulopathy. Surgery 1973;73:535-540. 

Rich NM, Hobson RW II, Collins GJ Jr: Traumatic 
arteriovenous fistulas and false aneurysms: 
A review of 558 lesions. Surgery 1975;78:817- 
828. 

Rob CG, Battle S: Arteriovenous fistula following 
the use of the Fogarty balloon catheter. Arch Surg 
1971;102:144-145. 

Sachtello C, Ernst CB, Griffen WJ: The acute 
ischemic upper extremity: Selected manage- 
ment. Surgery 1974;76:1002. 

Sako Y, Varco R: Arteriovenous fistula: Results of 
management of congenital and acquired forms, 
blood flow measurements and observations on 
proximal arterial degeneration. Surgery 1970;67: 
40-61. 

Schenk WJ, Bahn RA, Cordell A, Stephens J: The 
regional hemodynamics of acute experimental 
arteriovenous fistulas. Surg Gynecol Obstet 1957; 
105:733. 

Schwartz A: The historical development of methods 
of hemostasis. Surgery 1958;44:604. 



Seeley S: Vascular surgery at Walter Reed Army 
Hospital. US Armed Forces Med J 1954;8. 

Seeley S, Hughes CW, Cooke F, Elkin DC: Traumatic 
arteriovenous fistulas and aneurysms in war 
wounded. Am J Surg 1952;83:471-479. 

Shumacker HBJr: Incisions in surgery of aneurysm: 
With special reference to explorations in the 
antecubital and popliteal fossae. Ann Surg 
1946;124:586-598. 

Shumacker HB Jr: Problem of maintaining the 
continuity of artery in surgery of aneurysms and 
arteriovenous fistulae; notes on the development 
and clinical application of methods of arterial 
suture. Ann Surg 1948;127:207-230. 

Shumacker HBJr, Carter KL: Arteriovenous fistu- 
las and false aneurysms in military personnel. 
Surgery 1946;20:9-25. 

Shumacker HBJr, Stahl N: A study of the cardiac 
frontal area in patients with arteriovenous 
fistulas. Surgery 1949;26:928-944. 

Shumacker HBJr, Wayson EE: Spontaneous cure 
of aneurysms and arteriovenous fistulas, with 
some notes on intravascular thrombosis. Am J 
Surg 1950;79:532-544. 

Singh I, Gorman JF: Vascular injuries in closed 
fractures near junction of middle and lower 
thirds of the tibia. J Trauma 1972;12:592-598. 

Smith L, Foran R, Caspar MR: Acute arterial injuries 
of the upper extremity. AmJ Surg 1963;106:144- 
151. 

Soubbotitch V: Military experiences of traumatic 
aneurysms. Lancet 1913;2:720-721. 

Stewart F: Arteriovenous aneurysm treated by 
angiography (angiorrhaphy) . Abb Surg 1 91 3;57: 
247-254. 

Vollmar J, Krumhaar D: Surgical experience with 
200 traumatic arteriovenous fistulae. In Hiertonn 
T, Rybeck B (eds): Traumatic Arterial Lesions. 
Stockholm, Forsvarets Forskningsanstalt, 1968. 

White J, TalbertJ, HallerJAJr: Peripheral arterial 
injury in infants and children. Ann Surg 
1968;167:757-766. 



ch25.qxd 4/16/04 3:22 PM Page 525 




Thromboembolic 
Complications in 
Trauma Patients 

M. MARGARET KNUDSON 





o 


HISTORICAL PERSPECTIVES 




o 


PATHOPHYSIOLOGY 




o 


CLINICAL EPIDEMIOLOGY AND RISK FACTOR ANALYSIS 




o 


PROPHYLACTIC MEASURES 
Mechanical Prophylactic Devices 
Unfractionated Heparin 
Low-Molecular-Weight Heparin 
Prophylactic Vena Cava Filters 




o 


DIAGNOSISANDTREATMENTOFPOST-TRAUMATICTHROMBOEMBOLIC 
COMPLICATIONS 


o 


OUTCOMES RESEARCH IN PREVENTION OF THROMBOEMBOLISM IN 




TRAUMA PATIENTS 


o 


CURRENT RECOMMENDATIONS AND FUTURE DIRECTIONS 



HISTORICAL PERSPECTIVES 

In the 1934 volume of the American Journal of 
Pathology, McCartney initially suggested that 
there was an association between trauma and 



death from pulmonary embolism (PE), and 
that this association was particularly strong in 
patients with lower extremity fractures. This 
observation was followed by a number of 
autopsy studies that not only confirmed the 
relationship between injury and throm- 

525 



ch25.qxd 4/16/04 3:22PM Page 526 



526 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



boembolic complications but also further 
suggested that these thromboembolic events 
were rarely diagnosed premortem. These 
preliminary studies stimulated the sentinel 
work by Freeark, Boswick, and Fardin (1967) , 
who performed venograms on 124 trauma 
patients, demonstrating venous thrombosis in 
35% of fracture patients. Thrombus forma- 
tion was observed within 24 hours of injury 
and involved both the injured and the unin- 
jured extremity. More than two thirds of the 
patients with roentgenographic evidence of 
deep venous thrombosis (DVT) had no symp- 
toms or physical findings to suggest its occur- 
rence. These authors were among the first to 
advocate studies to examine the effectiveness 
of prophylactic measures in reducing postin- 
jury thromboembolism. 



PATHOPHYSIOLOGY 



The basic factors leading to the development 
of venous thrombosis have long been defined 
by the Virchow triad, which includes stasis, 
endothelial damage, and a prothrombotic 
state. In the microcirculation, a series of steps 
linking thrombosis and inflammation has 
been suggested. This inflammatory process 
involves platelets, neutrophils, monocytes, 
and substances released from the activated 
platelets and neutrophils, such as adenosine 
diphosphate, neutrophil-activating pep tide-2, 
and cathepsin G. More recently, direct injury 
to the venous endothelium, induced by the 
venodilation that occurs under anesthesia, has 
been implicated as the initiating step in this 
inflammatory process. The exposed suben- 
dothelial surface acts as a nidus for platelets 
and leukocytes, thus setting the stage for clot 
formation. 

Trauma patients are at risk for throm- 
boembolic complications for a number of 
reasons. Trauma patients are normally in a 
hypercoagulable state by the third day after 
trauma and often have depressed levels of 
an ti thrombin. Most trauma patients are immo- 
bilized for at least some period, and many are 
paralyzed to facilitate respiratory care or sec- 
ondary to neurologic injuries. Additionally, 
many trauma patients have direct venous 



injuries either associated with fractures or fol- 
lowing penetrating trauma. We have detected 
lower extremity thrombi by duplex sono- 
graphy within 12 hours of injury, and other 
investigators have documented that 6% of 
post-traumatic pulmonary emboli occur on 
day 1 following injury. Many of these injured 
patients are young and have no known pre- 
existing risk factors for thromboembolism. It 
is thus imperative that research in this field 
be directed toward understanding both the 
pathogenesis and the prevention of post- 
traumatic thromboembolism. 



CLINICAL EPIDEMIOLOGY AND 
RISK FACTOR ANALYSIS 

The overall incidence of post-traumatic DVT 
is estimated to be between 10% and 20% in 
patients who are not receiving any method of 
prophylaxis. The actual incidence will vary 
with such factors as the age of the patient, the 
nature of the injuries, the geographic loca- 
tion, and the method used to detect occult 
DVT. In addition to its association with 
potentially fatal PE, undetected (and thus 
untreated) DVT can also result in permanent 
postphlebitic changes. PE occurs in at least 
1% to 2% of injured patients, with an associ- 
ated mortality as high as 50%. The true inci- 
dence of PE is probably much higher, as most 
PEs in trauma patients are clinically silent. 
Additionally, a practical method of screening 
high-risk trauma patients for PE has not 
been developed. Importantly, many of the 
deaths attributable to PE occur in trauma 
patients who would otherwise recover fully 
from their injuries. This recognition has 
inspired many clinical investigators to attempt 
to describe the risk factors associated with the 
development of post-traumatic thromboem- 
bolic complications as the first step in pre- 
venting them. 

As mentioned, early autopsy studies docu- 
mented the association between injured 
patients with fractures and deaths from PE. 
Burned patients were also found to be at high 
risk for thromboembolism. More recently, 
various investigators have identified increased 
age, the presence of head injury, spinal cord 



ch25.qxd 4/16/04 3:22 PM Page 527 



25 • THROMBOEMBOLIC COMPLICATIONS 



527 



injury with paralysis, and prolonged immo- 
bilization as important risk factors for post- 
traumatic thromboembolism. Direct venous 
injury, either resulting from the trauma itself 
or induced by large-caliber venous access 
devices, has also been implicated. Burch and 
colleagues (1990) were among the first to 
warn of the risk of DVT and/or fatal PE fol- 
lowing ligation or repair of penetrating iliac 
vascular injuries. A report from Sue, Davis, 
and Parks (1995) supports the concept that 
direct iliac venous injuries are associated 
with a significant risk for thromboembolic 
complications. 

In a recent study that used venography 
to identify distal (calf) and proximal (thigh/ 
pelvic) lower extremity DVT, the incidence of 
DVT was found to be 54% in patients with 
head injuries and 62% in those with spinal 
injuries. Sixty-nine percent of injured patients 
with lower extremity or pelvic fractures who 
underwent venography had evidence of DVT 
within 14 to 21 days after the injury. The overall 
incidence of proximal DVT was 18%. Prophy- 
laxis against thromboembolism was not used 
in any of the patients in this study. 

A group of investigators dedicated to the 
field of thromboembolic research in injured 
patients recently collaborated in compiling a 
risk factor assessment scale. The risk factors 
included underlying conditions, iatrogenic 
factors, injury-related factors, and age (Table 
25-1) . Each factor was given a weight, based 
on the perceived association with the devel- 
opment of DVT/PE (i.e., a weight of 2 was 
relatively low risk and a score of 4 represented 
the highest risk factors) . When adding up the 
weighted scores for each patient, a trauma 
patient with a score of 5 or greater was con- 
sidered at high risk for thromboembolic com- 
plications and a candidate for prophylaxis. 
Further research by this group, using a 
prospective study, confirmed that five of these 
factors were significantly associated with the 
development of post-traumatic DVT. Patients 
with one or more of these five factors had an 
overall rate of DVT at 10% despite aggressive 
prophylaxis. It should be noted that although 
the authors of this particular study did not 
find spinal cord injury to be a significant risk 
factor, this likely represents a type II statisti- 
cal error due to a low number of patients 



TABLE 25-1 




RISK ASSESSMENT PROFILE 




Risk Factors Points 


Assigned 


Underlying condition 




Obesity* 


2 


Malignancy 


2 


Abnormal coagulation 


2 


History of thromboembolism 


3 


Iatrogenic factors 




Femoral venous lines 


2 


Transfusion > 4 units* 


2 


Operation > 2 hours* 


2 


Major venous repair 


3 


Injury-related factors 




ChestAIS>2 


2 


Abdomen AIS > 2 


2 


Head AIS>2* 


2 


Spinal fractures 


3 


Glasgow Coma Scale score < 8 


3 


Severe lower extremity fracture* 


4 


Pelvic fracture 


4 


Spinal cord injury 


4 


Age (yr) 




40-59 


2 


60-74 


3 


>75 


4 



'Factors found to be significantly associated with deep 
venous thrombosis/pulmonary embolism on subsequent 
prospective analysis. 
AIS, Abbreviated Injury Severity Score. 
From Greenfield LJ, Proctor MC, Rodriquez JL, et al: Post- 
traumatic thromboembolic prophylaxis. J Trauma 
1997;42:100-103. 



(seven total) enrolled with this injury. All other 
studies have included patients with spinal cord 
injuries as among the highest risk patients, 
with rates of DVT approaching 80% and PE 
rates at 5%. In fact, PE is one of the most 
common causes of death following spinal cord 
injury. 



PROPHYLACTIC MEASURES 



Definitive randomized controlled clinical 
studies on prophylactic measures in trauma 
patients with multiple injuries do not exist. 
Unlike other surgical patients with isolated 
disease (e.g., hip-replacement patients and 



ch25.qxd 4/16/04 3:22PM Page 528 



528 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



colectomy patients), the injured patients are 
a heterogeneous group and difficult to "cat- 
egorize." They can have isolated injuries or 
any combination of injuries, making stratifi- 
cation extremely difficult. Additionally, many 
patients are excluded from one type of pro- 
phylactic measure or another by the very 
nature of their injuries. For example, bilat- 
eral leg compression devices cannot be used 
with external fixators, and some head-injured 
patients cannot receive anticoagulants. Con- 
sidering all of these factors, a large multicenter 
trial with thousands of patients and defined 
endpoints would be needed to definitively 
answer the questions of which trauma patients 
need prophylaxis and which prophylactic 
measures are effective for a given combina- 
tion of traumatic injuries. To date, no funding 
source for this important study has been iden- 
tified. However, several smaller prospective 
studies on injured patients have been 
attempted, and recommendations can be 
made based on the best available data. 

Prophylactic measures can generally be 
divided into two categories: mechanical and 
pharmacologic. Mechanical measures are 
aimed at reducing stasis, whereas drug therapy 
attempts to alter some part of the extrinsic 
clotting system. An extreme example of a 
mechanical measure is a "prophylactic" vena 
cava filter (VCF) , placed before the develop- 
ment of PE/DVT in a high-risk patient. Each 
of these methods is described in the follow- 
ing sections. 



Mechanical Prophylactic 
Devices 

The mechanical devices, which vary from a 
simple elastic stocking to a full-length sequen- 
tial compression sleeve, are attractive because 
of their safety. Few if any complications can 
be attributed to the use of these devices if they 
are properly fit according to the directions 
supplied by the manufacturer for each patient 
and used appropriately. The only real "com- 
plication" is the lack of compliance in patients 
who are awake enough to remove the devices. 
Despite their widespread use, however, there 
are no level I trials demonstrating protection 



from DVT/PE in trauma patients using any 
type of mechanical device. Knudson and col- 
leagues (1991) demonstrated that the sequen- 
tial pneumatic compression device (SCD) was 
more effective than no prophylaxis in head- 
injured patients, but not in trauma patients 
with other injuries. In another study, which 
included trauma patients without orthopedic 
injuries, 62 were randomized to wear calf-thigh 
sequential pneumatic compression and 62 
wore plantar compression devices ("foot 
pumps") only. DVT developed in 21% of the 
patients wearing the plantar device and in 
6.5% of those wearing the calf-thigh device 
(P=.009). Studies by Ginzburg and col- 
leagues (2001) and Knudson and colleagues 
(1996) have demonstrated clearly that com- 
pression devices are less effective than 
low-molecular-weight heparin (LMWH) in 
preventing thromboembolic complications 
after trauma. No data exist on the use of 
mechanical devices combined with anticoag- 
ulant therapy in patients with multiple injuries, 
and there is no documented benefit in com- 
pressing only one leg or an arm, in hopes of 
stimulating fibrinolytic activity. Based on the 
current available data, trauma patients who 
are considered at risk for DVT/PE and who 
cannot safely be given an anticoagulant drug 
should receive bilateral whole leg pneumatic 
compression. Anything short of that should 
be considered inadequate protection. 



Unfractionated Heparin 

Of all the methods of prophylaxis, low-dose 
unfractionated heparin (LDUH, 5000 units 
given subcutaneously 2 hours before surgery 
and then every 12 hours for 7 days postoper- 
atively) has been the most widely studied 
and the most effective method of preventing 
thromboembolic complications in surgical 
patients. In 20 trials in which more than 8000 
general surgery patients were enrolled, LDUH 
reduced the incidence of leg DVT from 25% 
to 8% and consistently reduced the incidence 
of/atoZPE by 50%. Unfortunately, in patients 
undergoing elective hip and knee surgery, in 
which the risk of proximal DVT is more than 
30% and fatal PE up to 6%, LDUH does not 



ch25.qxd 4/16/04 3:22PM Page 529 



25 • THROMBOEMBOLIC COMPLICATIONS 



529 



offer sufficient protection. Similarly, in trauma 
patients, LDUH does not appear to be any 
more effective than no prophylaxis, and its 
use in trauma patients as the sole method of 
protection should be discouraged. 



Low-Molecular -Weight Heparin 

LMWHs are fragments of unfractionated 
heparin, induced by a controlled enzymatic 
or chemical depolymerization process that 
yields chains of glycosaminoglycans with a 
mean molecular weight of around 5000. 
These shorted chains retain their anticoagu- 
lant activity by their ability to interact with 
antithrombin but have relatively less activity 
against thrombin and platelets. LMWH has a 
more predictable anticoagulant response than 
LDUH because of increased bioavailability, 
longer half-life, and dose-independent clear- 
ance. Thus, LMWH results in improved anti- 
coagulant activi ty while causing less bleeding. 
In hip and knee replacement surgery, LMWH 
has been demonstrated to be highly effec- 
tive in preventing DVT/PE even when given 
postoperatively. 

To date, only the LMWH enoxaparin 
(Lovenox) has been studied in trauma 
patients. However, two large prospective 
studies have documented the effectiveness of 
the LMWH enoxaparin in preventing post- 
traumatic thromboembolism. Geerts and col- 
leagues (1994) compared LDUH with LMWH 
(30 mg given subcutaneously every 1 2 hours) , 
both started within 36 hours after injury, in 
344 major trauma patients without frank 
intracranial bleeding. Bilateral contrast venog- 
raphy was performed between postinjury days 
10 and 14. The proximal DVT rate was 15% 
with LDUH and 6% with LMWH (risk reduc- 
tion with LMWH of 58%, P= .01) . The overall 
rate of major bleeding was less than 2% with 
no significant differences between the groups, 
thus demonstrating both the efficacy and the 
safety of LMWH in trauma patients. The study 
by Knudson and colleagues (1996) included 
372 patients with multiple trauma and com- 
pared LMWH to mechanical compression. 
Patients were followed with serial duplex ultra- 
sound examinations. Of the 120 patients who 



were randomized to receive LMWH, only 1 
developed DVT by ultrasound (0.8%). In 
the mechanical compression group (199 
patients), the incidence of DVT was 2.5%. 
Only one patient had a major bleeding com- 
plication with LMWH. Although LMWH was 
withheld in patients with injuries to the spleen 
and/or liver who were being managed without 
operation, recent data suggest that LMWH can 
be given safely in these situations, without 
inducing bleeding. In patients with major 
head injury and evidence of bleeding on a 
head computed tomographic (CT) scan, 
LMWH is generally withheld until the injury 
has been demonstrated to be stable. For now 
then, LMWH is considered the most effective 
form of prophylaxis against DVT/PE in 
trauma patients and it should be initiated once 
bleeding is under control and early postin- 
jury coagulopathy has been corrected. 

Prophylactic Vena Cava Filters 

The effectiveness of a VCF in the prevention 
of PE in patients with proximal DVT has been 
well established. Traditionally, these filters 
have been placed in patients with acute prox- 
imal DVT or a recent PE who have a con- 
traindication to anticoagulation. Filters can 
be placed percutaneously with relative ease 
and have long-term patency rates of more than 
95%. Some trauma surgeons have advocated 
the prophylactic placement of a VCF in high- 
risk trauma patients, especially in those 
patients who have relative contraindications 
to anticoagulation. A recently described tech- 
nique of placing Inferior Vena Cava (IVC) 
filters at the bedside using ultrasound guid- 
ance makes it even easier to advocate for an 
aggressive approach in critically injured 
patients. 

The problems associated with the use of 
IVC filters in trauma patients include the 
following: 

1. Recurrent PE: Despite the presence of a 
filter, recurrent PE occurs in 3% of trauma 
patients. This complication may result 
from filter tilt or strut malposition and has 
been fatal in a few reported injured 
patients. 



ch25.qxd 4/16/04 3:22PM Page 530 



530 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



2. -DVT: An IVC filter does nothing to prevent 
DVT and, in fact, may promote thrombo- 
sis. In studies reported by Rodriquez and 
colleagues (1996), 10% of injured patients 
who had a prophylactic filter demon- 
strated caval thrombosis and 50% of these 
patients had long-term lower extremity 
edema. 

3. Permanence: Because all currently mar- 
keted VCFs are designed to be permanently 
implanted, patients are at risk for compli- 
cations for their lifetime. In addition to 
filter-associated thromboembolic events 
and filter migration, complicationswith vas- 
cular access procedures including trapping 
of the guidewire in the filter have been 
described. 

4. Timing: We and others have documented 
PE as early as 12 to 24 hours postinjury. In 
the study by Owings and colleagues (1997) , 
4 (6%) of 63 patients had embolism within 
1 day of their trauma. It would be highly 
unlikely that a prophylactic filter would be 
placed within such a narrow time frame in 
patients with multiple trauma who are in 
need of various other procedures to address 
their injuries. 

No data support the routine use of pro- 
phylactic VCFs in high-risk trauma patients. 
Their use should be restricted to the occa- 
sional injured patients who are at prolonged 
exceedingly high risk for PE, as described in 
Table 25-1, and in whom no other prophy- 
lactic measures can be used. 



DIAGNOSIS AND TREATMENT 
OF POST-TRAUMATIC 
THROMBOEMBOLIC 
COMPLICATIONS 



Most venous thrombi are clinically silent, pre- 
sumably because they do not totally obstruct 
the vein and because of the existence of col- 
lateral circulation. Even when symptoms do 
develop, they are nonspecific and may include 
pain, swelling, or fever. In trauma patients, 
these symptoms may be totally overlooked or 
attributed to bone or soft tissue injury. In most 



trauma patients, DVT is clinically occult. The 
symptoms associated with PE depend on the 
quantity of the embolus involved and the car- 
diopulmonary status of the patient. Signs and 
symptoms may include chest pain, dyspnea, 
tachypnea, anxiety, cyanosis, and fever. Arte- 
rial blood gas analyses may reveal hypoxia and 
an acute decrease in carbon dioxide. Unfor- 
tunately, the first sign of PE in many injured 
patients is sudden death. 

For many years, the standard diagnostic test 
for DVT was ascending phlebography (venog- 
raphy) . Venography can reliably detect both 
proximal (pelvic, thigh, popliteal) and distal 
(calf) thrombosis. Side effects include aller- 
gic reactions to the contrast, renal toxicity, and 
a 2% to 3% incidence of contrast-induced 
DVT. Additionally, venography cannot easily 
be repeated and is thus impractical for sur- 
veillance in high-risk patients. Thrombi have 
also been visualized incidentally during com- 
puterized scanning of the abdomen or pelvis 
performed for other indications. Recently, 
duplex ultrasound (color flow Doppler [CFD] 
imaging) has become the method of choice 
for detecting DVT in many centers. CFD 
imaging allows information on flow to be over- 
laid onto the real time two-dimensional image 
of the vein. Venous sonography is 90% sensi- 
tive (100% specific) in detecting proximal 
DVT in symptomatic patients, but the sensi- 
tivity drops significantly in asymptomatic 
patients, most likely related to the small size 
of the clot in this latter group of patients. The 
most reliable sign of DVT is lack of com- 
pressibility of the vein on ultrasound imaging 
(Fig. 25-1). Other signs of acute DVT include 
the loss of flow augmentation with the Val- 
salva maneuver or with muscle contraction and 
the presence of a homogenous thrombus with 
low echogenicity. The involved vein is dis- 
tended and noncompressible, with decreased 
or absent flow and no collateral channels. 
The limitations of color duplex sonography 
include the difficulty in imaging pelvic veins 
and the fact that the quality of the examina- 
tion is highly dependent on the experience 
and expertise of the sonographer. 

We have performed venous ultrasound 
examinations on hundreds of trauma patients 
during our research studies investigating 
thromboembolism prophylaxis. DVT detected 



ch25.qxd 4/16/04 3:22 PM Page 531 



25 • THROMBOEMBOLIC COMPLICATIONS 



531 




■ FIGURE 25-1 

Appearance of a clot in the iliac vein by color 
flow duplex sonography. ■ 



via ultrasound was the main outcome measure 
in three prospective studies on trauma patients 
at our center, and we have been impressed 
with our ability to detect clinically silent DVT 
with surveillance scanning. Our results have 
been confirmed by other investigators, in 
which color flow imaging surveillance in 
high-risk trauma patients revealed an overall 
DVT incidence of between 10% and 18%. 
Interestingly, up to 30% of the cases of DVT 
involved the upper extremity, an area not rou- 
tinely studied with venography. Some authors 
have argued that there is no need to detect 
clinically occult DVT, and that the number of 
cases detected does not justify the expense of 
surveillance. Mostwould agree, however, that 
given the incomplete protection offered by 
available methods of prophylaxis, some type 
of surveillance is warranted in extremely high- 
risk trauma patients (see Fig. 24-3) . Treatment 
of occult DVT is most likely the key factor 
contributing to the low incidence in of PE 
in our center and in other centers that 
perform venous imaging liberally in trauma 
patients. 

Pulmonary angiography remains the gold 
standard for the diagnosis of pulmonary 
emboli. Ventilatory-perfusion scans have not 
been useful in trauma patients, primarily 
because few trauma patients at risk for PE have 
anormal chestx-rayfilm. Additionally, the risks 
associated with full-dose anticoagulation in 
injured patients are significant, and the diag- 
nosis must be secured by the most sensitive 
and specific study. Pulmonary angiography 



requires transportation to the radiology suite, 
and there is the potential for allergic reactions, 
contrast-induced nephropathy, or vascular 
injuries during the procedure. Combining 
noninvasive diagnostic tests has been advo- 
cated for critically ill patients who cannot 
be moved out of the intensive care unit. 
For example, the combination of a positive 
ultrasound of the lower extremities and a 
transthoracic echocardiogram showing right 
ventricular hypokinesis is pathognomonic for 
PE. Whole-blood D-dimer levels are always ele- 
vated in patients with thrombosis, but this test 
is not very specific. Still, the combination of 
a normal D-dimer level and a negative venous 
ultrasound examination virtually rules out PE. 
Recently, calculation of the late pulmonary 
dead space fraction, calculated from the C0 2 
expirogram, was found to be valuable as a 
bedside screening technique for detection 
ofPE. 

Spiral CT of the chest with contrast is 
another excellent screening tool for PE. This 
study is most sensitive (90%) in detecting 
emboli that are located in the proximal pul- 
monary vascular tree. In the presence of a pos- 
itive CT scan, anticoagulant therapy should 
be initiated immediately (Fig. 25-2). If more 
distal emboli are suspected and the initial CT 
scan is negative, pulmonary angiography 
should be performed. In a recent analysis of 
15 combinations of diagnostic tests (spiral CT, 
lower limb ultrasonography, ventilation- 
perfusion scintigraphy, pulmonary arteri- 
ography, and D-dimer plasma levels), the 
combination of spiral CT and lower limb 
ultrasonography was found to be the least 
morbid and the most cost-effective combina- 
tion of studies per life saved. 

Heparin remains the first line of treatment 
for both DVT and PE. In patients with DVT, 
a bolus of heparin (5000 units) is given imme- 
diately, followed by a continuous drip of 
18U/kg per hour. The heparin dose is 
adjusted to keep the partial thromboplastin 
time twice normal or between 60 and 80 
seconds. For PE, especially when accompanied 
by hypoxia, a bolus of 10,000 units of heparin 
is initiated, followed by a continuous drip. 
Alternatively, a fully anticoagulating dose of 
enoxaparin (lmg/kg given subcutaneously 
twice daily) has been found to be effective 



ch25.qxd 4/16/04 3:23PM Page 532 



532 



V • SPECIAL PROBLEMS AND COMPLICATIONS 




■ FIGURE 25-2 

A, Spiral computed tomographic scan of the chest demonstrating pulmonary emboli in the right 
pulmonary artery. B, Lung window on the same patient. Note the area of infarction in the right lower 
lobe. ■ 



treatment for both DVT and PE. The advan- 
tage of enoxaparin is the ability to treat DVT 
as an outpatient with twice-daily injections 
performed by the patient. We have used this 
approach in patients with venous injuries who 
are anticoagulated for a short period (1 to 
2 months) . For documented cases of DVT/PE 
however, we advocate anticoagulation for 
6 months, and this is best accomplished by 
converting the patient from heparin (either 
unfractionated or LMWH) to Coumadin 
before discharge from the hospital. In patients 
with documented DVT and/or PE who cannot 
receive full-dose anticoagulation because of 
the nature of their injuries, a VCF should be 
placed. On rare occasions, trauma patients 
with life-threatening PE and persistent hypoxia 
may be candidates for transvenous catheter 
embolectomy. 



OUTCOMES RESEARCH IN 
PREVENTION OF 
THROMBOEMBOLISM IN 
TRAUMA PATIENTS 

As mentioned, no large-scale, prospective, ran- 
domized trials that definitively establish the 
safety and effectiveness of any prophylactic 



measures have been performed in trauma 
patients. Several investigators have attempted 
to review all of the data available to assist in 
the development of outcome-driven guide- 
lines for DVT/PE prophylaxis in injured 
patients. Brasel, Borgstrom, and Weigelt 
(1997) used decision-tree analysis to compare 
three approaches for PE prevention in trauma 
patients: no intervention, surveillance ultra- 
sound, and VCF. The probabilities in each 
subtree were taken from available published 
data. Their findings support the use of sur- 
veillance duplex ultrasound examinations, 
with a cost/PE prevented of $46,300 compared 
to $93,700 per PE prevented with the use of 
a prophylactic VCF. However, these costs were 
based on the assumption that the filter was 
placed in the radiology suite and that the 
patient was hospitalized for at least 2 weeks. 
A VCF may be more cost-effective in patients 
requiring prolonged hospitalization. 

Three other groups have attempted to 
develop cost-effective guidelines for DVT 
prevention in trauma patients by employing 
Cochrane-type principles in literature reviews. 
These groups include the Eastern Association 
for the Surgery of Trauma (EAST) , the Amer- 
ican College of Chest Physicians (ACCP) , and 
the Southern California Evidence-Based Prac- 
tice Center (SCEPC). The recommendations 
developed by each of these groups are 



ch25.qxd 4/16/04 3:23PM Page 533 



25 • THROMBOEMBOLIC COMPLICATIONS 



533 



TABLE 25-2 

SUMMARY OF RECOMMENDATIONS FOR THROMBOEMBOLIC PROPHYLAXIS FOR 
THREE INDEPENDENT REVIEWS 



Group 


LDUH 


SCD 


LMWH 


Foot Pump 


IVC Filter 


Ultrasound 


EAST 
ACCP 
SCEPC 


N/R 

N/R 

Equal to LMWH 


Level II 
Level II 
R 


Level II 
Level 1 

Equal to LDUH 


Level III 

N/A 

N/A 


Level II 
Level III 
No data 


Level II 
Level III 
N/A 



ACCP, American College of Chest Physicians; EAST, Eastern Association for the Surgery of Trauma; IVC, Inferior Vena Cava; 
LDUH, low-dose unfractionated heparin; LMWH, low-molecular-weight heparin; N/R, not recommended; R, recommend use; 
SCD, sequential pneumatic compression device; SCEPC, Southern California Evidence-Based Practice Center. 



summarized in Table 25-2. The EAST guide- 
lines recommend LMWH for patients with one 
ofthe following injury patterns: (1) pelvicfrac- 
tures requiring operative fixation or pro- 
longed bed rest (>5 days) ; (2) complex lower 
extremity fractures (defined as open fractures 
or multiple fractures in one extremity) requir- 
ing operative fixation or prolonged bed rest 
(>5 days) ; and (3) spinal cord injury with com- 
plete or incomplete motor paralysis. SCDs 
were recommended for high-risk patients with 
head injuries, spinal cord injuries, or pelvis 
or hip fractures. Foot pumps were given only 
a level III recommendation, due to insufficient 
data. Prophylactic VCFs in trauma patients 
without established PE or DVT were recom- 
mended only in patients who (1) could not 
receive anticoagulation because of increased 
bleeding risk and (2) who had one ofthe fol- 
lowing injuries: 

• Severe closed head injury (Glasgow Coma 
Scale score <8) 

• Incomplete spinal cord injury with para- 
plegia or quadriplegia 

• Complex pelvic fracture with associated 
long bone fractures 

• Multiple long bone fractures 

These recommendations are based on class 
II/III data. The ACCP recommendations are 
similar to EAST. However, the SCEPC inves- 
tigators concluded that there was no evidence 
that any existing method of prophylaxis was 
clearly superior to other methods or even to 
no prophylaxis. They concluded that LMWH 
was not superior to LDUH and they could 



make no conclusions on the role of IVC filters, 
based on their review of the literature. 
Although I do not agree with the conclusions 
drawn by the SCEPC investigators, I do agree 
that a large, multicenter, prospective trial on 
prophylactic methods in trauma patients is 
long overdue. 



CURRENT RECOMMENDATIONS 
AND FUTURE DIRECTIONS 



At the San Francisco General Hospital, we have 
had a long-standing interest in the prevention 
of thromboembolic complications in trauma 
patients. Before implementing a standardized 
protocol for DVT/PE prevention, our inci- 
dence of DVT was 10% (established by sur- 
veillance ultrasound scanning). Since 1992, 
we have prospectively followed more than 
12,000 patients for the clinical development 
of DVT or PE and used a standardized man- 
agement algorithm for DVT prophylaxis (Fig. 
25-3) . Our overall incidence of DVT is 0.003% 
and the PE rate is 0.002%. The death rate for 
PE, however, was 17%. In a detailed audit of 
our patients, we found that 20% ofthe time, 
trauma clinical case managers had to inter- 
vene to ensure that the house staff ordered 
the proper prophylactic measure. All cases of 
DVT/PE are thoroughly reviewed to assess 
whether the protocol was implemented and 
followed correctly. In our experience, in most 
patients in whom thromboembolic compli- 
cations occurred, prophylactic measures were 
inappropriately withheld early in the patients' 
postinjury course. Only one patient with PE 



ch25.qxd 4/16/04 3:23PM Page 534 



534 



V • SPECIAL PROBLEMS AND COMPLICATIONS 



■ FIGURE 25-3 

Algorithm for deep venous 
thrombosis prophylaxis in the 
trauma patient from the San 
Francisco General Hospital. ■ 





Trauma Patient Requiring Hospital Admission 






V 






Does patient have any risk factors for DVT? 






No 


V 


Yes 


" 




> r 


No indication for specific DVT 
prophylaxis 




Does the patient have 
contraindication for Heparin? 


No 






" 


Yes 


" 




v 


Begin low-molecular-weight 
Heparin within 36 hours 




Does the patient have 

contraindication for 

(lower extremity) intermittent 

pneumatic compression? 






No 




" 


Yes 


" 




V 


Begin intermittent pneumatic 
compression (IPC) 




Is the patient HIGH RISK 

for DVT? 

(multiple risk factors) 






No 




" 


Yes 


' 


r 




V 


Apply bilateral foot pumps. 
Consider serial monitoring 
using color duplex scans. 




Is the DVT risk permanent 

or long term? 

(e.g., spinal cord injury) 


No 






' 


f 


Yes 


■ 


t 




i 


r 


Apply bilateral foot pumps. 

Monitor closely using serial 

color duplex scans. 




Consider placement of an 

IVC filter and/or serial 

color duplex scans. 



fell outside of our established risk factors and 
thus did not receive prophylaxis. We encour- 
age other institutions to prospectively study 
the guidelines in place at their institution, so 
that high-quality, evidence-based outcome 
data will be available for analysis. 

Future investigations will include new and 
potentially more effective drugs that target not 
only the various steps in the coagulation 
cascade, but perhaps even the venous wall 
itself. One such agent, Aristra/Xantidar, a syn- 
thetic pentasaccharide that acts as an indirect 
inhibitor of factor Xa, has recently been shown 
to be more effective than LMWH in the 



prevention of DVT following orthopedic 
surgery. In the area of mechanical prophylaxis, 
a prototype removable IVC filter has been 
developed. This Tulip filter can be used as 
either a permanent or a temporary device and 
has been removed from a small number of 
patients as early as 5 days after insertion 
without injuring the vena cava. A temporary 
filter would obviously be more attractive in 
young trauma patients in whom the risk of 
developing thromboembolic complications is 
also temporary. Armed with an appreciation 
for and a better understanding of throm- 
boembolism in trauma patients, and coupled 



ch25.qxd 4/16/04 3:23PM Page 535 



25 • THROMBOEMBOLIC COMPLICATIONS 



535 



with effective and safe prophylactic mea- 
sures, we should be able to offer our future 
trauma patients protection from both the mor- 
bidity of DVT and the risk of a premature death 
from PE. 



REFERENCES 

Anderson JT, OwingsJT, Goodnight JE: Bedside 
noninvasive detection of acute pulmonary 
embolism in critically ill surgical patients. Arch 
Surg 1999;134:869-875. 

Brasel KJ, Borgstrom DC, Weigelt JA: Cost- 
effective prevention of pulmonary embolus in 
high-risk trauma patients. J Trauma 1 997;42:456- 
462. 

Burch JN, Richardson RJ, Martin RR, Mattox KL: 
Penetrating iliac vascular injuries: recent expe- 
rience with 233 consecutive patients. J Trauma 
1990;30:1450-1990. 

Claget GP, Anderson FA, Geerts W, Heit JA, 
Rnudson MM et al: Prevention of venous throm- 
boembolism. Chest 1998;114:531S-560S. 

Coon WW: Risk factors in pulmonary embolism. 
Surg Gynecol Obstet 1976;143:385-390. 

Elliott CG, Dudney TMN, Egger M, e t al: Calf-thigh 
sequential pneumatic compression compared 
with plantar venous pneumatic compression to 
prevent deep-vein thrombosis after non-lower 
extremity trauma. J Trauma 1999;47:25-32. 

Freeark RJ, Boswick J, Fardin R: Posttraumatic 
venous thrombosis. Arch Surg 1967;95:567-573. 

Gearhart MM, Luchette FA, Proctor MA, et al: The 
risk assessment profile score identified trauma 
patients at risk for deep vein thrombosis. Surgery 
2000;128:631-640. 

Geerts WH, Code KI, Jay RM, et al: A prospective 
study of venous thromboembolism after major 
trauma. N EnglJ Med 1994;331:1601-1606. 

Merli G, Geerts W, Ginzburg E, et al: for the Spinal 
Cord Injury Thromboprophylaxis Investigators: 
Prevention of venous thromboembolism in the 
acute treatment phase after spinal cord injury: 
A randomized multicenter trial comparing 
low-dose heparin plus intermittent pneumatic 
compression with Enoxaparin. J Trauma 2003; 
54:1116-1126. 

Knudson MM, Lewis FR, Clinton A et al: Preven- 
tion of venous thromboembolism in trauma 
patients. J Trauma 1994;37:480-487. 

Knudson MM, Morabito D, Paiement GD, Shack- 
leford S: Use of low-molecular-weight heparin 



in preventing thromboembolism in trauma 
patients. J Trauma 1996;41 :446-459. 

McCartney JS: Pulmonary embolism following 
trauma. Am J Pathol 1934;10:709-710. 

Meredith JW, Young JS, O'Neil EA, et al: Femoral 
catheters and deep venous thrombosis: a prospec- 
tive evaluation with venous duplex sonography. 
J Trauma 1993;35:187. 

Millward SF, Bhargava A, Aquino J, et al: Gunther 
Tulip filter: preliminary clinical experience with 
retrieval. JVIR 2000;1 1 :75-82. 

OwingsJT, Kraut E, Battistella F et al: Timing of 
occurrence of pulmonary embolism in trauma 
patients. Arch Surg 1997;132;862-867. 

Roergs FB, Cipolli MD, Velmahos GC, et al: Prac- 
tice guidelines for the prevention of venous 
thromboembolism in trauma patients: The EAST 
Practice Management Guidelines Work Group. 
J Trauma 2002;53:142-164. 

Rodriquez JL, Lopez JM, Proctor MC, et al: Early 
placement of prophylactic vena caval filters in 
injured patients at high risk for pulmonary 
embolism. J Trauma 1996;40:797-804. 

Rogers FB, Cipolle MD, Velmahos G, et al: Prac- 
tice management guidelines for the preven- 
tion of venous thromboembolism in trauma 
patients: The EAST practice management 
guidelines work group. J Trauma 2002;53(1): 
142-164. 

Rogers FB, Shackford ST, Ricci MA, et al: Routine 
prophylactic vena cava filter insertion in severely 
injured trauma patients decreases the incidence 
of pulmonary embolism. J Am Coll Surg 
1995;180:641-647. 

Sevitt S, Gallagher N: Venous thrombosis and pul- 
monary embolism: a clinico-pathological study 
in injured and burned patients. Br J Surg 
1960;48:475-488. 

Sue LP, Davis JW, Parks SN: Iliofemoral venous 
injuries: an indication for prophylactic caval filter 
placement. J Trauma 1995;39:693-695. 

Turpie AGG, Gallus AS, HoekJA: A synthetic pen- 
tasaccharide for the prevention of deep vein 
thrombosis after total hip replacement. N Engl 
J Med 2001;344:619-625. 

Van Erkel AR, van Rossum AB, Bloem JL, et al: Spiral 
CT angiography for suspected pulmonary 
embolism: a cost-effectiveness analysis. Radiol- 
ogy 1996;201:29-36. 

Velmahos GC, Kern JK, Chan LS et al: Prevention 
of venous thromboembolism after injury: an 
evidence-based report. J Trauma 2000:49:132- 
144. 



RE.qxd 4/16/04 3:19 PM Page 539 




Rich's Historic Collection of 
Vascular References 






Aaland M, Bryan FC, Sherman R: Two dimensional echocardiogram in hemo- 

dynamically stable victims of penetrating precordial trauma. Am Surg 

1994;60:412-415. 
Aarabi B, McQueen JD: Traumatic internal carotid occlusion at the base of the 

skull. Surg Neurol 1978;10:233-236. 
Abbe R: The surgery of the hand. N YMed J 1894. 
Abbott JA, Cousineau M, Cheitlin M, et al: Late sequelae of penetrating cardiac 

wounds. J Thorac Cardiovasc Surg 1978;75:510-518. 
Abbott WM, Darling RC: Axillary artery aneurysms secondary to crutch trauma. 

Am J Surg 1973;125:515-520. 
Abdo F, Massad M, Slim M, et al: Wandering intravascular missiles: Report of 

five cases from the Lebanon war. Surgery 1988;103:376-380. 
Abdul-Razek MS, Mnaymneh W, Yacoubian HD: Acute injuries of peripheral 

arteries with associated bone and soft tissue injury. J Trauma 1973;13:907- 

910. 
Abouljoud M: Arterial injuries of the thoracic outlet: A ten-year review. Am 

Surgeon 1993;59:590-595. 
Accola KD, Feliciano DV, Mattox KL, et al: Management of injuries to the supe- 
rior mesenteric artery. J Trauma 1986;26:313-317. 
Accola KD, Feliciano DV, Mattox KL, et al: Management of injuries to the 

suprarenal aorta. Am J Surg 1987;154:613-618. 
Adar R, Bass A, Walden R: Five years' experience in general and vascular surgery 

in a university hospital. Ann Surg 1982;196:725-729. 
Adar R, Nerubay J, Katznelson A: Management of acute vascular injuries. J 

Cardiovasc Surg 1970;11:435-439. 
Adar R, Schramek A, Khodadadi J, et al: Arterial combat injuries of the upper 

extremity. J Trauma 1980;20:297-302. 
Adebonojo SA: Management of chest trauma: A review. West Afr J Med 1993; 

12:122-132. 
Adinolfi MF, Hardin WD, O'Connell RC, Kerstein MD: Amputations after vas- 
cular trauma in civilians. South Med J 1983;76:1241-1248. 
Adkins RB Jr, Bitseff ELJr, Meacham PW: Abdominal vascular injuries. South 

MedJ 1985;78:1152-1160. 
Adkins RB, WhiteneckJM, Woltering EA: Penetrating chest wall and thoracic 

injuries. Am Surg 1985;51:140-148. 

539 



RE.qxd 4/16/04 3:19 PM Page 540 



540 VI • THE RICH REFERENCE COLLECTION 

Agee CK, Metzler MH, Churchill RJ, Mitchell FL: Computed tomographic eval- 
uation to exclude traumatic aortic disruption. J Trauma 1992;33:876-881. 
Agnew D II: The Principles and Practice of Surgery. Philadelphia, JB Lippin- 

cott Co, 1978. 
Agrwal N, Shah PM, Clauss RH, et al: Experience with 115 civilian venous injuries. 

J Trauma 1982;22:827-832. 
Aitken RJ, Matley PJ, Immelman EJ: Lower limb vein trauma: A long-term 

clinical and physiological assessment. Br J Surg 1989;76:585-588. 
Akins CW, Buckley MJ, Daggett W, et al: Acute traumatic disruption of the 

thoracic aorta: A ten-year experience. Ann Thorac Surg 1981;31:305-309. 
Akins EW, Carmichael MJ, HillJA, Mancuso AA: Preoperative evaluation of he 

thoracic aorta using MRI and angiography. Ann Thorac Surg 1987;44:499- 

507. 
Al Zharani HA: False aneurysm of the abdominal aorta after blunt trauma. Eur 

J Vase Surg 1991;5:685-687. 
Alberty RE, Goodfried G, Boyden AM: Popliteal artery injury with fracture dis- 
location of the knee. Am J Surg 1981;142:36-40. 
Albo D Jr, Christensen C, Rasmussen BL: Massive liver trauma involving the 

suprarenal vena cava. Am J Surg 1969;118:960-963. 
Albrik MH, Rodriguez E, England GJ, et al: Importance of designated thoracic 

trauma surgeons in the management of traumatic aortic transection. South 

MedJ 1994;87:497-501. 
Alexander EL, Szentpetery S, Greenfield L: Posttraumatic thoracic aortic 

aneurysm associated with a diaphragmatic hernia. Ann Thorac Surg 

1985;40:195-198. 
Alexander JJ, Imbembo AL: Aorta-vena cava fistula. Surgery 1989;105:1-12. 
Ali IS, Fitzgerald PG, Gillis DA, Lau HYC: Bunt traumatic disruption of the 

thoracic aorta: A rare injury in children 10-year-old .J Pediatr Surg 1992;27: 

1281-1284. 
Ali J, Vanderby B, Purcell C: The effect of the pneumatic antishock garment 

(PASG) on hemodynamics, hemorrhage, and survival in penetrating tho- 
racic aortic injury. J Trauma 1991;31:846-851. 
Allen MJ, Stirling AJ, Crawshaw CW, Barnes MR: Intracompartmental pressure 

monitor of leg injuries: An aid to management. J Bone Joint Surg 1985;67- 

B:53-57. 
Allen MJ, White GH, Harris JP, et al: Iatrogenic vascular trauma associated with 

intra-aortic balloon pumping: Identification of risk factors. Am Surg 

1993;59:813-817. 
Allen RE, Blaisdell FW: Injuries to the inferior vena cava. Surg Clin North Am 

1972;52:699-710. 
Allen TR, FranklinJD, Withers EH, DavisJL: Extremity salvage utilizing microvas- 
cular free tissue transfer. Surgery 1981;90:1047-1054. 
Allen TW, Reul GJ, Morton JR, Beall AC Jr: Surgical management of aortic 

trauma. J Trauma 1972;12:862-868. 
AlpertJ, Bhaktan EK, Gielchinsky I: Vascular complications of intra-aortic balloon 

counterpulsation. Arch Surg 1976;111:1190-1195. 
AlpertJ, O'DonnellJA, Parsonnet V, et al: Clinically recognized limb ischemia 

in the neonate after umbilical artery catheterization. AmJ Surg 1980;140:413- 

418. 
AlpertJ, Parsonnet V, Goldenkranz RJ, et al: Limb ischemia during intra-aortic 

balloon pumping: Indication for femoro-femoral crossover graft. J Thorac 

Cardiovasc Surg 1980;79:729-734. 



RE.qxd 4/16/04 3:19 PM Page 541 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 541 

Alsofrom DJ, Marcus NH, Seigel RS, et al: Shotgun pellet embolization from 
the chest to the middle cerebral arteries. J Trauma 1982;22:155-157. 

Althaus SJ, Keskey TS, Harker CP: Percutaneous placement of self expanding 
stent for acute traumatic arterial injury. J Trauma 1996;41:145-148. 

Amato JJ, Billy LJ, Gruber RP, et al: Vascular injuries: An experimental study 
of high and low velocity missile wounds. Arch Surg 1970;101:167-174. 

Amato JJ, Billy LJ, Gruber RP, Rich NML: Temporary cavitation in high veloc- 
ity pulmonary missile injury. Ann Thor Surg 1974;18:565-570. 

Amato JJ, Billy LJ, Lawson NS, Rich NM: High velocity missile injury. Am J Surg 
1974;127:454-459. 

Amato JJ, Rich NM: Temporary cavity effects in blood vessel injury by high 
velocity missiles. J Cardiovasc Surg 1972;13:147-155. 

Amato JJ, Rich NM, Billy LJ, et al: High velocity arterial injury: A study of the 
mechanism of injury. J Trauma 1971;11:412-416. 

Amato JJ, Syracuse D, Seaver PRJr, Rich NM: Bone as a secondary missile: An 
experimental study in the fragmenting of bone by high missiles. J Trauma 
1989;29:609-612. 

Amato JJ, Vanecko RM, Yao JST, Weinberg M Jr: Emergency approach to 
the subclavian and innominate vessels. Ann Thorac Surg 1969;8:537-541. 

Amin A, Alexander JB, O'Malley KF, Doolin E. Blunt abdominal aortic trauma 
in children: Case report. J Trauma 1993;34:293-297. 

Amine ARC, Sugar O: Repair of severed brachial plexus: A plea to emergency 
room physicians. JAMA 1976;235:1039. 

Ammons MA, Moore EE, Moore FA, Hopeman AR: Intra-aortic balloon pump 
for combined myocardial contusion and thoracic aortic rupture. J Trauma 
1990;30:1606-1608. 

Anamah NV, Konstam PG: Traumatic dissecting aneurysm of the abdominal 
aorta. Br J Surg 1965;52:981-982. 

Anastasia LF: Traumatic disruption of the thoracic aorta treated with external 
shunt. Am J Surg 1970;120:810-812. 

Anderson RJ, Hobson RW II, Lee BC, et al: Reduced dependency on arterio- 
graphy for penetrating extremity trauma: Influence of wound location and 
non-invasive vascular studies. J Trauma 1990;30:1059-1065. 

Anderson RJ, Hobson RW II, Padberg FT Jr: Reduced dependency on arteri- 
ography for penetrating extremity trauma (PET) : Influence of wound loca- 
tion and noninvasive studies. J Trauma 1989;29:1024. 

Anderson RJ, Hobson RW II, Padberg FT Jr, et al: Penetrating extremity trauma: 
Identification of patients at high-risk requiring arteriography. J Vase Surg 
1990;11:544-548. 

Andrade-Alegre R, Mon L: Subxiphoid pericardial window in the diagnosis of 
penetrating cardiac trauma. Ann Thorac Surg 1994;58:1139-1141. 

Anfossi A, Bertoglio C, Iurilli L, et al: Delayed development and rupture of an 
aortic aneurysm after closed abdominal trauma. J Cardiovasc Surg 1987; 
28:35-37. 

Annandale T: Traumatic popliteal arteriovenous aneurysm treated successfully 
by ligature of the popliteal artery and vein. Lancet 1875;1:568. 

Anthopoulos LP, Johnson JB, Spellman M: Arteriovenous fistula in multiple 
saccular arterial aneurysms of a finger, following childhood human bite. 
Angiology 1965;16:89. 

Anton GE, Hertzer NR, Beven EG, et al: Surgical management of popliteal 
aneurysms: Trends in presentation, treatment, and results from 1952 to 
1984. J Vase Surg 1986;3:125-134. 



RE.qxd 4/16/04 3:19 PM Page 542 



542 VI • THE RICH REFERENCE COLLECTION 



Antonovic R, RoschJ, Dotter CT: Complications of percutaneous transaxillary 
catheterization for arteriography and selective chemotherapy. Am J 
Roentgenol 1976;126:386-393. 

An tunes MJ: Acute traumatic rupture of the aorta: Repair by simple aortic cross- 
clamping. Ann Thorac Surg 1987;44:257-259. 

An tunes MJ, Fernandes LE, Oliveira JM: Ventricular septal defects and arteri- 
ovenous fistulas, with and without valvular lesions, resulting from pene- 
trating injury of the heart and aorta. J Thorac Cardiovasc Surg 1988; 
95:902-907. 

Antyllus. Oribasius4:52 (Daemberg Edition). Cited by: Olser. Lancet 1915;1:949. 

Appelbaum A, Karp RB, Kirklin JW: Surgical treatment for closed thoracic aortic 
injuries. J Thorac Cardiovasc Surg 1976;71:458-460. 

Applebaum R, Yellin AE, Weaver FA, et al: Role of routine arteriography in 
blunt lower-extremity trauma. Am J Surg 1990;160:221. 

Aprahamina C, Thompson BM, TowneJB, Darin JC: The effect of a paramedic 
system on mortality open intra-abdominal vascular trauma. J Trauma 
1983;23:687-690. 

Arajarvi E, Santavirta S: Chest injuries sustained in severe traffic accidents by 
seatbelt wearers. J Trauma 1989;29:37-41. 

Arajarvi E, Santavirta S, TolonenJ: Aortic ruptures in seat belt wearers. J Thorac 
Cardiovasc Surg 1989;98:355-361. 

Archambault R, Archambault FLA, Mizeres NJ: Rupture of the thoracoacromial 
artery and anterior dislocation of the shoulder. Am J Surg 1959;97:782- 
783. 

Armstrong K, Sfeir R, Rice J, Kerstein M: Popliteal vascular injuries and war: 
Are Beirut and New Orleans similar? J Trauma 1988;28:836-839. 

Arnon RG, Allen RG: Traumatic aortic aneurysm in a child. J Pediatr Surg 
1981;16:72-74. 

Arom KV, Richardson JD, Webb G, et al: Subxyphoid pericardial window in 
patients with suspected traumatic pericardial tamponade. Ann Thorac Surg 
1977;23:545-549. 

Aronstam EM, Gomez AC, O'Connell TJ, Geiger JP: Recent surgical and phar- 
macologic experience with acute dissecting and traumatic aneurysms. 
J Thorac Cardiovasc Surg 1970;59:231-238. 

Aronstam EM, Strader LD, Geiger JP, Gomez AC: Traumatic left ventricular 
aneurysms. J Thorac Cardiovasc Surg 1970;59:239-242. 

Artz T, Huffer JM: A major complication of the modified Bristow procedure 
for recurrent dislocation of the shoulder. J Bonejoint Surg 1972;54-A: 1293- 
1296. 

Asensio J, Hanpeter D, Demetriades D: The Futility of Liberal Utilization of 
Emergency Department Thoracotomy: Proceedings of the American 
Association for the Surgery of Trauma, 58th Annual Meeting. 1998, 
p210. 

AsensioJ, Ierardi R: Exsanguination: Evolving issues in emergency trauma. Emerg 
Care Q 1991;9:59-75. 

AsensioJ, Murray J, Demetriades D: Penetrating cardiac injuries: A prospective 
study of variables predicting outcomes. JACS 1998;186:24-34. 

Asensio JA: Exsanguination from penetrating injuries. Trauma Q 1989;16:1-25. 

Asensio JA, BerneJD, Chahwan S, et al: Traumatic injury to the superior mesen- 
teric artery. Am J Surg 1999;178:235-239. 

Asensio JA, BerneJD, Demetriades D: One hundred five penetrating cardiac 
injuries. J Trauma 1998;44:1073-1082. 



RE.qxd 4/16/04 3:19 PM Page 543 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 543 

Asensio JA, Britt LD, Borzotta A: Multi-institutional experience with the man- 
agement of superior mesenteric artery injuries. J Am Coll Surg 2001 ;193:354- 

366. 
Asensio JA, Chahwan S, Hanpeter D. Operative management and outcome of 

302 abdominal vascular injuries: AAST-OIS Correlates well with mortality. 

Am J Surg 2000;180:528-534. 
Asensio JA, Forno W, Gambaro E, et al: Abdominal vascular injuries: The trauma 

surgeon's challenge. Anales Chirurgiae Gynaecol 2000;89:71-78. 
Asensio JA, Lejarraga M: Abdominal vascular injury. In Demetiades D, Asensio 

JA (eds). Trauma Handbook. Austin, Tex, Landes Bioscience, 2000, pp 

356-362. 
Asensio JA, Stewart BM, MurrayJ: Penetrating cardiac injuries. Surg Clin North 

Am 1996;76:685-724. 
Asensio JA, VoystockJ, Khatri VJ, Kerstein MD: Toracotomia en el Centro de 

Urgencias. Procedimientos en el Paciente Critico 1993;111:337-341. 
Asensio JA, Wall M, Minel J: Practice management guidelines for emergency 

department thoracotomy. J Am Coll Surg 2001;193:303-309. 
Asfaw I, Arbulu A: Penetrating wounds of the pericardium and heart. Surg Clin 

North Am 1977;57:37-48. 
Asfaw I, Ramadan H, Talbert JG, Arbulu A: Double traumatic rupture of the 

thoracic aorta. J Trauma 1985;25:1102-1104. 
Ashbell TS, Kleinert HE, KutzJE: Vascular injuries about the elbow. Clin Orthop 

1967;50:107-127. 
Ash ton F, Slaney G: Arterial injuries in civilian surgical practice. Injury 1970;1 :303- 

314. 
Ashworth EM, Daising MC, Glover JL, Reilly MK: Lower extremity vascular trauma. 

J Trauma 1988;28:329-336. 
Assalia A, Schein M: Resuscitation of haemorrhagic shock. Br J Surg 1993;80:213. 
Attar S, Ayella RJ, McLaughlin JS: The widened mediastinum in trauma. Ann 

Thorac Surg 1972;13:435-449. 
Aufranc OE, Jones WN, Stewart WGJr: Common fracture with unusual associ- 
ated vascular injury. JAMA 1965;191:1073-1075. 
Aust JC, Bredenberg CE, Murray DG: Mechanisms of arterial injuries associ- 
ated with total hip replacement. Arch Surg 1981;116:345-349. 
Austin JH, Carsen GM: Acute traumatic hematoma of aorta. J Thorac Cardio- 

vascSurgl976;71:321. 
Ayella RJ, Hankins JR, Turney SZ, Cowley RA: Ruptured thoracic aorta due to 

blunt trauma. J Trauma 1977;17:199-205. 
Ayres ML, Winspur I: Major injuries and bullet wounds. Injury 1976;7:221-224. 
Ayuyao AM, Kaledzi YL, Parsa MH, Freeman HP: Penetrating neck wounds: 

Mandatory versus selective exploration. Ann Surg 1985;202:563-567. 
Baadsgaard SE, Bille S, Egeblad K: Major vascular injury during gynecologic 

laparoscopy: Report of case and review of published cases. Acta Obstet 

Gynecol Scand 1986;68:283-285. 
Babu SC, Piccorelli GO, Shah PM, et al: Incidence and results of arterial com- 
plications among 1 6,350 patients undergoing cardiac catheterization.J Vase 

Surg 1989;10:113-116. 
Bacharach JM, Garratt KN, Rooke TW: Chronic traumatic thoracic aneurysm: 

Report of two cases with the question of timing for surgical intervention. 

J Vase Surg 1993;17:780-783. 
Baek SM, Brown RS, Shoemaker WC: Cardiac tamponade following wound tract 

injection. J Trauma 1973;13:85-87. 



RE.qxd 4/16/04 3:19 PM Page 544 



544 VI • THE RICH REFERENCE COLLECTION 



Baik S, Uku JM, Joo KG: Seat belt injuries to the left common carotid 
artery and left internal carotid artery. Am J Forens Med Pathol 1988;9:38- 
39. 

Bailey H: Wounds of the neck. In Bailey H (ed): Surgery of Modern Warfare. 
Edinburgh, E & S Livingston, 1944. 

Baillot R, Dontigny L, Verdant A, et al: Penetrating chest trauma: A 20-year 
experience. J Trauma 1987;27:994-997. 

Baillot R, Dontigny L, Verdant A, et al: Intrapericardial trauma: Surgical expe- 
rience. J Trauma 1989;29:736-740. 

Baird RJ, Doran ML: The false aneurysm. Can Med Assoc J 1964;91:281- 
284. 

Baker CC, Thomas AN, Trunkey DD: The role of emergency room thoraco- 
tomy on trauma. J Trauma 1980;20:848-855. 

Baker MS: Recognition of vascular injury in the trauma patient. Milit Med 
1990;155:225. 

Baker SP, O'Neill B, Haddon WJr, Long WB: A method for describing patients 
with multiple injuries and evaluating emergency care.J Trauma 1974; 14: 187- 
196. 

Bakker FC, Patka P, Haarman HJ: Combined repair of a traumatic rapture of 
the aorta an anterior stabilization of a thoracic spine fracture: Case report. 
J Trauma 1996;40:128-129. 

Bakker KW, Cast LF: Retroperitoneal haemorrhage from the superior gluteal 
artery: A late complication of total hip arthroplasty. Clin Rheumatol 
1990;9:249. 

Ballance C: The surgery of the heart (Bradshaw Lecture). Lancet 1920;1:1. 

BaltschJ, Klose KJ, GonningerJ: Angiography in acute arterial trauma of periph- 
eral vessels and the aorta. Vasa 1987;16:246-250. 

Bandt RL, Foley WJ, FinkGH, Regan WJ: Mechanism of perforation of the heart 
with production of hydropericardium by a venous catheter and its pre- 
vention. Am J Surg 2001;33:311-316. 

Banks E, Chun J, Weaver FA: Chronic innominate artery dissection after blunt 
thoracic trauma: Case report. J Trauma 1995;38:975-978. 

Barcia PJ, Nelson TG, Whelan TJ: Importance of venous occlusion in arterial 
repair failure: An experimental study. Ann Surg 1972;175:223-227. 

Barcia TC, Livoni JP: Indications for angiography in blunt thoracic trauma. 
Radiology 1983;147:15-19. 

Baret AC, DeLong RP, Tukanowicz SA, Blakemore WS: Transfixation of the 
aorta accompanied by a Browne-Syndrome: A case report. J Trauma 
1958;35:359-362. 

Barker NW, Hines EAJr: Arterial occlusion in the hands and fingers associated 
with repeated occupational trauma. Proc Staff Meet Mayo Clin 1944;19:345- 
349. 

Barker JE, Knight KR, Romeo R, et al: Targeted disruption of the nitric oxide 
synthase 2 gene protects against ischaemia/reperfusion injury to skeletal 
muscle. J Pathol 2001;194:109-115. 

Barkun JS, Terazza O, Daignault P, et al: The fate of venous repair after shock 
and trauma. J Trauma 1988;28:1322-1329. 

Barner HB: Sutureless intraluminal aortic prosthesis in traumatic aortic rupture: 
Is it appropriate? J Trauma 1988;28:1607-1608. 

Barnes RW, Petersen JL, Krugmire RB, Strandness DEJr: Complications of per- 
cutaneous femoral arterial catheterization: Prospective evaluation with the 
Doppler ultrasonic velocity detector. Am J Cardiol 1974;33:259-263. 



RE.qxd 4/16/04 3:19 PM Page 545 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 545 

Barone GW, Kahn MB, Cook JM, et al: Traumatic left renal artery stenosis 
managed with splenorenal bypass: Case report. J Trauma 1990;30:1594- 
1596. 

Barros D'Sa AAB: A decade of missile-induced vascular trauma. Ann Surg 
1981;64:37-44. 

Barros D'Sa AAB: Management of vascular injuries of civil strife. Injury 1982;14:51- 
57. 

Barros D'Sa AAB: The Rationale for Arterial and Venous Shunting in the Man- 
agement. Belfast, Northern Ireland, Grune &Stratton, 1989. 

Barros D'Sa AAB, Hassard TH, Livingston RH, Irwin JW: Missile-induced vas- 
cular trauma. Injury 1980;12:13-30. 

Bass A, Papa M, Morag B, Adar R: Aortic false aneurysm following blunt trauma 
of the abdomen. J Trauma 1983;23:1072-1073. 

Bass A, Allison EJ, Reines HD: Thigh compartment syndrome without lower 
extremity trauma following application of pneumatic antishock trousers. 
Ann Emerg Med 1983;12:382-384. 

Bassett FH, Houck WS: False aneurysm of profunda femoris artery after sub- 
trochanteric osteotomy and nail-plate fixation. J Bone Joint Surg 
1964;46A:583-585. 

Bassett FH, Silver D: Arterial injury associated with fractures. Arch Surg 
1966;92:13-19. 

BattM, Hassen-KhoudjaR, BayadaJM, etal: Traumatic fistula between the aorta 
and the left renal vein: Case report and review of the literature. J Vase Surg 
1989;9:812-816. 

Batzdorf U, BentsonJR, Machleder HI: Blunt trauma to the high cervical carotid 
artery. Neurosurgery 1979;5:192-201. 

Baumgartner F, White GH, White RA, et al: Delayed, exsanguinating pelvic 
hemorrhage after blunt trauma without bony fracture: Case report.J Trauma 
1990;30:1603-1605. 

Baxter BT, Moore EE, Synhorst DR et al: Graded experimental cardiac contu- 
sion: Impact on cardiac rhythm, coronary artery flow, ventricular function, 
and myocardial oxygen consumption. J Trauma 1988;28:1411-1417. 

Beall ACJr: Penetrating wounds of the aorta. Am J Surg 1960;99:770-774. 

Beall ACJr, Arbergast NR, Hallman GL: Complete transection of the thoracic 
aorta due to rapid deceleration. Am J Surg 1967;114:769-773. 

Beall ACJr, Arbergast NR, Ripepi AC, et al: Aortic laceration due to rapid dece- 
leration. Arch Surg 1969;98:595-601. 

Beall ACJr, Crosthwait RW, Crawford ES, DeBakey ME: Gunshot wounds of the 
chest: A plea for individualization. J Trauma 1964;4:382. 

Beall ACJr, Diethrich EB, Cooley DA, DeBakey ME: Surgical management of 
penetrating cardiovascular trauma. South Med J 1967;60:698-704. 

Beall ACJr, Diethrich EB, Crawford HW, et al: Surgical management of pene- 
trating cardiac injuries. Am J Surg 1966;112:686-692. 

Beall ACJr, Diethrich EB, Morris GC Jr, DeBakey ME: Surgical management of 
vascular trauma. Surg Clin North Am 1966;46:1001-1011. 

Beall ACJr, Gasior RM, Bricker DL: Gunshot wounds of the heart: Changing 
patterns of surgical management. Ann Thorac Surg 1971;11:523-531. 

Beall ACJr, Harrington OB, Crawford ES, DeBakey ME: Surgical management 
of traumatic arteriovenous aneurysms. Am J Surg 1963;106:610-618. 

Beall ACJr, Morris GC Jr, Cooley DA: Temporary cardiopulmonary bypass in 
the management of penetrating wounds of the heart. Surgery 1962;52:330- 
337. 



RE.qxd 4/16/04 3:19 PM Page 546 



546 VI • THE RICH REFERENCE COLLECTION 



Beall AC Jr, Ochsner JL, Morris GC Jr, et al: Penetrating wounds to the heart. 
J Trauma 1961;1:195-207. 

Beall AC Jr, Patrick TA, Okies JE, et al: Penetrating wounds of the heart: Chang- 
ing patterns of surgical management. J Trauma 1972;12:468-473. 

Beall AC Jr, Roof WR, DeBakey ME: Successful surgical management of through 
and through stab wound of the aortic arch. Ann Surg 1962;156:283-286. 

Beall AC Jr, ShirkeyAL, DeBakey ME: Penetrating wounds of the carotid arter- 
ies. J Trauma 1963;3:276-287. 

Beattie EJJr, Greer D: Laceration of the aorta: Case report of successful repair 
forty-eight hours after injury. J Thorac Surg 1952;23:293-298. 

Beattie EJJr, Oldham JB, RossJA: Superior thyroid arteriovenous aneurysm. 
Br J Surg 1961;48:456-457. 

Beatty RA: Dissecting hematoma of the internal carotid artery following 
chiropractic cervical manipulation. J Trauma 1977;17:248-249. 

BeckCS: Wounds of the heart: The technique of suture. Arch Surg 1926;13:205- 
227. 

Beck CS: Further observations on stab wounds of the heart. Ann Surg 
1942;115:698-704. 

Beck DE, Robison JG, Hallett JE Jr: Popliteal artery pseudoaneurysm following 
arthroscopy. J Trauma 1986;26:87-89. 

Becker G, Palmaz J, Rees C: Angioplasty-induced dissections in human iliac 
arteries: Management with Palmaz-balloon-expandable intraluminal 
stents. Radiology 1990;176:31. 

Becker HM, Ramirez J, Echave V, Heberer G: Traumatic aneurysms of the 
descending thoracic aorta. Ann Vase Surg 1986;1:196-200. 

Becker HM, WexlerJ, Frater RW: False aneurysm of aorta secondary to partial 
occlusion clamp injury diagnosis by nuclear flow study. Chest 1981;80:331- 
333. 

BednarskiJJ, Nayduch DA: Thoracic aorta rupture as a cause of paraplegia: A 
diagnostic dilemma (case report). J Trauma 1989;29:531-533. 

Beebe GW, Bergan JJ, Bergqvist D, et al: Classification and grading of chronic 
venous disease in the lower limbs. A consensus statement. Eur Vase Endovasc 
Surg 1996;12:487-492. 

Beebe HG: Complications in Vascular Surgery. Philadelphia, JB Lippincott Co, 
1973. 

Beel T, Harwood AL: Traumatic rupture of the thoracic aorta. Ann Emerg Med 
1980;9:483-486. 

BeggJW: Recanalization of autogenous vein graft. Southern MedJ 1988;81:1446- 
1447. 

Beggs CW, Helling TS, Evans LL, et al: Evaluation of cardiac injury by two- 
dimensional echocardiography in patients suffering blunt chest trauma. 
Ann Emerg Med 1987;16:542-545. 

Belcaro G, Nicolaides AN: Pressure index in hypotensive of hypertensive patients. 
J Cardiovasc Surg 1989;30:614-617. 

Beless DJ, Muller DS, Perez H: Aortoiliac occlusion secondary to atheroscle- 
rotic plaque rapture as the result of blunt trauma. Ann Emerg Med 
1990;19:922-924. 

Belkin M, Dunton R, Crombie HD Jr, Lowe R: Preoperative percutaneous intra- 
luminal balloon catheter control of major arterial hemorrhage. J Trauma 
1988;28:548-550. 

Bell D, Cockshott WP: Angiography of traumatic arterio-venous fistulae. Clin 
Radiol 1965;16:241-247. 



RE.qxd 4/16/04 3:19 PM Page 547 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 547 

Bell J: Principles of surgery. Discourse 1801;9:4. 

Bell KA, Simon BK: Aneurysm of popliteal artery after medial meniscectomy: 

An unusual complication. South MedJ 1979;72:1126-1134. 
Bellamy RF: The causes of death in conventional land warfare: Implications for 

combat casualty care research. Mil Med 1984;149:55-62. 
Belley G, Gallix BP, Derossis AM, et al: Profound hypotension in blunt trauma 

associated with superior gluteal artery rupture without pelvic fracture. 

J Trauma 1997;43:703-705. 
Ben-Menachem Y: Vascular injuries of the extremities: Hazards of unnecessary 

delays in diagnosis. Orthopedics 1986;9:333-338. 
Ben-Menachem Y: Avulsion of the innominate artery associated with fracture 

of the sternum. Am J Cardiol 1988;150:621-622. 
Ben-Menachem Y: Rupture of the thoracic aorta by broadside impacts in road 

traffic and other collisions: Further angiographic observations. J Trauma 

1993;36:363-367. 
Ben Hur N, Gemer M, Milwidsky H: Perforating injury of the heart caused by 

nail fired from a studgun. J Trauma 1964;84:850-853. 
Benckart DH, Magovern GJ, Lievler GA, et al: Traumatic aortic transection: 

Repair using left arterial to femoral bypass. J Cardiac Surg 1989;4:43-49. 
Benitez PR, Newell MA: Vascular trauma in drug abuse: Patterns of injury. Ann 

Vase Surg 1986;1:175-180. 
Benito MC, Garcia F, Fernandez-Quero L, et al: Lesion of the internal carotid 

artery caused by a car safety belt. J Trauma 1990;30:116-117. 
Bennett DE, CherryJK: The natural history of traumatic aneurysms of the aorta. 

Surgery 1967;61:516. 
Bennett JE: Expanding forearm hematoma after apparent minor injury. Plastic 

Reconstr Surg 1965;36:622-625. 
BenvenutoR, Rodman FS, Glimour J, etal: Composite venous grafts for replace- 
ment of the superior vena cava. Arch Surg 1962;84:570-573. 
BerendesJN, BredeeJJ, SchipperheynJJ, Mashhour YAS: Mechanisms of spinal 

cord injury after cross-clamping of the descending thoracic aorta. Circu- 
lation 1982;66:112-116. 
BerganF: Traumatic intimal rupture of the popliteal artery with acute ischemia 

of the limb in cases with supracondylar fractures of the femur. J Cardiac 

Surg 1963;4:300-302. 
Bergan JJ, Conn J Jr, Trippel OH: Severe ischemia of the hand. Ann Surg 

1971;173:301-307. 
Bergan JJ, Dean RH, Yao JST: Vascular injuries in pelvic cancer surgery. Am J 

Obstet Gynecol 1976;124:562-566. 
Bergan JJ, Yao JST: Symposium on Venous Problems in Honor of Geza de Takats. 

Chicago, Year Book Medical Publishers, 1978. 
Bergan JJ, Yao JST: Surgery of the Veins. Orlando, Grune & Stratton, 1985. 
Bergen tz SE, Bergqvist D: Iatrogenic Vascular Injuries. New York: Springer-Verlag, 

1989. 
Bergen tz SE, Hansson LO, Norback B: Surgical management of complications 

to arterial puncture. Ann Surg 1966;164:1021-1026. 
Bergin PJ: Aortic thrombosis and peripheral embolization after thoracic gunshot 

wound by transesophageal echocardiography. Am Heart 1990;1 19:688- 

690. 
Berglund B, Swanbeck G, Hedin H: Low molecular weight dextran therapy for 

digital ischemia due to collagen vascular disease. Dermatologica 1981; 

163:353-357. 



RE.qxd 4/16/04 3:19 PM Page 548 



548 VI • THE RICH REFERENCE COLLECTION 



Bergman K, Spence L, Wesson D, Dykes E: Thoracic vascular injuries: A post 
mortem study. J Trauma 1990;30:604-606. 

Bergquist D, Takolander R: Aortic occlusion following blunt trauma of the 
abdomen. J Trauma 1981;21:319-322. 

Bergquist D, Carlsson AS, Ericsson BF: Vascular complications after total hip 
arthroplasty. Acta Orthop Scand 1983;54:157-163. 

Berguer R, Feldman AJ, Wilner HI, Lazo A: Arteriovenous vertebral fistulae: 
Cure by combination of operation and detachable intravascular balloon. 
Ann Surg 1982;196:65-68. 

Berkoff HA, Carpenter EW, Frey CF: Evaluation of balloon tamponade of the 
abdominal aorta. J Surgery 1971;11:496-500. 

Bernheim BM: Surgery of the vascular system. Philadelphia, JB Lippincott Co, 
1913. 

Bernheim BM: Bloodvessel surgery in the war. Surg Gynecol Obstet 1920;30:564- 
567. 

BertiniJEJr, Flechner SM, Miller P, et al: The natural history of traumatic branch 
renal artery injury. J Urol 1986;135:228-230. 

Bezreh AA: Injuries resulting from hostile action against army aircrew members 
in flight over Vietnam. Aerospace Med 1970;41:763. 

Bhatnagar MK, Smith GS: Trauma in the Afghan Guerilla War: Effects of lack 
of access to care. Surgery 1989;105:699. 

Bickell WH, Shaftan GW, Mattox KL: Intravenous fluid administration and uncon- 
trolled hemorrhage. J Trauma 1989;29:409. 

Bickell WH, Wall MJ Jr, Pepe PE, et al: Immediate versus delayed fluid resusci- 
tation for hypotensive patients with penetrating thoracoabdominal trauma. 
N EnglJ Med 1994;331:1105-1109. 

Bickham WS: Arteriovenous aneurysm. Ann Surg 1904;39:767-775. 

Bigger IA: Heart wounds: A report of seventeen patients operated upon in the 
Medical College of Virginia Hospitals and discussion of the treatment and 
prognosis. J Thorac Surg 1939;8:239-253. 

Billings KJ, Nasca RJ, Griffin HA: Traumatic arteriovenous fistula with sponta- 
neous closure. J Trauma 1973;13:741-743. 

Billroth T: The surgery of the heart (Bradshaw Lecture). Lancet 1920;1-1:73- 
134. 

Billroth T: Wounds of the heart: The technique of suture. Arch Surg 1926; 13:205- 
227. 

Billroth T: Historical studies on the nature and treatment of gunshot wounds 
from the 15th century to the present time. YaleJ Biol Med 1931;4:16-36. 

Billy LJ, Amato JJ, Rich NM: Aortic injuries in Vietnam. Surgery 1971;70:385- 
391. 

BinetJP, LangloisJ, Cormier JM, Saint Florent GD: A case of recent traumatic 
avulsion of the innominate artery at its origin from the aortic arch: Suc- 
cessful surgical repair with deep hypothermia. J Thorac Cardiovasc Surg 
1962;43:670-676. 

Binkley FM, Wylie EJ: A new technique for obliteration of cerebrovascular arte- 
riovenous fistulae. Arch Surg 1973;106:524-527. 

Birchall D, Fields JM, Chalmers N, Walker MG: Case report: Delayed superfi- 
cial femoral artery pseudoaneurysm rupture following successful com- 
pression therapy. Clin Radial 1997;52:629-630. 

BirchardJD, Pichara DR, Brown PM: External iliac artery and lumbosacral plexus 
injury secondary to an open book fracture of the pelvis: Report of a case. 
J Trauma 1990;30:906-908. 



RE.qxd 4/16/04 3:19 PM Page 549 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 549 

Birkeland IW, Taylor TK: Major vascular injuries in lumbar disc surgery. J Bone 
Joint Surg 1969;51B:4-19. 

Birnholz JC: Radionuclide angiography in the diagnosis of aortocaval fistula. 
J Thorac Cardiovasc Surg 1973;65:292-295. 

Bisenkov LN, Isamukhamedov T, Panin SM: Heart wound with migration of a 
bullet into the abdominal aorta. Vestn Khir 1983;131:97-98. 

Bishara RA, Pasch AR, Lim LT, et al: Improved results in the treatment of civil- 
ian vascular injuries associated with fractures and dislocations. J Vase Surg 
1986;3:707-711. 

BizerL: Peripheral vascular injuries in the Vietnam War. Arch Surg 1969;98:165- 
166. 

Blackford JM, McLaughlin JS: Pseudoaneurysm of the carotid artery. Am Surg 
1973;39:257-260. 

Blacklay PF, Duggan E, Wood RFM: Vascular trauma. Br J Surg 1987;74:1077- 
1083. 

Blackmore TL, Whelan TJ: Arteriovenous fistula as a complication of gastrec- 
tomy. Am J Surg 1965;109:197. 

Bladergroen M, Brockman R, Luna G, et al: A twelve-year survey of cervicothoracic 
vascular injuries. Am J Surg 1989;157:483-486. 

Blaisdell FW: Discussion in Buscaglia: Penetrating abdominal vascular injuries. 
Arch Surg 1969;109:197. 

Blaisdell FW, Cooley DA: The mechanism of paraplegia after temporary tho- 
racic aortic occlusion and its relationship to spinal fluid pressure. Surgery 
1962;51:351-355. 

Blaisdell FW, Hall AD, Lim RC: Aortoiliac arterial substitution utilizing subcu- 
taneous grafts. Ann Surg 1970;172:775-780. 

Blake HA, Inmon TW, Spencer FC: Emergency use of antegrade aortography 
in diagnosis of acute aortic rupture. Ann Surg 1960;152:954-956. 

Blakemore AH: Restorative endoaneurysmorrhaphy by vein graft inlay. Ann 
Surg 1947;126:841-849. 

Blakemore AH, Lord JE Jr: A non-suture method of blood vessel anastomosis: 
Review of experimental study and report of clinical cases. Ann Surg 1945;121: 
435-453. 

Blakemore AH, Lord JW, Stefko PL: The severed primary artery in the war 
wounded; a non suture method of bridging arterial defects. Surgery 
1942;12:488-508. 

Blalock A: A successful suture of a wound of the ascending aorta. JAMA 
1934;103:1617-1618. 

Blalock A, Ravitch MM: Consideration of the non-operative treatment of cardiac 
tamponade resulting from wounds to the heart. Surgery 1943;14:157- 
162. 

Blanchard DG, Kimura BJ, Dittrich HC, DeMaria AN: Transesophageal echocar- 
diography of the aorta. JAMA 1994;272:546-551. 

Bland-Sutton J: A lecture on missiles as emboli. Lancet 1919;1:773-775. 

Bland EF, Beebe GW: Missiles in the heart. A twenty-year follow-up report of 
World War II cases. New Engl J Med 1966;274:1039-1046. 

Blatchford JW III, Anderson RW: The evolution of the management of pene- 
trating wounds of the heart. Ann Surg 1985;202:615-623. 

Blegvad S, Lippert H, Lund O, et al: Acute delayed surgical treatment of trau- 
matic rupture of the descending aorta. J Cardiovasc Surg 1989;30:559-564. 

Blickenstaff KL, Weaver FA, Yellin AE, et al: Trends in management of trau- 
matic vertebral artery injuries. Am J Surg 1989;158:101-106. 



RE.qxd 4/16/04 3:19 PM Page 550 



550 VI • THE RICH REFERENCE COLLECTION 



Blohme I, Brynger H: Emergency ligation of the external iliac artery. Ann Surg 

1985;201:505-510. 
Bloom AI, Sasson T, Ravkind A, Pikarski B-ZJ: Diagnosis of traumatic rupture 

of the thoracic aorta using dynamic rapid sequence axial computerized 

tomography. Isr J Med Sci 1995;31:314-320. 
Bloom JD, Mozersky DJ, Buckley CG, Hagood COJr: Defective limb growth as 

a complication of catheterization of the femoral artery. Surg Gynecol Obstet 

1974;138:524-526. 
Blumoff RL, Powell T, Johnson GJr: Femoral venous trauma in a university 

referral center. J Trauma 1982;22:703-705. 
Blumoff RL, Proctor HJ, Johnson GJr: Recanalization of a saphenous vein inter- 
position venous graft. J Trauma 1981;21:407-408. 
Bodai BI, Smith P, Ward RE: Emergency thoracotomy in the management of 

trauma: A review. JAMA 1983;249:1891. 
Bodily K, Perry JF, Strate R, Fischer RP: The salvageability of patients with post- 
traumatic rupture of the descending thoracic aorta in a primary trauma 

center. J Trauma 1977;17:754-760. 
Boerhaave H: Cited in Elkin 1941. Aphorismi de Cognoscendis et Curandis 

Morbis (aphorism 170). 1709. 
Bogaert AM, DeScheerder I, Calardyn F: Successful treatment of aortic rupture 

presenting as a syncope: The role of echocardiography in diagnosis. IntJ 

Cardiology 1987;16:212-214. 
Bolanowki PJP, Swaminatham AP, Neville WE: Aggressive surgical management 

of penetrating cardiac injuries. J Thorac Cardiovasc Surg 1973;66:52-57. 
Bolasny BL, Killen DA: Surgical management of arterial injuries secondary to 

angiography. Ann Surg 1971;174:962-964. 
Bole P, Andronaco JT, Purdy R: Superior mesenteric arteriovenous fistula 

secondary to a gunshot wound. J Cardiac Surg 1973;14:456. 
Bole PV, Cortes LE, Munda RT, et al: Repair of false aneurysm of the abdomi- 
nal aorta. J Trauma 1975;15:255-259. 
Bole PV, Katz M, Cohen NH, Clauss RH: Carotid crossover bypass: Cerebral 

revascularization after ligation of common carotid artery. Cardiovasc Surg 

1977;18:533-538. 
Bole PV, Munda R, Purdy RT: Traumatic pseudoaneurysms: A review of 32 cases. 

J Trauma 1976;16:63-70. 
Bole PV, Purdy RT, Munda RT, et al: Civilian arterial injuries. Ann Surg 

1976;183:13-23. 
Bollett AJ: Wounded presidents: 1981 almost repeats the events of 1881. Mil 

Med 1981;109:19-23. 
Bongard F, Dubrow T, Klein S: Vascular injuries in the urban battleground: 

Experience at a metropolitan trauma center. Ann Vase Surg 1990;4:415. 
Bongard F,Johs SM, Leighton TA, Klein SR: Peripheral arterial shotgun missile 

emboli: Diagnostic and therapeutic management case reports. J Trauma 

1991;31:1426-1431. 
Bongard FS, White GH, Klein SR: Management strategy of complex extremity 

injuries. Am J Surg 1989;158:151-155. 
Bongard FS, Wilson SE, Perry MO: Vascular injuries in surgical practice. In 

Bongard FS, Wilson SE, Perry MO (eds) : Norwalk, Conn, Appleton & Lange, 

1991. 
Boontje AH: Iatrogenic vascular trauma. J Cardiac Surg 1978;19:335-340. 
Boontje AH: True aneurysm of the abdominal aorta due to blunt trauma. 

J Cardiac Surg 1978;19:359-363. 



RE.qxd 4/16/04 3:19 PM Page 551 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 551 

BordeauxJ, GuysJM, Magnan PE: Multiple aneurysms in a seven-year-old child. 

Ann Vase Surg 1990;4:26-28. 
Borja AR, Lansing AM: Thrombosis of the abdominal aorta caused by blunt 

trauma. J Trauma 1970;10:499-501. 
Borja AR, Lansing AM: Traumatic rupture of the heart: A case successfully treated. 

J Vase Surg 1970;171:438-440. 
Borlase BC, Metcalf RK, Moora EE: Penetrating wounds to the anterior chest. 

Analysis of thoracotomy and laparotomy. Am J Surg 1986;6:649. 
Borlase BC, Moore EE, Moore FA: The abdominal trauma index: A critical 

reassessment and validation. J Trauma 1990;30:1340-1344. 
Borman KR, Aurbakken CM, WeigeltJA: Treatment priorities in combined blunt 

abdominal and aortic trauma. Am J Surg 1982;144:728-732. 
Borman KR, Jones GH, Snyder WH Jr: A decade of lower extremity venous 

trauma: Patency and outcome. Am J Surg 1987;154:608-612. 
Borman KR, Snyder WH III, WeigeltJA: Civilian arterial trauma of the upper 

extremity: 1 1-year experience in 267 patients. Am J Surg 1984;148:796-799. 
Boruchow IB, Iyengar R,JudeJR: Injury to ascending aorta root trauma.J Thorac 

Cardiovasc Surg 1977;73:303-305. 
Boruchow IB: Delayed death from aortic root trauma. Ann Thorac Surg 

1991;51:317-319. 
Bosher LH, Freed TA: The surgical treatment of traumatic rupture of avulsion 

of the innominate artery. J Thorac Cardiovasc Surg 1967;54:732-739. 
Bouchard JF, Lamontagne D: Protection afforded by preconditioning to the 

diabetic heart against ischaemic injury. Cardiovasc Res 1998;37:82-90. 
Bourland WA, Kispert JF, Hyde GL, Kazmers A: Trauma to the proximal supe- 
rior mesenteric artery: A case report and review of the literature. J Vase 

Surg 1992;15:669-674. 
Bower TC, Murray MJ, Gloviczki P: Effects of thoracic aortic occlusion and cere- 
brospinal fluid drainage on regional spinal cord blood flow in dogs: Cor- 
relation with neurologic outcome. J Vase Surg 1989;9:135-144. 
Boyd DP, Farha GJ: Arteriovenous fistula and isolated vascular injuries secondary 

to intervertebral disk surgery: Report of four cases and review of the lit- 
erature. Ann Surg 1965;161:524-531. 
Boyd TF, Strieder JW: Immediate surgery for traumatic heart disease. J Thorac 

Cardiovasc Surg 1965;50:305-315. 
Boyden AM: Personal communication, 1970. 
Boyle EM Jr, Canty T Jr, Morgan E, et al: Treating myocardial ischemia- 

reperfusion injury by targeting endothelial cell transcription. Ann Thor 

Surg 1999;68:1949-1953. 
Bradham GB, Nunn D, Brailsford LE: Successful repair of a bullet wound of 

the abdominal aorta. Ann Surg 1962;155:86-89. 
Bradham RR, Buxton J, StallworthJ: Arterial injury of the lower extremity. Surg 

Gynecol Obstet 1964;118:995-1000. 
Bradham WG, Lewis J, Sewell D, Garrett A: Bullet embolus to the ascending 

aorta following a gunshot wound to the chest. J Tenn Med Assoc 

1991;84:592-593. 
Bradley EL III: Management of penetrating carotid injuries: An alternative 

approach. J Trauma 1973;13:248-255. 
BrandayJM, Kumar E, Spencer H: Intrapericardial heart and great vessel injuries. 

West Indian Med J 1984;33:176-119. 
Branham HH: Aneurysmal varix of the femoral artery and vein following a 

gunshot wound. IntJ Surg 1890;3:250. 



RE.qxd 4/16/04 3:19 PM Page 552 



552 VI • THE RICH REFERENCE COLLECTION 



Brantigan CO: Delayed major vessel hemorrhage following tracheostomy. 
J Trauma 1973;13:235-237. 

Brathwaite CE, Rodriguez: Injuries of the abdominal aorta from blunt trauma. 
Am Surg 1992;58:350-352. 

Brathwaite CE, CilleyJ, O'Connor W, et al: The pivotal role of transesophageal 
echocardiography in the management of traumatic thoracic aortic rupture 
with associated intra-abdominal hemorrhage. Chest 1994;105:1899. 

Brawley RK, Murray G, Crisler C, Cameron J: Management of wounds of the 
innominate, subclavian and axillary blood vessels. Surg Gynecol Obstet 
1970;131:1130-1140. 

Breaux EP, Dupont J Jr, Alvert HM, et al: Cardiac tamponade following pene- 
trating mediastinal injuries: Improved survival with early pericardiocen- 
tesis. J Trauma 1979;19:461-466. 

Brener BJ, Couch N: Peripheral arterial complications of left heart catheteri- 
zation and their management. Am J Surg 1973;125:521-526. 

Brengman ML, O'Donnell S, Mullenix P, et al: The fate of a patent carotid 
artery contralateral to an occlusion. Ann Vase Surg 2000;14:77-81. 

Brentlinger A, Hunter J: Perforation of the external iliac artery and ureter pre- 
senting as acute hemorrhagic cystitis after total hip replacement. J Bone 
Joint Surg 1987;69:620-622. 

Brewer LA III: Wounds of the chest in war and peace, 1943-1968. Ann Thorac 
Surg 1969;7:387-408. 

Brewer LA III: The contributions of the Second Auxiliary Surgical Group to 
military surgery during World War II with special reference to thoracic 
surgery. Ann Surg 1983;197:318-326. 

Brewer LA III, Carter R: Wounds of the great vessels of the thorax: Diagnosis 
and surgical approach in twenty-four cases. Am J Surg 1967;1 14:340- 
350. 

Brewer LA III, Carter R: Elective cardiac arrest for the management of mas- 
sively bleeding heart wounds. JAMA 1967;200:1023-1025. 

Brewer LA III, Carter R: A rational treatment of small and large wounds of the 
heart. Surg Gynecol Obstet 1968;126:977-985. 

Brewer PL, Schramel R, Menendez C, Creech O Jr: Injuries of the popliteal 
artery: A report of sixteen cases. Am J Surg 1969;118:36-42. 

Brewster D, May A, Darling RC: Variable manifestations of vascular injury during 
lumbar disk surgery. Arch Surg 1979;114:1026-1030. 

Brewster SA, Thirlby R, Synder WH: Subxiphoid pericardial window and 
penetrating cardiac trauma. 1988. 

Bricker D, Hallman GL: Complete transection of the thoracic aorta: Manage- 
ment of a case associated with massive total body injury. J Trauma 
1970;10:420-426. 

Bricker DL, Beall ACJr, DeBakey ME: The differential response to infection of 
autogenous vein versus Dacron arterial prosthesis. Chest 1970;58:566- 
570. 

Bricker DL, Morton JR, Okies JE, Beall ACJr: Surgical management of injuries 
the vena cava: Changing patterns of injury and newer techniques of repair. 
J Trauma 1971;11:725-735. 

Bricker DL, Noon G, Beall ACJr: Vascular injuries of the thoracic outlet.J Trauma 
1970;10:1-15. 

Bricker DL, Waltz T, Telford R, Beall ACJr: Major abdominal and thoracic 
trauma associated with spinal cord injury: Problems in management. 
J Trauma 1971;11:63-75. 



RE.qxd 4/16/04 3:19 PM Page 553 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 553 

Brigham RA, Eddleman W, Clagett GP, Rich NM: Isolated venous injury pro- 
duced by penetrating trauma to the lower extremity. J Trauma 1983;23:255- 
257. 

Brigham RA, Salander J, Rich NM: Abdominal venous trauma. In Scribner RG, 
Brown WH, Tawes RL (eds): Decision Making in Vascular Surgery. 
Toronto, BC Decker, 1988. 

Brigham RA, Salander JM, Rich NM: Extremity venous trauma. In Scribner RG, 
Brown WH, Tawes RL (eds): Decision Making in Vascular Surgery. 
Toronto, BC Decker, 1988. 

Bright EF, Beck CS: Nonpenetrating wounds of the heart. Am Chest J 1935; 
10:293-320. 

Brink BE: Vascular trauma. Surg Clin North Am 1977;57:189-196. 

Brinton M, Miller S, Lim RC Jr, Trunkey DD: Acute abdominal aortic injuries. 
J Trauma 1982;22:481-486. 

Brisbin RL, Geib PO, Eiseman B: Secondary disruption of vascular repair fol- 
lowing war wounds. Arch Surg 1969;99:787-791. 

Brittain RS, Marchioro TL, Hermann G, et al: Accidental hepatic artery liga- 
tion in humans. Am J Surg 1964;107:822-832. 

Bromley LL, HobbsJT, Robinson RE: Early repair of traumatic rupture of the 
aorta. Br MedJ 1965;2:17-19. 

Brooks AL, Fowler SB: Axillary artery thrombosis after prolonged use of crutches. 
J Bone Joint Surg 1964;46-A:863-864. 

Brooks AP, Olson LK, Shackford SR: Computed tomography in the diagnosis 
of traumatic rupture of the thoracic aorta. Clinical Radiology 1989;40:133- 
138. 

Brooks SW, Cmolik BL, Young JC, et al: Transesophageal echocardiographic 
examination of a patient with traumatic aortic transection from blunt chest 
trauma: A case report. J Trauma 1991;31:841-845. 

Brooks SW, Young JC, Cmolik BL, et al: The use of transesophageal echocar- 
diography in the evaluation of chest trauma. J Trauma 1992;23:761-765. 

Brophy DP, Sheiman RG, Amatulle P, Akbari CM: Iatrogenic femoral pseudoa- 
neurysms: Thrombin injection after failed US-guided compression. Radi- 
ology 2001;214:278-282. 

Bross W: Injuries of the thoracic aorta. J Cardiac Surg 1971;12:95-103. 

Brotman S, Browner B, Cox E: MAST trousers improperly applied causing com- 
partmental syndrome in lower extremity trauma. J Trauma 1982;22:598- 
599. 

Brown AJ: Old masterpieces in Surgery: The surgery of HieronymusBrunschwig. 
Surg Gynecol Obstet 1924;38:133. 

Brown C: Inadvertent prolonged cannulation of the carotid artery. Anesth Analg 
1982;61:150-152. 

Brown JJ, Greene FL, McMillan RD: Vascular injuries associated with pelvic frac- 
tures. Am J Surg 1984;140:802-806. 

Brown MF, Graham JM, Feliciano DV, et al: Carotid artery injuries. Am J Surg 
1982;144:748-753. 

Brown OW, Kerstein MD: Injuries to the peripheral vasculature: In Manage- 
ment of Vascular Trauma. Baltimore, University Park Press, 1985. 

Brown PS Jr, Nath R, Votapka T, et al: Traumatic ventricular septal defect and 
disruption of the descending thoracic aorta. Ann Thorac Surg 1991 ;52: 143- 
144. 

Brown RS, Boyd DR, Masuda T, Lowe R: Temporary internal vascular shunt for 
retrohepatic vena cava injury. J Trauma 1971;11:736-737. 



RE.qxd 4/16/04 3:19 PM Page 554 



554 VI • THE RICH REFERENCE COLLECTION 



Brunner JH, Stanley RJ: Superior mesenteric arteriovenous fistula. JAMA 

1973;223:316. 
Brunsting LA, Ouriel K: Traumatic fracture of the abdominal aorta: Rupture 

of a calcified abdominal aorta with minimal trauma.J Vase Surg 1988;8:184- 

186. 
Bryant MF Jr, Lazenby WD, Howard JM: Experimental replacement of short 

segment of veins. Arch Surg 1958;76:289-293. 
Brzowski BK, Mills JL, Beckett M, Beckett WC: Iatrogenic subclavian artery 

pseudoaneurysms: Case reports. J Trauma 1990;30:1-3. 
Buchbinder D, Karmody A, Leather R, Shah D: Hypertonic mannitol: Its use 

in prevention of revascularization syndrome after acute arterial ischemia. 

Arch Surg 1981;116:414-421. 
Buchman RJ, Thomas PAJ, Park B: Carotid artery injuries: Follow-up of fifteen 

patients treated in Vietnam. Angiology 1972;23:97-102. 
Buchmaster MJ, Kearney PA, Johnson SB, et al: Further experience with trans- 
esophageal echocardiography in the evaluation of thoracic aortic injury. 

J Trauma 1994;37:989-995. 
Buchness MP, Lo Gerfo FW, Mason GR: Gunshot wounds of the suprarenal 

abdominal aorta. Am Surg 1976;42:1-7. 
Buckman RF, Badellino MM, Mauro LH, AsensioJA: Penetrating cardiac wounds: 

Prospective study of factors influencing initial resuscitation. J Trauma 

1993;34:717-727. 
Buckner F, Lyons C, Perkins R: Management of lacerations of the great vessels 

of the upper thorax and base of the neck. Surg Gynecol Obstet 1958;107:135- 

142. 
Buechter KH, Gomez GA, Zeppa R: A new technique for exposure of injuries 

at the confluence of the retrohepatic veins and the retrohepatic vena cava. 

J Trauma 1990;30:328-331. 
Buechter KJ, Sereda D, Gomez G, Zeppa R: Retrohepatic vein injuries: Expe- 
rience with twenty cases. J Trauma 1989;29:1698-1704. 
BuntTJ: Iatrogenic vascular injury: A discourse on surgical technique. Mt. Kisco, 

Futura Publishing, 1990. 
Bunt TJ, MaloneJM, Moody M, et al: Frequency of vascular injury with blunt 

trauma-induced extremity injury. Am J Surg 1190;160:226. 
Bunt TJ, Manship L, Moore W: Iatrogenic vascular injury during peripheral 

revascularization. J Vase Surg 1985;2:491-498. 
Burch JM, Brock JC, Gevirtzman L, et al: The injured colon. Ann Surg 

1986;203:701-711. 
Burch JM, Feliciano DV, Mattox KL: The atriocaval shunt: Facts and fiction. 

Ann Surg 1988;207:555-568. 
Burch JM, Feliciano DV, Mattox KL: Colostomy and drainage for civilian rectal 

injuries: Is that all? Ann Surg 1989;209:600-611. 
Burch JM, Feliciano DV, Mattox KL, Edelman M: Injuries of the inferior vena 

cava. Am J Surg 1988;156:548-552. 
Burch JM, Richardson RJ, Martin RR, Mattox KL: Penetrating iliac vascular 

injuries: Recent experience with 233 consecutive patients. J Trauma 

1990;30:1450-1459. 
Burihan I, Pepe EVA, Miranda R: Bullet embolism following gunshot wound 

of the chest. J Cardiovasc Surg 1980;21:711-716. 
Burnett HF, Parnell CL, Williams GD, Campbell GS: Peripheral arterial injuries: 

A reassessment. Ann Surg 1976;183:701-709. 



RE.qxd 4/16/04 3:19 PM Page 555 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 555 

BurneyRE, Gundry SR, MacKenzieJR, etal: Comparison of mediastinal width, 
mediastinal-thoracicandcardioratios, and "mediastinal widening" in detec- 
tion of traumatic aortic rupture. Ann Emerg Med 1983;12:668-671. 

Burney RE, Gundry SR, MacKenzie JR, et al: Chest roentgenograms in diag- 
nosis of traumatic rupture of the aorta. Chest 1984;85:605-609. 

Burns GR, Sherman RT: Trauma of the abdominal aorta and inferior vena cava. 
Am Surg 1972;38:303-306. 

Burnsed DW, Weiss JB, Campbell GS, William GD: The relative merits of the 
heparin-bonded shunt vs. femoral bypass of aortic arch injury. Surgery 
1975;78:176-180. 

Burrell HL: Ligature of the innominate artery with a report of a case. Am Soc 
A1895;13:291. 

Burrows PE, Tubman DE: Multiple extracranial arterial lesions following closed 
cranial facial trauma. J Trauma 1981;21:497-498. 

Buscaglia LC, Blaisdell FW, Lim RCJr: Penetrating abdominal vascular injuries. 
Arch Surg 1969;99:764-769. 

Buscaglia LC, Matolo N, Macbeth A: Common iliac artery injury from blunt 
trauma: Case reports. J Trauma 1989;29:697-699. 

Busuttil RW, Kitahama A, Cerise E, et al: Management of blunt and penetrat- 
ing injuries to the porta hepatis. Ann Surg 1980;191:641-648. 

Byrne DE, Pass HI, Crawford FAJr: Traumatic vena caval injuries. Am J Surg 
1980;140:600-602. 

Cabrera A, Palomar S, Pastor E: Aortic rupture in a child diagnosed by cross 
sectional echocardiography. IntJ Cardiol 1987;15:249-252. 

Caes LF, Cham B, Sacre J, et al: A patient with two chronic posttraumatic 
aneurysms of the thoracic aorta. Thorac Cardiovasc Surg 1989;37:105-106. 

Caldeira CCB, Velmahos GC, Modrall JG: Acute suprarenal aortogastric fistu- 
lae caused by gunshot wounds: The importance of early recognition in 
directing surgical strategy. J Trauma 1996;40:838-839. 

CalhoonJH, Hoffmann TH, TrinkleJK, et al: Management of blunt rupture of 
the heart. J Trauma 1986;26:495-502. 

Callander CL: Study of arteriovenous fistula with an analysis of 447 cases. Ann 
Surg 1920;19:428-459. 

Cameron HS, Laird JJ, Carroll SE: False aneurysms complicating closed frac- 
tures. J Trauma 1972;12:67-74. 

Cammack K, Rapport R, Paul J, Baird WC: Deceleration injuries of the thoracic 
aorta. Arch Surg 1959;79:244-251. 

Campbell DK, Austin RF: Seat belt injury: Injury of the abdominal aorta. Radi- 
ology 1969;92:123-124. 

Campt PC, Rogers FB, Shackford SR, et al: Blunt traumatic thoracic aortic 
lacerations in the elderly: An analysis of outcome. J Trauma 1994;37: 
418-425. 

Capone AC, Safar P, Stezoski W, et al: Improved outcome with fluid restriction 
in treatment of uncontrolled hemorrhagic shock. J Am Coll Surg 
1995;180:49-56. 

Cappelen A: Vulnus cordis, sutur of hjrtet. Nor Mag f Laegevidensk 1 896;1 1 :285- 
288. 

Carden DL, Granger DN: Pathophysiology of ischaemia-reperfusion injury. 
J Pathol 2000;190:255-266. 

Cargile JS III, Hunt JL, Purdue GF: Femoral vascular trauma: A review of 218 
cases. J Trauma 1991;31:1025. 



RE.qxd 4/16/04 3:19 PM Page 556 



556 VI • THE RICH REFERENCE COLLECTION 



Cargile JS III, Hunt JL, Purdue GF: Acute trauma of the femoral artery and 

vein. J Trauma 1992;32:364-371. 
Carlsson E, Silander T: Rupture of the subclavian and innominate artery due 

to nonpenetrating trauma of the chest. Acta Chir Scand 1963;125:294-300. 
Carrasquilla C, Weaver AW: Aneurysm of the saphenous graft to the common 

carotid artery. Vase Surg 1972;6:66-68. 
Carrasquilla C, Wilson RE Walt AJ, Arbulu A: Gunshot wounds of the heart. 

Ann Thorac Surg 1972;13:208-213. 
Carrel A: La technique operatories des anastomoses vasculaires et la trans- 
plantation des visceres. Lyon Medical 1902;98:859. 
Carrel A: The surgery of blood vessels. Bull Johns Hopkins Hosp 1907;18:18. 
Carrel A: Results of transplantation of blood vessels, organs and limbs. JAMA 

1908;51:1662-1667. 
Carrel A, Guthrie CC: Uniterminal and biterminal venous transplantations. 

Surg Gynecol Obstet 1906;2:266-286. 
Carroll PR, McAninch JW, Klosterman P, Greenblatt M: Renovascular trauma: 

Risk assessment, surgical management, and outcome.J Trauma 1990;30:547- 

554. 
Carter BN, DeBakey ME: Current observations on war wounds of the chest. 

J Thorac Surg 1944;13:271-293. 
Carter SL, McKenzie JG, Hess DR: Blunt trauma to the common femoral artery. 

J Trauma 1981;21:178-179. 
Cass AS, Luxemberg M: Management of renal artery injuries from external 

trauma. J Urol 1987;138:266-268. 
Cass AS, Luxemberg M: Traumatic thrombosis of a segmental branch of the 

renal artery. J Urol 1987;137:1115-1116. 
Catinella FP, De Laria GA, De Wald RL: False aneurysm of the superior gluteal 

artery: A complication of iliac crest bone grafting. Spine 1990;15:1360. 
Catoire P, Bonnet F, Delaunay L, et al: Traumatic laceration of the ascending 

aorta detected by transesophageal echocardiography. Ann Emerg Med 

1994;23:356-359. 
Catoire P, Orliaguet G, Liu N, et al: Systematic transesophageal echocardio- 
graphy for detection of mediastinal lesions in patients with multiple injuries. 

J Trauma 1995;38:96-102. 
Caudros CL, Hutchinson JE, Mogtader AH: Laceration of a mitral papillary 

muscle and the aortic root as a result of blunt trauma to the chest. J Thorac 

Cardiovasc Surg 1984;88:134-140. 
Cerino M, McGrawJY, LukeJC: Autogenous vein graft replacement of throm- 
bosed deep veins: Experimental approach to the treatment of the post 

phlebitic syndrome. Surgery 1964;55:123-134. 
Cernaianu AC, Cilley JH Jr, Baldino WA, et al: Determinants of outcome in 

lesions of the thoracic aorta in patients with multiorgan system trauma. 

Chest 1992;101:331-335. 
Cernaianu AC, Olah A, Cilley JH Jr, et al: Effect of sodium nitroprusside on 

paraplegia during cross-clamping of the thoracic aorta. Ann Thorac Surg 

1993;56:1035-1037. 
Chaikof EL, Shamberger RC, Brewster DC: Traumatic pseudoaneurysms of the 

abdominal aorta. J Trauma 1985;25:169-173. 
Champion HR, Sacco WJ, Carnazzo AJ, et al: Trauma score. Crit Care Med 

1981;9:672-676. 
Champion MC, Sullivan SN, Coles JC, et al: Aortoenteric fistula: Incidence, 

presentation, recognition and management. Ann Surg 1982;195:314-317. 



RE.qxd 4/16/04 3:19 PM Page 557 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 557 

Chan EL, Bardin JA, Bernstein EF: Inferior vena cava bypass. J Vase Surg 
1984;1:675-680. 

Chandler JG, Knapp RW: Early definitive treatment of vascular injuries in the 
Vietnam conflict. JAMA 1967;202:960-966. 

Chang FC, Harrison PB, Beech RR, Helmer SD: PASG: Does it help in the man- 
agement of traumatic shock? J Trauma 1995;39:453-456. 

Chang H, Chu SH, Lee YT: Traumatic aorto-right arterial fistula blunt chest 
injury. Ann Thorac Surg 1989;47:778-779. 

Chapman AJ, McClain J: Wandering missiles: Autopsy study. J Trauma 
1984;24:634-637. 

Charles KP, Davidson KG, Miller H, Caves PK: Traumatic rupture of the ascend- 
ing aorta and aortic valve following blunt chest trauma. J Thorac Cardio- 
vasc Surg 1977;73:208-211. 

Chase CW, Layman TS, Barker DE, Clements JB: Traumatic abdominal aortic 
pseudoaneurysm causing biliary obstruction: A case report and review of 
the literature. J Vase Surg 1997;25:936-940. 

Chase MD, Schwartz SI: Suture anastomosis of small arteries. Surg Gynecol 
Obstet 1963;117:44-46. 

Chaudry IH: Cellular mechanisms in shock and ischemia and their correction. 
Am J Physiol 1983;245:R117-R134. 

Chaudry IH: ATP-MgC12 and liver blood flow following shock and ischemia. 
Prog Clin Biol Res 1989;299:19-31. 

Chaudry IH: The use of ATP following shock and ischemia. In Dubyak BR, 
FedanJS (eds): Biological Actions of Extracellular ATP. Ann NY Acad Sci, 
1990: 130-141. 

Chaudry IH, Ohkawa M, Clemens MG: Improved mitochondrial function 
following ischemia and reflow by ATP-MgCL. Am J Physiol 
1984;246:R799-R804. 

Chaudry IH, Ohkawa M, Clemens MG, Baue AE: Alterations in electron trans- 
port and cellular metabolism with shock and trauma. Prog Clin Biol Res 
1983;111:67-88. 

Chedid MK, Deeb ZL, Rothfus WE, et al: Major cerebral vessel injury caused 
by a seatbelt strap: Case report. J Trauma 1989;29:1601-1603. 

Cheek RC, Pope JC, Smith HF, et al: Diagnosis and management of major 
vascular injuries: A review of 200 operative cases. Am Surg 1975;41:755- 
760. 

Cherng WJ, Bullard MJ, Chang HJ, Lin FC: Diagnosis of coronary artery dis- 
section following blunt chest trauma by transesophageal echocardiogra- 
phy. J Trauma 1995;39:772-774. 

CherryJK, Bennett DE: False aneurysm of the aorta following penetrating trauma: 
Report of a case with secondary infection and delayed rupture. Arch Surg 
1966;93:404-408. 

Chervu A, Quinones-Baldrich WJ: Vascular complications in orthopaedic surgery. 
Clin Orthop 1988;235:275-288. 

Chew FS, Panicek DM, Heitzman ER: Late discovery of a posttraumatic right 
aortic arch aneurysm. AJR Am J Roentgenol 1985;145:1001-1002. 

Childs D, Wilkes RG: Puncture of the ascending aorta. A complication of sub- 
clavian venous cannulation. Anesthesia 1986;41:331-332. 

Chitwood W, Austin E: Cardiac trauma: Penetrating and blunt. In Trauma 
Surgery. Philadelphia, JB Lippincott Co, 1988. 

Chiu CL, Roelofs JD, Go RT: Coronary angiographic and scintigraphic find- 
ings in experimental cardiac contusion. Radiology 1975;116:679-683. 



RE.qxd 4/16/04 3:19 PM Page 558 



558 VI • THE RICH REFERENCE COLLECTION 



Choo MH, Chia BL, Chia FK: Penetrating cardiac injury evaluated by two- 
dimensional echocardiography. Am HeartJ 1984;108:417-420. 

Ciaravella JM, Ochsner JL, Mills NL: Traumatic avulsion of the innominate 
artery: A case report and literature review. J Trauma 1976;16:751-754. 

Cikrit DF, Dalsing MC, Bryant BJ, Lalka SG: An experience with upper- 
extremity vascular trauma. Am J Surg 1990;160:229. 

Cioffi WG Jr, Pruitt BA Jr: Vascular abnormalities associated with thermal 
and electrical trauma. In Flanigan DP (ed): Civilian Vascular Trauma. 
Philadelphia, Lea & Febiger, 1992, pp 255-264. 

Clark DE, Georgia JW, Ray FS: Renal arterial injuries caused by blunt trauma. 
Surgery 1981;90:87-96. 

Clark DE, Zeiger MA, Wallace KL, et al: Blunt aortic trauma: Signs of high risk. 
J Trauma 1990;30:701-705. 

Clarke CP, Brandt PW, Cole DS, Barratt-Boyes BG: Traumatic rupture of the 
thoracic aorta: Diagnosis and treatment. Br J Surg 1967;54:353-358. 

Clarke P, Whittaker M: Traumatic aneurysm of the internal carotid artery and 
rupture of the duodenum following seatbelt injury. Injury 1980;12:158- 
160. 

Clemens MG, McDonagh PF, Chaudry IH, Baue AE: Hepatic microcirculatory 
failure afterischemia and reperfusion: improvement with ATP-MgCl 2 treat- 
ment. Am J Physiol 1985;248:H804-H811. 

Clermont G: Suture laterale et circulaire des veines. Presse Med 1901;1: 
229. 

Cleveland JC, Cleveland RJ: Successful repair of aortic root and aortic valve 
injury caused by blunt chest trauma in a patient with prior aortic dissec- 
tion. Chest 1974;66:447-450. 

Cleveland JC, Ellis J, DagueJ: Complete disruption of axillary artery caused 
by severe atherosclerosis and trivial non-penetrating trauma. J Trauma 
1979;19:635-636. 

Clyne CAC, Ashbrooke EA: Seat-belt aorta: Isolated abdominal aortic injury 
following blunt injury. Br J Surg 1985;72:239. 

Coelho JC, Siegel B, Flanigan DP, et al: Arteriographic and ultrasonic evalua- 
tion of vascular clamp injury using an in vitro human experimental model. 
Surg Gynecol Obstet 1982;155:506-512. 

Cogbill TH, Moora EE, Jurkovich GJ, et al: Severe hepatic trauma: A multi- 
center experience with 1,335 liver injuries. J Trauma 1988;28:1433-1438. 

Cogbill TH, Moore EE, Milikan JS, Cleveland HC: Rationale for selective appli- 
cation of emergency department thoracotomy in trauma. J Trauma 
1983;23:453-460. 

Cohen A, Baldwin JN, Grant RN: Problems in the management of battlefield 
vascular injuries. Am J Surg 1969;118:526-530. 

Cohen A, Brief D, Mathewson CJr: Carotid artery injuries: An analysis of 85 
cases. Am J Surg 1970;120:210-214. 

Cohen AM, Crass JR, Thomas HA, et al: CT evidence for the "osseous pinch" 
mechanism for traumatic aortic injury. AJR Am J Roentgenol 1992; 159:271- 
274. 

Cohn SM, Burns GA,Jaffe C, Milner KA: Exclusion of aortic tear in the unsta- 
ble trauma patient: The utility of transesophageal echocardiography. 
J Trauma 1995;39:1087-1090. 

Coimbra R, Pinto MC, Razuk A: Penetrating cardiac wounds: Predictive value 
of trauma indices and the necessity of terminology of standardization. Am 
Surg 1995;61:448-452. 



RE.qxd 4/16/04 3:19 PM Page 559 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 559 

Coin D, Crighton J, Schorn L: Successful management of hepatic vein injury 

from blunt trauma in children. Am J Surg 1980;140:858. 
Coley RW. Case of rupture of the carotid artery and wound of several of its 

branches successfully treated by tying off the common trunk of the carotid 

itself. Med ChirJ (Lond) 1817;3:2. 
Collicott PE: Initial assessment of the trauma patient. In Mattox KL, Moore EE, 

Feliciano DV (eds) : Trauma. EastNorwalk, Conn, Appleton & Lange, 1988, 

pp 107-124. 
Collins HA, Jacobs JK: Acute arterial injuries due to blunt trauma. J Bone Joint 

Surg 1961;43-A:193-197. 
Collins JA: Recent developments in the area of massive transfusion. World J 

Surg 1987;11:75-81. 
Collins PS, Golocvsky M, Salander JM, et al: Intra-abdominal vascular 

injury secondary to penetrating trauma. J Trauma 1988;28:S165- 

S168. 
Collins RE, Douglass FM: Small vein anastomosis with and without operative 

microscope: A comparative study. Arch Surg 1964;88:740-742. 
Colon R, Frazier OH, Cooley DA, McAllistr HA: Hypothermic regional perfu- 
sion for protection of the spinal cord during periods of ischemia. Ann 

Thorac Surg 1987;43:639-643. 
ConeJB: Vascular injury associated with fracture-dislocations of the lower extrem- 
ity. Clin Orthop 1989:30. 
Conkle DM, Richie RE: Surgical treatment of popliteal artery injuries. Arch 

Surg 1975;110:1351-1353. 
Conn J, Trippel OH, Bergan JJ: A new atraumatic aortic occluder. Surgery 

1968;64:1158. 
ConnJH, Hardy JD, Chavez CM, Fain WR: Challenging arterial injuries. J Trauma 

1971;11:167-177. 
Connelly JE, Brownell DA, Levine EF: Complications of renal dialysis access 

procedures. Arch Surg 1984;119:1325-1328. 
Connery C, Geller E, Dulchavsky S, Kreis DJ: Paraparesis following emergency 

room thoracotomy: Case report. J Trauma 1990;30:362-363. 
Connolly J, Williams E, Whittaker D: The influence of fracture stabilization on 

the outcome of arterial repair in combined fracture-arterial injuries. Surg 

Forum 1969;20:450-452. 
ConnollyJ: Management of fractures associated with arterial injuries. Am J Surg 

1970;120:331. 
Connolly JE: Prevention of paraplegia secondary to operations on the aorta. 

J Cardiovasc Surg 1986;27:410-417. 
Connolly JE, KwaanJHM, McCart PM: Complications after percutaneous trans- 
luminal angioplasty. Am J Surg 1981;142:60-66. 
Connolly JF, Whittaker D, Williams E: Femoral and tibial fractures combined 

with injuries to the femoral or popliteal artery. J Bone Joint Surg 1971;53- 

A56-67. 
Conti VR, Calverley J, Safley WL, et al: Arterial spinal artery syndrome with 

chronic traumatic thoracic aortic aneurysm. Ann Thorac Surg 1982;33:81- 

85. 
Contino JP, Follette DM, Berkoff HA, et al: Use of Carmeda-coated femoral- 
femoral bypass repair of traumatic aortic pseudoaneurysm. Arch Surg 

1994;129:933-937. 
Cook FW, Haller JAJr: Penetrating injuries of the subclavian vessels associated 

with venous complications. Ann Surg 1962;155:370-372. 



RE.qxd 4/16/04 3:19 PM Page 560 



560 VI • THE RICH REFERENCE COLLECTION 



Cook P, Erdoes L, Selzer P: Dissection of the external iliac artery in highly 

trained athletes. J Vase Surg 1995;22:173. 
Cook TA, Jones AJ, Webb AK, Baird RN: Traumatic abdominal aortic aneurysm. 

Eur J Vase Surg 1994;8:364-365. 
Cooley DA, Dunn JR, Brockman HL, DeBakey ME: Treatment of penetrating 

wounds of the heart: Experimental and clinical observations. Surgery 

1955;37:882. 
Cooper B: Wounds in arteries and traumatic aneurysms. Guys Hosp Rep 

1852;8:195. 
Cope C, Zeit R: Coagulation of aneurysms by direct percutaneous thrombin 

injection. AJR Am J Roentgenol 1986;147:383-387. 
Copper ES: Aneurysm of the right carotid and subclavian arteries. Am J Med 

Sci 1859;38:395. 
Cornwell EE, Kennedy F, Berne TV, et al: Gunshot wounds to the thoracic aorta 

in the '90s: Only prevention will make a difference. Am Surg 1995;61:721- 

723. 
Cosby RL, Miller PD, Schirir RW: Traumatic renal artery thrombosis. Am J Med 

1986;81:890-894. 
CoselliJS, Crawford ES: Surgical treatment of aneurysms of the intrathoracic 

segment of the subclavian artery. Chest 1987;91:704-708. 
Costa M, Robbs JV: Management of retroperitoneal haematoma following 

penetrating trauma. Br J Surg 1985;72:662-664. 
Costa MC, Robbs JV: Nonpenetrating subclavian artery trauma. J Vase Surg 

1988;8:71-75. 
Couch NP, Cerundolo D: A clip for the rapid control of the vascular injuries. 

Surg Gynecol Obstet 1981;153:408-409. 
Courcy PA, Brotman S, Oster-Granite ML, et al: Superior mesenteric artery and 

vein injury from blunt abdominal trauma. J Trauma 1984;24:843-845. 
Couves CM, Lumpkin MB, Howard JM: Arterial injuries due to blunt- 

nonpenetrating trauma: Experiences with 15 patients. Can J Surg 1958; 

1:197-200. 
Cowgill LD, Campbell DN, Clarke DR, et al: Ventricular septal defect due to 

non-penetrating chest trauma: Use of the intra-aortic balloon pump. 

J Trauma 1987;27:1087-1090. 
Cranley JJ, Krause RJ: Injury to the axillary artery following anterior disloca- 
tion of the shoulder. Am J Surg 1958;95:524-526. 
Crawford ES, FenstermacherJM, Richardson W, Frank S: Reappraisal of adjuncts 

to avoid ischemia in the treatment thoracic aortic aneurysms. Surgery 

1970;67:182-196. 
Crawford ES, Mizrahi EM, Hess KR, et al: The impact of distal aortic perfusion 

and somatosensory evoked potential monitoring on prevention of 

paraplegia after aortic aneurysm operation. J Thorac Cardiovasc Surg 

1988;95:357-367. 
Crawford ES, Morris GCJr, Myhre HO, Roehm JO: Celiac axis, superior mesen- 
teric artery and inferior mesenteric artery occlusion: considerations. Surgery 

1977;82:856-866. 
Crawford ES, Svensson LG, Hess KR, et al: A prospective randomized study of 

cerebrospinal fluid drainage to prevent paraplegia after high-risk surgery 

on the thoracoabdominal aorta. J Vase Surg 1991;13:36-46. 
Creagh TA, Broe PJ, Grace PA, Bouchier-Hayes DJ: Blunt trauma induced upper 

extremity vascular injuries. J R Coll Surg Edinb 1991;36:158. 
Creech O Jr: Acute arterial injuries. Postgrad Med 1961;29:581. 



RE.qxd 4/16/04 3:19 PM Page 561 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 561 

Creech O Jr, Gantt J, Wren H: Traumatic arteriovenous fistula at unusual sites. 

Ann Surg 1965;161:908-920. 
CreppsJTJr, Rodriguez A: Combined abdominal aortic and visceral artery injury 

secondary to blunt trauma. Injury 1989;20:176-177. 
Crissey MM, Bernstein EF: Delayed presentation of carotid intimal tear following 

blunt craniocervical trauma. Surgery 1974;75:543-549. 
CronenwettJL, Walsh DB, Garrett HE: Tibial artery pseudoaneurysms: delayed 

complication of balloon catheter embolectomy. J Vase Surg 1988;8:483- 

488. 
CronenwettJL, McDaniel M, Zwolak R, et al: Limb salvage despite extensive 

tissue loss: Free tissue transfer combined with distal revascularization. Arch 

Surg 1989;124:609-615. 
Cryer HM, Miller FB, Evers M, et al: Pelvic fracture classification: Correlation 

with hemorrhage. J Trauma 1988;28:973-980. 
Cterctko G, Mok CK: Aorto-esophageal fistula induced by a foreign body: The 

first recorded survival. J Thorac Cardiovasc Surg 1980;80:233-235. 
Curley SA, Demarest G, Hauswald M: Pericardial tamponade and hemothorax 

after penetrating injury to the internal mammary artery. J Trauma 

1987;27:957-958. 
Currarino G, Engle ME: The effects of ligation of the subclavian artery on the 

bones and soft tissues of the arms. J Pediatr 1965;67:808-811. 
Curtin JJ, Goodman LR, Thorsen MR: Aortic traumatic lesion: Has tomoden- 

sitometry a role? J Radiol 1993;74:523-530. 
CusickJF, Daniels D: Spontaneous reversal of internal carotid artery occlusion: 

Case report. J Neurosurg 1981;54:811-813. 
DainkoEA: Complications of the use of the Fogarty balloon catheter. Arch Surg 

1972;105:79-82. 
Dajani OM, Haddad FF, Hajj HA, et al: Injury to the femoral vessels: The Lebanese 

War experience. Eur J Vase Surg 1988;2:293-296. 
Dajee H, Richardson IW, Iype MO: Seatbelt aorta: Acute dissection and throm- 
bosis of the abdominal aorta. Surgery 1979;85:263-267. 
Dale WA: Chronic iliofemoral venous occlusion including seven cases of cross- 
over vein grafting. Surgery 1966;59:117-132. 
Dale WA: Management of arterial occlusive disease (national conference on 

management of arterial occlusive disease, Naashville, 1970). In Dale WA 

(ed): Chicago, Year Book Publisher Inc, 1971. 
Dale WA: The beginnings of vascular surgery. Surgery 1974;76:849-866. 
Dale WA, Harris J, Terry RB: Polytetrafluoroethylene reconstruction of the 

inferior vena cava. Surgery 1984;95:625-630. 
Dalton HC: Report of a case of stab wound of the pericardium, terminating in 

recovery after resection of a rib and suture of the pericardium. Ann Surg 

1895;21:147-152. 
Dameron TB Jr: False aneurysm of femoral profundus artery resulting 

from internal-fixation device (screw). J Bone Joint Surg 1964;46A:577- 

580. 
Damron T, McBeath A: Diagnosis and management of vascular injuries associ- 
ated with skeletal trauma. Orthop Rev 1990;19:1063. 
Dana A, Skarli M, Papakrivopoulou J, Yellon DM: Adenosine A(l) receptor 

induced delayed preconditioning in rabbits: Induction of p38 mitogen- 

activated protein kinase. Circ Res 2000;86:989-997. 
Danielson GK, Wood R, Holloway JB: Traumatic avulsion of the innominate 

artery from the aorta. Ann Thorac Surg 1968;5:451-458. 



RE.qxd 4/16/04 3:19 PM Page 562 



562 VI • THE RICH REFERENCE COLLECTION 



Danto LA, Wolfman EFJr: Linear abdominal trauma. J Trauma 1976;16:179- 

183. 
Danza R, Mauro L, Arias J: Reconstruction of the femoro-popliteal vessels with 

a double graft (arterial and venous) in severe injury of the limb. J Cardiac 

Surg 1970;11:60-64. 
Dart CH Jr, Braitman HE: Traumatic rupture of the thoracic aorta: diagnosis 

and management. Arch Surg 1976;111:697-702. 
Dart CH Jr, Braitman HE: Popliteal artery injury following fracture or disloca- 
tion at the knee: Diagnosis and management. Arch Surg 1977;112:969-973. 
David D, Blumenberg RM: Subintimal aortic dissection with occlusion after 

blunt abdominal trauma. Arch Surg 1970;100:302-304. 
Davie JC, Coxe W: Occlusive disease of the carotid artery in children. Arch 

Neurol 1967;17:313-323. 
Davies MG, Hagen PO: The vascular endothelium: A new horizon. Ann Surg 

1993;218:593-609. 
Davis. 1834. Cited in: Straus R. Pulmonary embolism caused by lead bullet fol- 
lowing gunshot wound of abdomen. Arch Path 1942;33:63-68. 
Davis JM, Zimmerman RA: Injury of the carotid and vertebral arteries. Neuro- 
radiology 1983;25:55-69. 
Davis TP, Feliciano DV, Rozycki GS: Results with abdominal vascular trauma in 

the modern era. Am Surg 2001;67:565-571. 
Dawson D, Johanse K, Jurkovich G: Injuries to the portal triad. Am J Surg 

1991;161:545. 
Daxini BV, Desai AG, Sharma S: Echo-Doppler diagnosis of aortocaval fistula 

following blunt trauma to abdomen. Am HeartJ 1989;118:843-844. 
de Guzman E, Shankar MN, Mattox KL: Limited volume resuscitation in 

penetrating thoracoabdominal trauma. AACN Clin Issues 1999;10:61-68. 
De Nayer P, Jaumin P, Linard D: Lesions osteo-articulaires des membres com- 

pliquees de traumatismes vascularies. Acta Chir Belg 1973;72:427. 
De Takats G: Vascular surgery in the war. War Med 1943;3:291-296. 
De Takats G: Vascular Surgery. Philadelphia, WB Saunders, 1959. 
De Takats G: Symptoms and signs of peripheral arterial disease. Med Clin North 

Am 1962;46:647-657. 
De Takats G: Trauma to the arteriosclerotic limb. J Trauma 1963;4:47. 
De Takats G, Pirani C: Aneurysms: General considerations. Angiology 1954;5: 1 73- 

208. 
De Wet LubbeJJ, Janson PM: Penetrating wounds of the heart and great vessels: 

Experience with cases including three with intracardiac defects. South Afr 

MedJ 1975;29:512-516. 
Dean RH: Management of renal artery trauma. J Vase Surg 1988;8:89-90. 
DeBakey ME, Simeone FA: Battle injuries of the arteries in World War II: An 

analysis of 2,471 cases. Ann Surg 1946;123:534-579. 
DeBakey ME, Cooley DA, Morris GCJr, Collins H: Arteriovenous fistula involv- 
ing the abdominal aorta: Report of four cases with successful repair. Ann 

Surg 1958;147:646-658. 
DeBakey ME, Beall ACJr, WukaschDC: Recent developments in vascular surgery 

with particular reference to orthopaedics. Am J Surg 1 965; 109: 134- 

142. 
DeBakey ME: The development of vascular surgery. Am J Surg 1979;137:697- 

738. 
Defore WWJr, Mattox KL, Jordan GLJr: Management of 1590 consecutive cases 

of liver trauma. Arch Surg 1976;111:493-497. 



RE.qxd 4/16/04 3:19 PM Page 563 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 563 

Degianni E, Levy RD, Sofianos C, et al: Arterial gunshot injuries of the extrem- 
ities: A South African experience. J Trauma 1995;39:570-575. 

DelRossi AJ, Cernaianu AC, Cilley JH Jr, et al: Multiple traumatic disruptions 
of the thoracic aorta. Chest 1990;97:1307-1309. 

DelRossi AJ, Cernaianu AC, Madden LD, et al: Traumatic disruptions of the 
thoracic aorta: Treatment and outcome. Surgery 1990;108:864-870. 

DeLucia A III, Fromm D: Retropancreatic control of the suprarenal aorta. Surg 
Gynecol Obstet 1988;166:475-476. 

Demetriades D: Cardiac wounds. Ann Surg 1986;203:315-317. 

Demetriades D, Rabinowitz B, Pezikis A, et al: Subclavian vascular injuries. Br 
J Surg 1987;74:1001-1003. 

Demetriades D, SkalkidesJ, Safianos C: Carotid artery injuries: Experience with 
124 cases. J Trauma 1989;29:91-94. 

Corrected 

Demetriades D, Charalam bides L, Sareli P: Later sequelae of penetrating cardiac 
injuries. Br J Surg 1990;77:813-814. 

Demetriades D, Theodorou D, Murray J, et al: Mortality and prognostic factors 
in penetrating injuries of the aorta. J Trauma 1996;40:761-763. 

Demetriades D, Chan L, Cornwell E, et al: Paramedic vs. private transportation 
of trauma patients. Arch Surg 1996;131:133-138. 

DeMeulesJE, Cramer G, Perry JFJr: Rupture of aorta and great vessels due to 
blunt thoracic trauma. J Thorac Cardiovasc Surg 1971;61:438-442. 

DeMuth WE: Bullet velocity makes the difference. J Trauma 1969;9:642- 
643. 

DeMuth WE Jr, Baue AE, Odom JA Jr: Contusions of the heart. J Trauma 
1967;7:443-445. 

DeMuth WE Jr, Roe H, Hobbie W: Immediate repair of traumatic rupture of 
the aorta. Arch Surg 1965;91:602-603. 

Dennis JW, Frykberg ER, Crump JM, et al: New perspectives on the manage- 
ment of penetrating trauma in proximity to major limb arteries. J Vase Surg 
1990;11:85-93. 

Dent RI,Jena GP: Missile injuries of the abdomen in Zimbabwe-Rhodesia. Br 
J Surg 1980;67:305-310. 

DePriest W, Barish R, Almquist T, Groleau G: Echocardiographic diagnosis of 
acute pericardial effusion in penetrating chest trauma. Am J Emerg Med 
1988;6:21-23. 

DeSa'Neto A, Padnick MB, Desser KB, Steinhoff NG: Right sinus of Valsalva- 
right atrial fistula secondary to nonpenetrating chest trauma. Circulation 
1979;60:205-209. 

Desai SB, ModheJM, Aulakh BG: Percutaneous transcatheter steel-coil emboliza- 
tion of a large proximal post-traumatic superior mesenteric arteriovenous 
fistula. J Trauma 1987;27:1091. 

DeSaussure RL: Vascular injury coincident to disc surgery. J Neurosurg 
1959;16:222-228. 

DesChamps GT, Morano JU: Intracranial bullet migration: A sign of brain abscess: 
Case report. J Trauma 1991;31:293-295. 

DesForges G, Ridder WP, Lenoci RJ: Successful suture of ruptured myocardium 
after nonpenetrating injury. N Engl J Med 1955;252:567-569. 

Detwiler K, GoderskyJC, Gentry L: Pseudoaneurysm of the extracranial verte- 
bral artery. J Neurosurg 1987;67:935-939. 

DeWeese JA, Mattox KL, Seltzer SE, BukleyJM: Symposium on traumatic tho- 
racic aortic rupture. Contemp Surg 1984;24:109-131. 



RE.qxd 4/16/04 3:19 PM Page 564 



564 VI • THE RICH REFERENCE COLLECTION 



Dharker SR, Dharker RS: Traumatic occlusion of internal carotid artery in an 
infant. Surg Neurol 1978;9:77-78. 

Dichtel WJ, Miller RH, Woodson GE, et al: Lateral mandibulotomy: A tech- 
nique of exposure for penetrating injuries of the internal carotid artery at 
the base of the skull. Laryngoscope 1984;94:1140-1144. 

Dickerman RM, Gewertz BL, Foley DW, Fry WJ: Selective intra-arterial tolazo- 
line infusion in peripheral arterial trauma. Surgery 1977;81:605-609. 

Diebel LN, Wilson RF, Bennder J, Paules B: A comparison of passive and active 
shunting for bypass of retrohepatic IVC. J Trauma 1991;31:987-990. 

Dieter RA, Asselmeier GH, Hamouda F, et al: Traumatic disruption of the tho- 
racic aorta: A community hospital experience. Mil Med 1983;148:502-506. 

Dieux JJ, Milani H, Maillet P, Brahy R: A case of 2-stage traumatic rupture of 
the abdominal aorta. Acta Chir Belg 1985;85:367-370. 

Dillard BM, Nelson DL, Norman HG: Review of 85 major traumatic arterial 
injuries. Surgery 1968;63:391-395. 

Dillard BM, Staple TW: Bullet embolism from the aortic arch to the popliteal 
artery. Arch Surg 1969;98:326-328. 

Dillman RO, Crumb CK, Lidsky MJ: Lead poisoning from a gunshot wound. 
Am J Med 1979;66:509-514. 

DiMaio VJ, DiMaio DJ: Bullet embolism: Six cases and a review of the literature. 
J Forens Sci 1972;17:394-398. 

Dimond FC Jr, Rich NM: M-16 wounds in Vietnam. J Trauma 1967;7:618- 
625. 

Dingeldein GR Proctor HJ, Jaques PF: Traumatic aorto-caval-portal-duodenal 
fistula: Case report. J Trauma 1977;17:474-476. 

Diveley WL, Daniel RAJr, Scott HWJr: Surgical management of penetrating 
injuries of the ascending aorta arch. J Thorac Cardiovasc Surg 1961;41:23- 
33. 

DiVincenti FC, Weber BB: Traumatic carotid artery injuries in civilian practice. 
Am Surg 1974;40:277-280. 

Dixon RG, McEwan P: Notes on a case of penetrating wound of the heart. Br 
MedJ1916;l:755. 

Doan E, Gorton ME: Traumatic disruption of the innominate and right pul- 
monary arteries: Case report. J Trauma 19;43:701-702. 

Dobell ARC, MacNaughton EA, Crutchlow EF: Successful early treatment of 
subadventitial rupture of the thoracic aorta. N Engl J Med 1964;270:410- 
412. 

Dobrin PB: Mechanisms and prevention of arterial injuries caused by balloon 
embolectomy. Surgery 1989;106:457-466. 

Donahue TK, Strauch GO: Ligation as definitive management of injury to the 
superior mesenteric vein. J Trauma 1988;28:541-543. 

Donchin Y, Rivkind Al, Bar-ZivJ, et al: Utility of postmortem computed tomo- 
graphy in trauma victims. J Trauma 1994;37:552-556. 

Donell ST, Hudson MJK: Iatrogenic superior mesenteric arteriovenous fistula: 
Report of a case. J Vase Surg 1988;8:335. 

Donovan DL, Sharp WV: Blunt trauma to the axillary artery. J Vase Surg 
1984;1:681-683. 

Dorfman GS, Cronan ]J: Bayesian analysis of chest plain film finding in trau- 
matic rupture of the aorta. Invest Radiol 1987;22:S4. 

Dorion RP, Hamati HF, Landis B: Risk and clinical significance of developing 
antibodies induced by topical thrombin preparations. Arch Pathol Lab 
Med 1998;122:887-894. 



RE.qxd 4/16/04 3:19 PM Page 565 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 565 

Dosios TJ, Magoven GJ, Gay TC, Joyner CR: Cardiac tamponade complicating 
percutaneous catheterization of subclavian vein. Surgery 1975;78:261-263. 

Doty DB, Baker WH: Bypass of superior vena cava with spiral vein graft. Ann 
Thorac Surg 1976;22:490-493. 

Doty DB, Treiman RL, Rothschild PD, Gaspar MR: Prevention of gangrene due 
to fractures. Surg Gynecol Obstet 1967;125:284-288. 

Downs AR, MacDonald P: Popliteal artery injuries: Civilian experience with sixty- 
three patients during a twenty-four year period. J Vase Surg 1986;4:55-62. 

DorflerJ: Uber arteriennaht. Beitr Klin Chir 1889;25:781. 

Dragojevic D, Hetzer R, Oelert H: Surgery for traumatic rupture of the tho- 
racic aorta. Thorac Cardiovasc Surg 1980;28:436-440. 

Dragon R, Saranchak H, Lakin P, Strauch G: Blunt injuries to the carotid ver- 
tebral arteries. Am J Surg 1981;141:497-500. 

Drapanas T, Hewitt RL, Weichert RF III, Smith AD: Civilian vascular injuries: 
Acritical appraisal of three decades of management. Ann Surg 1970;172:351- 
360. 

Dronen SC, Stern SA, Wang X, Stanley M: A comparison of the response of 
near fatal acute hemorrhage mode with and with out a vascular injury to 
rapid volume expansion. Am J Emerg Med 1993;11:331-335. 

Drost TF, Rosemurgy AS, Proctor D, Kearney RE: Outcome of treatment of 
combined orthopedic and arterial trauma to the lower extremity. J Trauma 
1989;29:1331-1334. 

Drury JK, Scullion JE: Vascular complications of anterior dislocation of the 
shoulder. Br J Surg 1980;67:579-581. 

Dshanelidze II. Manuskript petrograd. 1922. Cited in: Lilianthal H: 1926. 

Du Toit DF, Greeff M: Low-velocity gunshot injury of the abdominal aorta 
managed by debridement and re-anastomosis. S Afr Med J 1986;18:139- 
140. 

Du Toit DF, Rademan F: Traumatic superior mesenteric arteriovenous fistula: 
A case report. S Afr Med J 1987;71:587. 

Dube P: Control of massive carotid hemorrhage by balloon catheter. Thorax 
1973;28:399-400. 

Dubinskiy MB: Suture of the abdominal aorta. Khirurgiya 1944;4:71. 

Duhaylongsod FG, Glower DD, Wolfe WG: Acute traumatic aortic aneurysm: 
The Duke experience from 1970 to 1990. J Vase Surg 1992;15:331-343. 

Dula DJ, Hughes HG, Majernick T: Traumatic disruption of the brachiocephalic 
artery. Ann Emerg Med 1983;12:639-641. 

Dulchavsky SA, Geller ER, Iorio DA: Analysis of injuries following the crash of 
Avianca flight 52. J Trauma 1993;34:282-284. 

Duncan A, Scalea T, Sclafani SJ: Evaluation of occult cardiac injuries using sub- 
xiphoid pericardial window. J Trauma 1989;29:955-960. 

Durham LA, Richardson RJ, Wall MJ: Emergency center thoracotomy impact 
of prehospital resuscitation. J Trauma 1992;32:775. 

Earle AS, HorsleyJS, Villavicencio JL, Warren R: Replacement of venous defects 
by venous autografts. Arch Surg 1960;80:119-124. 

Eastcott HH: The management of arterial injuries. J Bone Joint Surg 
1965;47B:394-398. 

Eastcott HHG: Arterial Surgery. Philadelphia, JB Lippincott Co, 1969. 

Eaves CC: Traumatic rupture of the thoracic aorta presenting as transient para- 
plegia. J Emerg Med 1990;8:429-431. 

EckNV: K voprosu o perevyazkie vorotnoisveni. Predvaritelnoye soobshtshjenye 
(Ligature of the portal vein). Voen Med J 2001;130:1-2. 



RE.qxd 4/16/04 3:19 PM Page 566 



566 VI • THE RICH REFERENCE COLLECTION 



Eddy AC, Misbach GA, Luna GK: Traumatic rupture of the thoracic aorta in 

the pediatric patient. Pediatr Emerg Care 1989;8:429-431. 
Eddy AC, Nance DR, Goldman MA, et al: Rapid diagnosis of thoracic aortic 

transection using intravenous digital subtraction angiography. Am J Surg 

1990;159:500-503. 
Eddy AC, Rusch VW, Fligner CL, et al: The epidemiology of traumatic rupture 

of the thoracic aorta in children: A 13-year review. J Trauma 1990;30:989- 

992. 
Eddy AC, Rusch VW, Marchioro T, et al: Treatment of traumatic rupture of the 

thoracic aorta. Arch Surg 1990;125:1351-1356. 
Edmundson K: Transfixion of the aorta. Br J Surg 1963;23:869-870. 
Edwards WS, Lyons C: Traumatic arterial spasm and thrombosis. Ann Surg 

1957;140:318-323. 
Egan TJ, Neiman RJ, Malave SR, Sanders JH: Computed tomography in the 

diagnosis of aortic aneurysm dissection or traumatic injury. Radiology 

1980;136:141-146. 
Eger M, Glocman L, Goldstein A, Hirsch M: The use of temporary shunt in the 

management of arterial vascular injuries. Surg Gynecol Obstet 1971 ; 1 32:67- 

70. 
Eger M, Glocman L, Torok G, Hirsch M: Inadvertent arterial stripping in the 

lower limb: Problems of management. Surgery 1973;73:23-27. 
Eger M, Glocman L, Schmidt D, Hirsch M: Problems in the management of 

popliteal artery injuries. Surg Gynecol Obstet 1973;134:921-926. 
Eger M, Schmidt B, Torok G: Replantation of the upper extremities. Am J Surg 

1974;4:47-50. 
Ehrenfeld WK, Wilber BG, Olcott CN, Stoney RJ: Autogenous tissue recon- 
struction in the management of infected prosthetic grafts. Surgery 

1979;85:82-91. 
Eifert S, Villavicencio JL, Kao TC, et al: Prevalence of deep venous anomalies 

in congenital vascular malformations of venous predominance. J Vase Surg 

2000;31:462-471. 
Eiseman B, Rainer WG: Clinical management of post traumatic rupture of the 

thoracic aorta. J Thorac Cardiovasc Surg 1958;35:347-358. 
EisenbreyAB: Arteriovenous aneurysm of the superficial femoral vessels. JAMA 

1913;61:2155-2157. 
Ekbom B, Towne JB, Majewski JT, Woods JH: Intra-abdominal vascular trauma: 

A need for prompt operation. J Trauma 1981;21:1040-1044. 
Eklof B, GjoresJE, Thulesius O, Bergqvist D: Controversies in the Management 

of Venous Disorders. Stoneham, Butterworths, 1989. 
Elderding SC, Manart FD, Moore EE: A reappraisal of penetrating neck injury 

management. J Trauma 1980;20:695-697. 
Elford J, Burrell C, Roobottom C: Ultrasound guided percutaneous thrombin 

injection for the treatment of iatrogenic pseudoaneurysms. Heart 

1999;82:526-527. 
Elkin DC: Diagnosis and treatment of cardiac trauma. Ann Surg 1941;114:169- 

185. 
Elkin DC: Vascular injuries of warfare. Ann Surg 1944;120:284-310. 
Elkin DC: Wounds of the heart. Ann Surg 1944;120:817-821. 
Elkin DC: Arteriovenous aneurysm. Surg Gynecol Obstet 1945;80:217- 

224. 
Elkin DC: Traumatic aneurysm: The Matas Operation 57years after. Surg Gynecol 

Obstet 1946;82:1-12. 



RE.qxd 4/16/04 3:19 PM Page 567 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 567 

Elkin DC, Banner EA: Arteriovenous aneurysms following surgical operations. 
JAMA 1946;131:1117-1119. 

Elkin DC, Campbell RE: Cardiac tamponade: Treatment by aspiration. Ann 
Surg 1941;133:623-630. 

Elkin DC, DeBakey ME: Vascular surgery in World War II. Washington, DC, US 
Government Printing Office, 1955. 

Elkin DC, Harris MH: Arteriovenous aneurysm of the vertebral vessels. Ann 
Surg 1946;124:934-951. 

Elkin DC, Shumacker HB Jr: In Elkin DC, DeBakey ME (eds) : Vascular Surgery 
in World War II. Washington, DC, US Government Printing Office, 1944. 

Elkin DC, Woodhall B: Combined vascular and nerve injuries of warfare. Ann 
Surg 1944;119:411-431. 

Elkins RC, DeMeester TR, Brawl ey RK: Surgical exposure of the upper abdomi- 
nal aorta and its branches. Surgery 1971;70:622-627. 

Eller JL, Ziter FMH: Avulsion of the innominate artery from the aortic arch. 
Radiology 1970;94:75-78. 

Elliot JA: Acute arterial occlusion: An unusual cause. Surgery 1956;39:825-827. 

Ellis F: Surgical repair of a traumatic rupture of the thoracic aorta. Br J Surg 
1959;46:495-499. 

Ellis J: Case of gunshot wound, attended with secondary hemorrhage in which 
both carotid arteries were tied at an interval of four and a half days. N YJ 
Med 1845;5:187. 

Ellis J: A case of probable lead poisoning, resulting fatally from a bullet lodged 
in the knee joint twelve years previously. Boston Med Surg J 1874;91: 
472. 

Ellison RG, Meares CH: Traumatically injured aortic arch repaired under pro- 
found hypothermia and circulatory arrest. Am Surg 1977;43:298-302. 

Elmore JR, Gloviczki P, Harper CM Jr, et al: Spinal cord injury in experimen- 
tal thoracic aortic occlusion: Investigation of combined methods of pro- 
tection. J Vase Surg 1992;15:789-799. 

Enderson BL, Reath DB, Meadors J, Dallas W: The tertiary trauma survey: A 
prospective study of missed injury. J Trauma 1990;30:666. 

Enge I, Aakhus T, Evensen A: Angiography in vascular injuries of the extremi- 
ties. Acta Radiol Diag 1975;16:193-199. 

Engleman RM, Clements JM, Herrmann JB: Stab wounds and traumatic false 
aneurysms in the extremities. J Trauma 1969;9:77-87. 

Epstein BM, Bocchiola FC, AndrewsJC, Bester L: Traumatic arteriovenous fistula 
involving the portal venous system. Clin Radiol 1987;38:91-93. 

Ergin MA, O'Connor JV, Blanche C, Griepp RB: Use of stapling instruments 
in surgery for aneurysms of the aorta. Ann Thorac Surg 1983;36:161-166. 

Ernst CB, Daugherty ME, Bristow DL: Supraceliac pseudoaneurysm of abdomi- 
nal aorta. Arch Surg 1975;110:211-225. 

Ernst CB, Kaufer F: Fibulectomy-fasciotomy: An important adjunct in the man- 
agement of lower extremity arterial trauma. J Trauma 1971;11:365. 

Erskine JM: Case report: A true traumatic aneurysm of the radial artery at the 
wrist successfully treated by resection and arterial repair. J Trauma 
1964;4:530-534. 

Escobar GA, Escobar SC, Marquez L, et al: Vascular trauma: Late sequelae and 
treatment. J Cardiac Surg 1980;21:35-40. 

Eshaghy B, Loeb HS, Miller SE, et al: Mediastinal and retropharyngeal hem- 
orrhage. A complication of cardiac catheterization. JAMA 1973;226:427- 
431. 



RE.qxd 4/16/04 3:19 PM Page 568 



568 VI • THE RICH REFERENCE COLLECTION 



Esmarch F: The Surgeons Handbook of the Treatment of the Wounded in War. 
New York, LW Schmidt, 1878. 

Espada R, Wisennand HH, Mattox KL, Beall AC Jr: Surgical management of 
penetrating injuries to the coronary arteries. Surgery 1975;78:755-760. 

Espinoza R, Sonneborn R: Contusion of the abdominal aorta and massive 
embolization from the use of a seat belt. Rev Med Chil 1990;118:1376-1379. 

Esposito TJ, Jurkovich G, Rice CL: Reappraisal of emergency room thoraco- 
tomy in a changing environment. J Trauma 1991;31:881-887. 

Esterera AS, Pass LJ, Piatt MR: Systemic arterial air embolism in penetrating 
lung injury. Ann Thorac Surg 1990;2:257. 

EttienJT: Crutch-induced aneurysms of the axillary artery. Am Surg 1980;46:267- 
269. 

Evans J, Gray LA, Rayner A, Fulton RL: Principles for the management of pen- 
etrating cardiac wounds. Ann Surg 1979;189:777-784. 

Evans WE, Bernhard VM: Tibial artery bypass for ischemia resulting from frac- 
tures. J Trauma 1971;11:999-1007. 

Fabian TC, Turkleson ML, Connelly TL, Stone HH: Injury to the popliteal artery. 
Am J Surg 1982;143:225-228. 

Fabian TC, Mangiante EC, Patterson CR, et al: Myocardial contusion in blunt 
trauma: Clinical characteristics, means of diagnosis and implications for 
patient management. J Trauma 1988;28:50-57. 

Fabian TC, George SM Jr, Croce MA, et al: Carotid artery trauma: Management 
based on mechanism of injury. J Trauma 1990;30:953-963. 

Fabian TC, Richardson JD, Croce MA, et al: Prospective study of blunt aortic 
injury: Multicenter trial of the American Association for the Surgery of 
Trauma. J Trauma 1997;42:748-755. 

Fabian TC, Richardson JD, Croce MA, et al: Perspective study of blunt aortic 
injury: Multicenter trial of the American Association for Surgery of Trauma. 
J Trauma 1997;42:374-387. 

Fakhry BM, Jaques FF, Proctor HJ: Cervical vessel injury after blunt trauma. 
J Vase Surg 1988;8:501-508. 

Falkmer L, Eriksson A, Arnerlov C, Domellof L: Arterial bullet embolism with 
radiologic demonstration of vessel entrance site. World J Surg 1987;1 1:548- 
558. 

Fallahnejad M, Kutty ACK, Menut H: The importance of intra-aortic balloon 
pumping in the management of coronary artery laceration. J Cardiac Surg 
1983;23:426-428. 

Fallahnejad M, Kutty ACK, Wallace HW: Secondary lesions of penetrating cardiac 
injuries: A frequent complication. Ann Surg 1980;191:228-233. 

Fallahnejad M, Wallace HW, Su CC, et al: Unusual manifestations of penetrat- 
ing cardiac injuries. Arch Surg 1975;110:1357-1362. 

Fallon G, Thomford NR: False aneurysm of the superficial femoral artery asso- 
ciated with fracture of the femur. Angiology 1970;21:120-123. 

Falor WH, Hansel JR, Williams GB: Gangrene of the hand: A complication of 
radial artery cannulation. J Trauma 1976;16:713-716. 

Fargally M, ChristensonJT: Combined aortic and inferior vena cava injury during 
laminectomy. Vasa 1988;17:288-292. 

Farhood A, McGuire GM, Manning AM, et al: Intercellular adhesion molecule 
1 (ICAM-1) expression and its role in neutrophil induced ischemia- 
reperfusion injury in rat liver. J Leukoc Biol 1995;57:368-374. 

Farina C, Cavallaro A, Schultz RD, et al: Popliteal aneurysms. Surg Gynecol 
Obstet 1989;169:7-13. 



RE.qxd 4/16/04 3:19 PM Page 569 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 569 

Faro RS, Monson DO, Weinberg M, Javid H: Disruption of aortic arch branches 
due to nonpenetrating chest trauma. Arch Surg 1983;118:1333-1336. 

Farret A, Da Ros CT, Fischer CA, et al: Suprarenal aorta reconstruction using 
a saphenous spiral graft: A case relate? J Trauma 1994;37:114-118. 

Fasol R, Irvine S, Zilla P: Vascular injuries caused by anti-personnel mines. 
J Cardiac Surg 1989;30:467. 

Fay R, Brosman S, Lidstrom R: Renal artery thrombosis: A successful revascu- 
larization by autotransplantation. J Urol 1974;111:572. 

Feczko JD, Lynch L, PlessJE, et al: An autopsy case review of 142 nonpene- 
trating (blunt) injuries of the aorta. J Trauma 1992;33:846-849. 

FeeJF, CavoyJM, Dainko EA: Pseudoaneurysm of the axillary artery following 
a modified Bristow operation: A report of a case and review. Cardiovasc 
Surg 1978;19:65-68. 

Feigenbaum H, Waldhausen JA, Hyde LP: Ultrasonic diagnosis of pericardial 
effusion. JAMA 1965;191:107. 

Feigenbaum H, Zaky A, Waldhausen JA: Use of ultrasound in diagnosis of peri- 
cardial effusion. Intern Med 1966;65:443. 

Feliciano DV: Managing peripheral vascular trauma. Infect Surg 1986;5:659- 
669. 

Feliciano DV: Pitfalls in the management of peripheral vascular injuries. In 
Roses D (ed): Problems in General Surgery. Philadelphia, JB Lippincott 
Co, 1986, pp 101-113. 

Feliciano DV: Vascular injuries. Adv Trauma 1987;179:206. 

Feliciano DV: Abdominal gunshot wounds. Ann Surg 1988;208:362-370. 

Feliciano DV: Approach to major abdominal vascular injury. J Vase Surg 
1988;7:730-736. 

Feliciano DV: Counterpoint. J Trauma 1992;32:553-554. 

Feliciano DV: Abdominal vessels. In Ivatury R, Cayten C (eds): The Textbook 
of Penetrating Trauma. Baltimore, Williams & Wilkins, 1996, pp 702-716. 

Feliciano DV: Abdominal vascular injuries. Surg Clin North Am 2001;68:741. 

Feliciano DV, Accola KD, BurchJM, Spjut-Patrinely V: Extra-anatomic bypass 
for peripheral arterial injuries. Am J Surg 1989;158:505. 

Feliciano DV, Bitondo CG, Mattox KL, et al: Civilian trauma in 1980s: A one- 
year experience with 456 vascular and cardiac injuries. Ann Surg 
1984;199:717-724. 

Feliciano DV, Bitondo CG, Mattox KL, et al: Combined tracheoesophageal 
injuries. Am J Surg 1985;150:710-715. 

Feliciano DV, BurchJM, Graham JM: Abdominal vascular injury. In Mattox KL, 
Feliciano DV, Moore EE (eds): Trauma. New York, McGraw-Hill, 1999, pp 
783-805. 

Feliciano DV, BurchJM, Mattox KL, et al: Balloon catheter tamponade in car- 
diovascular wounds. Am J Surg 1990;160:583-587. 

Feliciano DV, Cosseli JS, Bitondo CG: Bilateral external iliac artery injury from 
a single missile. J Cardiac Surg 1986;27:46. 

Feliciano DV, Cruse PA, BurchJM, Bitondo CF: Delayed diagnosis of arterial 
injuries. Am J Surg 1987;154:579-584. 

Feliciano DV, Cruse PA, Spjvi-Patrinely V, et al: Fasciotomy after trauma to the 
extremities. Am J Surg 1988;156:533-536. 

Feliciano DV, Gentry LO, Bitondo CG, et al: Single agent cephalosporin pro- 
phylaxis for penetrating abdominal trauma. Am J Surg 1986;152:674-681. 

Feliciano DV, Herskowitz K, O'Gorman RB: Management of vascular injuries 
in lower extremities. J Trauma 1988;28:319-328. 



RE.qxd 4/16/04 3:19 PM Page 570 



570 VI • THE RICH REFERENCE COLLECTION 



Feliciano DV, Mattox KL: Major complications of percutaneous subclavian vein 
catheters. Am J Surg 1979;138:869-874. 

Feliciano DV, Mattox KL, Graham JM, Bitondo CG: Five-year experience with 
PTFE grafts in vascular wounds. J Trauma 1985;25:71-82. 

Feller I: Surgical anatomy of the abdominal aorta. Ann Surg 1961;154:239. 

Fellmeth BD, Roberts AC, Bookstein JJ: Postangiographic femoral artery injuries: 
Nonsurgical repair with US-guided compression. Radiology 1991;178:671- 
675. 

Fenner MN, Fisher KS, Sergei NL, et al: Evaluation of possible traumatic tho- 
racic aortic injury using aortography. Am Surg 1990;56:497-499. 

Ferguson IA, Byrd WM, McAfee DK: Experiences in the management of arte- 
rial injuries. Ann Surg 1961;153:980-986. 

Ferguson WM: Arteriovenous aneurysm following osteotomy for genu valgum. 
Lancet 1914;1:532. 

Fernandez LG, Lain KY, Messersmith RN, et al: Transesophageal echocardio- 
graphy for diagnosing aortic injury: A case report and summary of current 
imaging techniques. J Trauma 1994;36:877-880. 

Fernandez LG, Radhakrishnan J, Gordon RT, et al: Thoracic injuries in pedi- 
atric patients. J Trauma 1995;38:384-388. 

Ferriter P, Hirschy J, Kesseler H, Scott WN: Popliteal pseudoaneurysm: A case 
report. J Bone Joint Surg 1983;65-A:695-697. 

Finkelmeier BA, Mentzer RM Jr, Kaiseer DL, et al: Chronic traumatic thoracic 
aneurysm: Influence of operative treatment on natural history: An analy- 
sis of reported cases. J Thorac Cardiovasc Surg 1982;84:257-266. 

Fish GDJr, Hockhauser M: Laceration of the popliteal artery due to blunt trauma. 
AmJSurgl957;94:651. 

Fisher GW: Acute arterial injuries treated by the United States Army Medical 
Service in Vietnam, 1965-1966. J Trauma 1967;7:844-855. 

Fisher RE), Rienhoff WF III: Subclavian artery laceration resulting from frac- 
ture first rib. J Trauma 1966;6:579. 

Fisher RG, Ben-Manachem Y: Penetrating injuries of the thoracic aorta and 
brachiocephalic arteries: Angiographic finding in 18 cases. AJR Am J 
Roentgenol 1987;149:607-611. 

Fisher RG, Chasen MH, Lamki N: Diagnosis of injuries of the aorta and bra- 
chiocephalic arteries caused by blunt chest trauma: CT vs aortography. AJR 
Am J Roentgenol 1994;162:1047-1052. 

Fisher RG, Oria RA, Mattox KL, et al: Conservative management of aortic 
lacerations due to blunt trauma. J Trauma 1990;30:1562-1566. 

Fisher RG, Ward RE, Ben-Manachem Y, et al: Arteriography and the fractured 
first rib: Too much or too little? AJR Am J Roentgenol 1982;1 17:657- 
661. 

Fitchett VH, Pomerantz M, Butsch DW, et al: Penetrating wounds of the neck. 
Arch Surg 1969;99:307-314. 

Fitts CT, Barnett LT, Webb CM, et al: Perforating wounds of the heart caused 
by central venous catheters. J Trauma 1970;10:764-769. 

Fitzgerald EJ, Brumsler WG, Ruttley ST: False aneurysm of the femoral artery: 
Computed tomography and ultrasound appearance. Clin Radiat 
1986;37:585-588. 

Fitzgerald JB, Crawford ES, DeBakey ME: Surgical consideration of non pene- 
trating abdominal injuries. Am J Surg 1960;100:22-29. 

Fitzgerald JF, KeatesJ: False aneurysm as a late complication of anterior dislo- 
cation of the shoulder. Ann Surg 1975;181:785-786. 



RE.qxd 4/16/04 3:19 PM Page 571 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 571 

Fitzsimons LE, Garvey FK: Inferior vena caval injury: Case report. J Urol 

1959;83:258-259. 
Flanigan DP: Civilian Vascular Trauma. Philadelphia, Lea & Febiger, 1992. 
Flanigan DP, Keifer TJ, Schuler JJ, et al: Experience with iatrogenic pediatric 

vascular injuries: Incidence, etiology, management, and results. Ann Surg 

1983;198:430-442. 
Fleckenstein JL, Schultz SM, Miller RH: Serial aortography assesses stability of 

"atypical" aortic arch ruptures. Cardiovasc Intervent Radiol 1987;10:194- 

197. 
Fleming A, Bishop M, Shoemaker W, et al: Prospective trial of supranormal 

values as goals of resuscitation in severe trauma. Arch Surg 1992; 1 27: 11 75- 

1181. 
Fleming AW, Green DC: Traumatic aneurysms of the thoracic aorta: A report 

of 43 patients. Ann Thorac Surg 1974;18:91-101. 
Fleming JI: Case of rupture of the carotid artery and wound of several of its 

branches successfully treated by tying off the common trunk of the carotid 

itself. (Translated by Coley RW). Med ChirJ (Lond) 1817;3:2. 
Fleming JFR, Petrie D: Traumatic thrombosis of the internal carotid artery with 

delayed hemiplegia. CanJ Surg 1968;11:166-172. 
Fleming WB: A case of attempted repair of the superior mesenteric vessels. Aust 

N Z J Surg 1961;31:151-154. 
Fletcher JP, Little JM: Injuries of branches of the aortic arch. Aust N Z J Surg 

1988;58:217-219. 
Flint LM: Arterial injuries with lower extremity fracture. Surgery 1983;93: 

5-8. 
Flint LM, Synder WH, Perry MO, Shires GT: Management of major vascular 

injuries in the base of the neck: An 11-year experience with 146 cases. Arch 

Surg 1973;106:407-413. 
Fogarty TJ: Catheter technique for arterial embolectomy. Ann Surg 1965;161:325- 

330. 
Fogarty TJ, CranleyJJ, Strasser ES, Hafner CD: A method for extraction of arte- 
rial emboli and thrombi. Surg Gynecol Obstet 1963;116:241-244. 
Fogelman MJ, Stewart RD: Penetrating wounds of the neck. Am J Surg 

1965;91:581-596. 
FomonJJ, Warren WD: Complications of peripheral arterial injuries. Arch Surg 

1965;91:610-616. 
Foramitti KI: Wien Klin Wschr 1910;25:46-55. (Cited in Foramitti.H) 
Forbes AD, Ashbaugh DG: Mechanical circulatory support during repair of tho- 
racic aortic injuries improves morbidity and prevents spinal cord injury. 

Arch Surg 1994;129:497-498. 
Forbes HW, Thompson CQ, Smith JW: Mesenteric arteriovenous fistula after a 

gunshot wound. J Trauma 1969;9:806. 
Forrest CR, Pang CY, Zhong AG, Kreidstein ML: Efficacy of intravenous infu- 
sion of prostacyclin (PG12) or prostaglandin Ej (PGEj) in augmentation 

of skin flap blood flow and viability in the pig. Prostaglandins 1991;225:518- 

527. 
Forster DA: Letter to editor. J Trauma 1985;25:1115. 
Foster JH, Carter JW, Grahamm CPJr, Edwards WH: Arterial injury secondary 

to the use of the Fogarty catheter. Ann Surg 1970;171:971-978. 
Foster RD, AlbrightJA: Acute compartment syndrome of the thigh: A case report. 

J Trauma 1990;30:108-110. 
Fouin A: Stir la sutre des vaisseaux. Presse Med 1908;16:233-236. 



RE.qxd 4/16/04 3:19 PM Page 572 



572 VI • THE RICH REFERENCE COLLECTION 



Fox S, Pierce WS, Waldhausen JA: Acute hypertension: Its significance in trau- 
matic aortic rupture. J Thorac Cardiovasc Surg 1979;77:622-625. 

Fradet G, Nelems B, Meuller NL: Penetrating injury of the torso with impale- 
ment of the thoracic aorta: Preoperative value of the computed tomographic 
scan. Ann Thorac Surg 1988;45:680-681. 

Frame SB, Timberlake GA, Rush DS, et al: Penetrating injuries of the abdomi- 
nal aorta. Am Surg 1990;56:651-654. 

Francis H III, Thai ER, Weigelt JA, Redman HC: Vascular proximity: Is it a valid 
indication for arteriography in asymptomatic patients? J Trauma 1991; 
31:512-514. 

Frank MJ, Nadimi M, Lesniak LJ: Effects of cardiac tamponade on myocardial 
performance blood flow and metabolism. Am J Physiol 1971;220:179-185. 

Franz JL, Simpson CR, Penny RM, et al: Avulsion of the innominate artery after 
blunt chest trauma. J Thorac Cardiovasc Surg 1974;67:478-480. 

Fraser GA: Closed traumatic rupture of common femoral artery. Ann Surg 
1965;161:539-544. 

Freeark RJ: Role of angiography in the management of multiple injuries. Surg 
Gynecol Obstet 1969;128:761-771. 

Freeman NE: Secondary hemorrhage arising from gunshot wounds of the peri- 
pheral blood vessels. Ann Surg 1945;122:631-640. 

Freeman NE: Arterial repair in the treatment of aneurysms and arteriovenous 
fistulae: A report of 18 successful restorations. Ann Surg 1946;124:888-919. 

Freeman NE, Shumacker HB Jr: Vascular surgery. In Elkin DC, DeBakey ME 
(eds): Washington, DC, US Government Printing Office, 1955. 

Freidman AL, GertlerJP: Thoracoabdominal repair of post-traumatic aorta- 
renal vein fistula following a previous transabdominal approach: A case 
report. J Trauma 1991;31:287-289. 

French BG, Hughes CF: Post-traumatic chromic false aneurysm of the ascend- 
ing aorta with long-term survival. Aust N Z J Surg 1994;64:284-285. 

Freshman SP, Wisner DH, Weber CJ: 2D echocardiography: Eemergent use in 
the evaluation of penetrating precordial trauma. J Trauma 1991;31:902- 
906. 

Fried G, Salerno T, Burke D, et al: Management of the extremity with combined 
neurovascular and musculoskeletal trauma. J Trauma 1978;18:481-484. 

Friedman SG: A History of Vascular Surgery. Mt. Kisco, NY, Futura Publishing, 
1989. 

Fromm SH, Lucas CE: Obturator bypass for mycotic aneurysm in the drug addict. 
Arch Surg 1970;100:82-83. 

Fromm SH, Carrasquilla C, Lucas C: The management of gunshot wounds of 
the aorta: The use of Dacron grafts to replace the injured aorta. Arch Surg 
1970;101:388-390. 

Fry RE, Fry WJ: Extracranial carotid artery injuries. Surgery 1980;88:581-587. 

Frydenberg M, Royle JP, Hoare M: Blunt abdominal aortic trauma. Aust N Z J 
Surg 1990;60:347-350. 

Frykberg ER: Arteriography of the injured extremity: Are we in proximity to 
an answer. J Trauma 1992;32:551-552. 

Frykberg ER, Crump JM, Dennis JW, et al: Nonoperative observation of clini- 
cally occult arterial injuries: Aprospective evaluation. Surgery 1991 ;1 09:85- 
96. 

Frykberg ER, Crump JM, Vines FS, et al: A reassessment of the role of arteri- 
ography in assessing acute vascular injuries. J Trauma 1989;29:1041- 
1052. 



RE.qxd 4/16/04 3:19 PM Page 573 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 573 

Frykberg ER, Crump JM, Vines FS, McLellan GL: A reassessment of the role 
of arteriography in penetrating proximity extremity: A prospective study. 
J Trauma 1989;29:1041-1052. 

Frykberg ER, Dennis JW, Bishop K, et al: The reliability of physical examina- 
tion in the evaluation of penetrating extremity trauma for vascular injury: 
Results at one year. J Trauma 1991;31:502-511. 

Frykberg ER, Vines FS, Alexander RH: The natural history of clinically occult 
arterial injuries: A prospective evaluation. J Trauma 1989;29:577-583. 

Fu WR: Angiography of Trauma. Springfield, 111, Charles C Thomas, 1972. 

Fulda G, Rodriguez A, Turney SZ, Cowley RA: Blunt traumatic pericardial rupture: 
A ten-year experience 1979-89. J Cardiac Surg 1990;31:525-530. 

Fullen WD, Hunt J, Altemeier WA: The clinical spectrum of penetrating injury 
to the superior mesenteric arterial circulation. J Trauma 1972;12:656-664. 

Furman S, Vijaynagar R, McMullen M, Scher DJW: Lethal sequelae of intra- 
aortic balloon rupture surgery. Surgery 1971;69:121-129. 

Furnas DW, Saliblan AH, Achauer BM: Genesis of a replantation program. Am 
J Surg 1978;136:21-25. 

Gaddard P,Jones AG, WisheartJD: Self-inflicted stab wound causing aorto-right 
ventricular fistula. Am HeartJ 1981;46:101-103. 

Gage M: Traumatic injuries to the peripheral vessels in both civil and military 
practice. Surgery 1942;11:983-986. 

Gainor BJ, Metzler M: Humeral shaft fracture with brachial artery injury. Clin 
Orthop Relat Res 1986;204:154-161. 

Gale SS, Scissons RP, Jones L, Salles-Cunha SX: Femoral pseudoaneurysm throm- 
binjection. Am J Surg 2001;181:379-383. 

Gallen J, Wiss DA, Cantelmo N, Menzoian JO: Traumatic pseudoaneurysm of 
the axillary artery: Report of three cases and literature review. J Trauma 
1984;24:350-354. 

Gammie JS, Katz WE, Swanson ER, Peitsman AB: Acute aortic dissection after 
blunt chest trauma. J Trauma 1996;40:126-127. 

GaramellaJJ, Schmidt WR, Jensen NK, Lynch MF: Traumatic aneurysms of the 
thoracic aorta. Report of four cases, including one of spontaneous rupture 
into the esophagus. N EnglJ Med 1962;266:1341-1348. 

Garcia-Rinaldi R, Defore WW, Mattox KL, Beall ACJr: Unimpaired renal myocar- 
dial and neurologic function after cross clamping of the thoracic aorta. 
Surg Gynecol Obstet 1976;143:249-252. 

Gardner C: Traumatic vasospasm and its complications. Am J Surg 1952;83:468- 
470. 

Gardner WJ, StorerJ: The use of the G Suit in control of intra-abdominal bleed- 
ing. Surg Gynecol Obstet 1966;123:792. 

Garrison RN, Richardson JD, Fry DE: Diagnostic transdiaphragmatic pericar- 
diotomy in thoracoabdominal trauma. J Trauma 1982;22:147-149. 

Garzon A, Gleidman ML: Peripheral embolization of bullet following perfora- 
tion of the thoracic aorta. Am Surg 1964;160:901. 

Caspar MR: Vascular injury secondary to drug abuse. In Ernst CB, Stanley JC 
(eds) : Current Therapy in Vascular Surgery. Philadelphia, BC Decker, 1987. 

Caspar MR, Hare RR: Gangrene due to intraarterial injection of drugs by drug 
addicts. Surgery 1987;72:573-577. 

Caspar MR, Treiman RL: The management of injuries to major veins. Am J 
Surg 1960;100:171-175. 

Caspar MR, Treiman RL, Payne JH, et al: Principles of treatment and special 
problems in vascular trauma. Surg Clin North Am 1968;48:1355. 



RE.qxd 4/16/04 3:19 PM Page 574 



574 VI • THE RICH REFERENCE COLLECTION 



Gaspard DJ, Caspar MR: Arteriovenous fistula after Fogarty catheter thrombec- 
tomy. Arch Surg 1972;105:90-92. 

Gasparini D, Lovaria A, Saccheri S: Percutaneous treatment of iliac aneurysms 
and pseudoaneurysms with Cragg Endopro System 1 stent-grafts. Cardio- 
vasc Intervent Radiol 1997;20:348-352. 

Gates JD, Clair DG, Hechtman DH: Thoracic aortic dissection with renal artery 
involvement following blunt thoracic trauma: Case report. J Trauma 
1994;36:430-432. 

Gates JD, Knox JB: Axillary artery injuries secondary to anterior dislocation of 
the shoulder. J Trauma 1995;39:581-583. 

Gauderer MW, Wolkoff JS, Izant RJ: Traumatic aneurysm of the suprarenal 
abdominal aorta: Surgical reconstruction in a 7-year-old patient. J Pediatr 
Surg 1982;17:940-945. 

Gaudio KM, Taylor MR, Chaudry IH, et al: Accelerated recovery of single nephron 
function by the postischemic infusion of ATP-MgC12. Kidney Int 1982;22:13- 
20. 

Gavant ML, Gold RE, Fabian TC, Tonkin LI: Vascular trauma to the extremi- 
ties and lower neck: Initial assessment with intravenous digital subtraction 
angiography. Radiology 1986;158:755-760. 

Gay W: Blunt trauma to the heart and great vessels. Surgery 1982;91:507-509. 

Gazzanaga AB, IKhuri EI, Mir-Sepasi HM, Bartlett RH: Rupture of the thoracic 
aorta following blunt trauma. Arch Surg 1975;110:1119-1123. 

Geer TM, Rich NM: Cardiac trauma in Vietnam: Unpublished data in the Vietnam 
Vascular Registry. 1972. 

Geer TM: Personal communication, 1972. 

Geiran O, Solheim K: Cardiac and aortic injuries. Scand J Thorac Cardiovasc 
Surg 1974;8:27-33. 

GeisWP, Johnson CF, ZajtchukR, Kittle CF: Extrapericardial (mediastinal) cardiac 
tamponade. Arch Surg 1970;100:305-306. 

GelbermanRH, BlasingameJP, ArnostF, et al: Forearm arterial injuries. J Hand 
Surg 1979;4:401-408. 

Gelberman RH, Menon J, Fronek A: The peripheral pulse following arterial 
injury. J Trauma 1980;20:948-950. 

Gelberman RH, NunleyJA, Koman LA, et al: The results of radial and ulnar 
arterial repair in the forearm. J Bone Joint Surg 1982;64:383-387. 

Geller DA, Chia SH, Takahashi Y, et al: Protective role of the L-arginine-nitric 
oxide synthase pathway on preservation injury after rat-liver transplanta- 
tion. J Parenter Enter Nutr 2001;25:142-147. 

Gensoul: Note sur les Bless es recus a l'Hotel Dieu de Lyon, pendant les trou- 
bles de 1831. Gaz Med Paris 1883;297. 

Gensoul: 1883 Cited in Simeone, et al. On the question of ligation of the con- 
comitant vein when a major artery is interrupted. Surgery 1951;29:932-951. 

Gentry LO, Feliciano DV, Lea AS, et al: Perioperative antibiotic therapy for 
penetrating injuries of the abdomen. Ann Surg 1984;200:561-566. 

George SM Jr, Croce MA, Fabian TC: Cervicothoracic arterial injuries: Recom- 
mendations for diagnosis and management. WorldJ Surg 1991;15:134-140. 

Ger R: The coverage of vascular repairs by muscle transposition. J Trauma 
1976;16:974-978. 

Gerbode F, Holman E, Dickenson EH, Spencer FC: Arteriovenous fistulas and 
arterial aneurysms: The repair of major arteries injured in warfare, and 
the treatment of an arterial aneurysm with a vein graft inlay. Surgery 1952; 
32:259-274. 



RE.qxd 4/16/04 3:19 PM Page 575 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 575 

Gerbode F, Braimbridge M, Osborn JJ, et al: Traumatic thoracic aneurysms: 

Treatment by resection and grafting with the use of an extracorporeal bypass. 

Surgery 1957;42:975-985. 
Gerlock AJ Jr: The use of pedal venous pressure (PVP) as a guide in evaluating 

the patency of venous repairs. J Trauma 1977;17:108-110. 
Gerlock AJ Jr, Mathis J, Goncharenko V, Maravilla A: Angiography of intimal 

and intramural arterial injuries. Radiology 1978;129:357-361. 
Gerlock AJJr, Muhletaler CA: Venography of peripheral venous injuries. Radi- 
ology 1979;133:77-80. 
Gerlock AJJr, Thai ER, Snyder WH III: Venography in penetrating injuries of 

the extremities. AJR Am J Roentgenol 1976;126:1023-1027. 
German DS, Shapiro MJ, Willman VL: Acute aortic valvular incompetence 

following blunt thoracic deceleration injury: Case report. J Trauma 

1990;30:1411-1412. 
Gertsch P: Lesions vasculaires des blesses de guerre Afghans Traitees a 

l'Hospital du CICR a Peshawar. Med Milit 1986;2:46. 
Ger vin AS, Fischer CA: The importance of prompt transport in salvage of patients 

with penetrating heart wounds. J Trauma 1982;22:443-448. 
Getzen LC, Bellinger SB, Kendall LW: Should all neck, axillary, groin or popliteal 

wounds be explored for possible vascular or visceral injuries. J Trauma 

1972;12:906. 
Geuder JW, Hobson RW II, Padberg FT Jr, et al: The role of contrast arterio- 
graphy in suspected arterial injuries of the extremities. Am Surg 1985;51:89- 

93. 
Gewertz B, O'Brien C, Kirsh MM: Use of the intra-aortic balloon support 

of refractory low cardiac output in myocardial contusion. J Trauma 

1977;17:325-327. 
Gewertz BL, Samson DS, Ditmore QM, Bone GE: Management of penetrating 

injuries of the internal carotid artery at the base of the skull utilizing 

extracranial-intracranial bypass. J Trauma 1980;20:365-369. 
GibbonsJR: Treatment of missile injuries of the chest: Belfast experience. J Car- 

diothorac Surg 1989;4:297. 
Gibson JMC: Rupture of axillary artery. J Bone Joint Surg 1962;44B:114- 

115. 
Gielchinsky I, McNamara JJ: Cardiac wounds at a military evacuation hospital 

in Vietnam: A review of one-year's experience. J Thorac Cardiovasc Surg 

1970;60:603-606. 
Gielchinsky I, McNamaraJJ: Flechette wounds of the heart. Surgery 1971;69:229- 

231. 
GigliaJS, Zelenock GB, D'Alecy L: Prevention of paraplegia during thoracic 

aortic cross-clamping: Importance of patent internal mammary arteries. 

J Vase Surg 1994;19:1044-1051. 
Gill SS, Eggleston FC, Singh CM: Arterial injuries of the extremities. J Trauma 

1976;16:766-772. 
Gingrass RP, Cunningham DS, Paletta FX: Skin grafting of exposed arterial 

grafts: A clinical and experimental study. J Trauma 1973;13:951-957. 
Giraud RM: Arteriovenous fistula of the aortic arch complicating stab wound 

of the neck. S Afr Med J 1965;12:474-476. 
Glaser RL, McKellar D, Scher KS: Arteriovenous fistulas after cardiac catheter- 
ization. Arch Surg 1989;124:1313-1315. 
Gltick T: Uber zwei falle von aortenaneurysmen nebst bemerkungen uber die 

naht der blutgefasse. Arch Klin Chir 1883;28:548. 



RE.qxd 4/16/04 3:19 PM Page 576 



576 VI • THE RICH REFERENCE COLLECTION 



Gnanadev DA, Fandrich BL: Popliteal artery trauma: Update and recent advances 
in management. Ann Vase Surg 1988;2:332-335. 

Goff JB, Gillespie DL, Rich NM: Long-term follow-up of a superficial femoral 
vein injury: A case report from the Vietnam Vascular Registry. J Trauma 
1998;44:209-211. 

Goff J, Gillespie D, Rich NM: Long-Term Follow-up of Military Superficial Femoral 
Vein Injury: A Case Report from Vietnam Vascular Registry. 1996 Febru- 
ary 23-25, San Diego. San Diego, American Venous Forum, 1996. 

Gold AH, Lee GW: Upper extremity replantation: Current concepts and patient. 
J Trauma 1981;21:551-557. 

Goldberg BB, Ostrium BJ, Isard JJ: Ultrasonic determination of pericardial effu- 
sion. JAMA 1967;202:103. 

Goldberger M: Clinical experience with intra-aortic balloon counter pulsation 
in 112 consecutive patients. Am Heart 1986;111:497-502. 

Goldman BS, Firor WB, Key JA: The recognition and management of peri- 
pheral arterial injuries. Can Med Assoc J 1965;92:1154-1160. 

Goldman LI, Maier WP, Drezner AD: Another complication of subclavian punc- 
ture: Arterial laceration. JAMA 1971;217:78. 

Goldman MH, Kent S, Schaumburg E: Brachial artery injuries associated with 
posterior elbow dislocation. Surg Gynecol Obstet 1987;164:95-97. 

Goletti O, Ghiselli G, Lippolis PV, et al: The role of ultrasonography in blunt 
abdominal trauma: Results in 250 consecutive cases. J Trauma 1994;36:1 78- 
181. 

Golman BS, Firor WB, KeyJA: The recognition and management of peripheral 
arterial injuries. Can Med Assoc J 1965;92:1154-1160. 

Golueke PJ, Goldstein AS, Sclanfani SJA, et al: Routine versus selective explo- 
ration of penetrating neck injuries: A randomized, prospective study. 
J Trauma 1984;24:1010-1014. 

Golueke PJ, Sclanfani S, Phillips T, et al: Vertebral artery injury: Diagnosis and 
management. J Trauma 1987;27:856-865. 

Gomes MMR, Gernatz PE: Arteriovenous fistulas: A review and 10-year experi- 
ence at the Mayo Clinc. Mayo Clin Proc 1970;45:81-102. 

Gomez GA, RreisDJJr, RatnerL, etal: Suspected vascular trauma of the extrem- 
ities: The role of arteriography in proximity injuries.JTraumal986;26:1005- 
1008. 

Gomez RL, Bole PV, Lande A, et al: Successful repair of two false aneurysms of 
the abdominal aorta. Arch Surg 1973;107:91-94. 

Goodman C: Suture of blood vessel from projectiles of war. Surg Gynecol Obstet 
1918;27:528. 

Goodman P, Jeffrey R, Brant-Zawadki M: Digital subtraction angiography in 
extremity trauma. Radiology 1984;153:61-64. 

Gordon RL, Landau RH, Shifrin E, Romanoff H: The use of a balloon catheter 
in the treatment of an iatrogenic pseudoaneurysm of the subclavian artery. 
J Cardiovasc Surg 1983;24:178-180. 

GormanJF: Combat wounds of the popliteal artery. Ann Surg 1968;24:178-180. 

Gorman JF: Combat arterial trauma: Analysis of 106 limb-threatening injuries. 
Arch Surg 1969;98:160-164. 

Gosselin RA, Yukka Siegberg CJ, Coupland R, Agerskov K: Outcome of arter- 
ial repairs in 23 consecutive patients at the ICRC-Peshawar hospital for war 
wounded. J Trauma 1993;34:373-376. 

Gott VL: Heparinized shunts for thoracic vascular operations. Ann Thorac Surg 
1972;14:219-220. 



RE.qxd 4/16/04 3:19 PM Page 577 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 577 

Goyanes J: Neuvos trabajos de chirurgial vascular: Substitution plastica de las 
the arterias por las venas, o arterio-plastia venosa, aplicado, como neuvo 
metodo, al traitamiento de los aneurismas. El Siglo Med 1906;53:546- 
561. 

Grablowsky OM, Weichert RF III, Goff JB, Schlegel JU: Renal artery 
thrombosis following blunt trauma: Report of four cases. Surgery 
1970;67:895. 

Graeber GM, Clagett GR Wolf RE, et al: Alteration in serum creatine kinase 
and lactate dehydrogenase: Association with abdominal aortic surgery, 
myocardial infarction and bowel necrosis. Chest 1990;97:521-527. 

Graham JM, Feliciano DV, Mattox KL: Combined brachial, axillary and sub- 
clavian artery injuries of the same extremity: A case report. J Trauma 
1980;20:899-901. 

Graham JM, Feliciano DV, Mattox KL, Beall AC Jr: Innominate vascular injury. 
J Trauma 1982;22:647-655. 

Graham JM, Feliciano DV, Mattox KL, et al: Management of subclavian vascu- 
lar injuries. J Trauma 1980;20:537-544. 

Graham JM, Mattox KL: Right ventricular bullet embolectomy without car- 
diopulmonary bypass. J Thorac Cardiovasc Surg 1981;82:310-313. 

Graham JM, Mattox KL, Beall AC Jr: Portal venous system injuries. J Trauma 
1978;18:419-422. 

Graham JM, Mattox KL, Beall AC Jr, DeBakey ME: Traumatic injuries of the 
inferior vena cava. Arch Surg 1978;113:413-418. 

Graham JM, Mattox KL, Beall AC Jr, DeBakey ME: Injuries to the visceral arter- 
ies. Surgery 1978;84:835-839. 

Graham JM, Mattox KL, Feliciano DV, DeBakey ME: Vascular injuries of the 
axilla. Ann Surg 1982;195:232-238. 

Gray WA: Prehospital resuscitation: The good, the bad, and futile. JAMA 
1993;22:1471-1472. 

Green NE, Allen BL: Vascular injuries associated with dislocation of the knee. 
J Bone Joint Surg 1977;59-A:236-239. 

Greenfield LJ, Ebert PA: Technical considerations in the management of axil- 
lobrachial arterial injuries. J Trauma 1967;7:606-612. 

Greenfield LJ: Perforation of the inferior vena cava with aortic and vertebral 
penetration by a suprarenal Greenfield filter. Radiology 1990;175:287. 

Greenholz SK, Moore EE, Peterson NE, Moore GE: Traumatic bilateral renal 
artery occlusion: Successful outcome with out surgical intervention. J Trauma 
1986;26:941-944. 

GreenoughJ: Operations on innominate artery: Report of a successful ligation. 
Arch Surg 1929;19:1484-1544. 

Griffen WO, BelinRP, Walder AL: Traumatic aneurysm of the abdominal aorta. 
Surgery 1966;60:813-816. 

Griffiths SJH: Traumatic rupture of the aorta. Br J Surg 1931;18:664-665. 

Grimley RP, Ashton F, Slaney G, Batten RL: Popliteal artery injuries associated 
with civilian knee trauma. Injury 1981;13:1-6. 

Griswold RA, Maguire CH: Penetrating wounds of the heart in pericardium. 
Surg Gynecol Obstet 1942;74:406-418. 

Griswold RA, Drye JC: Cardiac wounds. Ann Surg 1954;139:783-785. 

Groms GA, FlintLT: Repair of two traumatic aneurysms of the supraceliac abdomi- 
nal aneurysm in a single patient. Surgery 1972;72:371-377. 

Gross WS, Flanigan P, Kraft RO, Stanley JC: Chronic upper extremity arterial 
insufficiency. Arch Surg 1978;113:419-423. 



RE.qxd 4/16/04 3:19 PM Page 578 



578 VI • THE RICH REFERENCE COLLECTION 



Grossi EA, Krieger KH, Cunningham JN Jr, et al: Venoarterial bypass: A tech- 
nique for spinal cord protection. J Thorac Cardiovasc Surg 1985;89:228- 
234. 

Grosso MA, Simson MA, Kobayaski K: Myocardial ischemia pattern determines 
predisposition to ventricular arrhythmias. Surg Forum 1984;34:239-240. 

Groves LK: Traumatic aneurysm of the thoracic aorta. N Engl J Med 1964; 
270:220-224. 

Gruber HE, Weisman WH: Aortic thrombosis during sigmoidoscopy in Behcet's 
syndrome. Arch Intern Med 1983;143:343-345. 

Gryska PF: Major vascular injuries: Principles of management in selected cases 
of arterial and venous injuries. N Engl J Med 1962;266:381-385. 

Gschaedler R, Dollfus P, LoebJP, et al: Traumatic rupture of the aorta and para- 
plegia. Paraplegia 1978;16:123-127. 

Gubler KD, Wisner DH, Blaisdell FW: Multiple vessel injury to branches of the 
aortic arch: Case report. J Trauma 1991;31:1566-1568. 

Guerriero WG, Carlton CE Jr, Scott R, Beall AC Jr: Renal pedical injuries. 
J Trauma 1971;11:53-62. 

Gugenheim S, Sanders RJ: Axillary artery rupture caused by shoulder disloca- 
tion. Surgery 1984;95:55. 

Guilfoil PH, Christiansen T: An unusual vascular complication of fractured clav- 
icle. JAMA 1967;200:72-73. 

Gundry SR, Willimams S, Burney RE, et al: Indications for aortography in blunt 
thoracic trauma: A reassessment. J Trauma 1982;22:664-671. 

Gundry SR, Burney RE, MacKenzieJR, et al: Clinical contact with the trauma 
patient enhances accuracy of chest roentgenogram interpretation in pre- 
dicting traumatic rupture of the aorta. J Cardiovasc Surg 1985;26:332-336. 

Gunnlaugnson GH, Hallgrimsson JG, Sigurdsson JL, et al: Complete traumatic 
avulsion of the innominate artery from the aortic arch with a unique mech- 
anism of injury. J Thorac Cardiovasc Surg 1973;66:235-240. 

Gupta BK, Khaneja SC, Flores L, et al: The role of intra-aortic balloon occlu- 
sion in penetrating abdominal trauma. J Trauma 1989;29:861-865. 

Gupta SK, Veith FJ: Management of juxtarenal aortic occlusion: Technique for 
suprarenal clamp placement. Ann Vase Surg 1992;6:306-310. 

Gurdjian ES, Hardy WG, Lindner DW, Thomas LM: Closed cervical cranial 
trauma, associated with involvement of carotid and vertebral arteries. Neu- 
rosurgery 1963;20:418. 

GurriJA, Johnson GJr: Management of brachial artery occlusion after cardiac 
catheterization. Am Surg 1980;46:233-235. 

Guthrie CC: Blood Vessel Surgery. London, Edward Arnold & Company, 
1912. 

Guthrie CC: Blood Vessel Surgery and Its Application. New York, Longmans, 
Green and Co, 1912. 

Guthrie GJ: Disease and injuries of arteries. 1830. 

Guvendik L, Davis NR, Starr A: Repair of traumatic aortic transection: A man- 
agement protocol and review of 21 patients. J Thorac Cardiovasc Surg 
1988;36:198-201. 

Gwathmey O, Byrd CW: Clinical experience with acute traumatic rupture of 
the thoracic aorta in a general hospital. Ann Surg 1964;159:846-857. 

Hafez HM, Woolgar J, Robbs JV: Lower extremity artery injury: Results of 550 
cases and review of risk factors associated with limb loss. J Vase Surg 
2001;33:1212-1219. 

Haimovici H: History of arterial grafting. J Cardiac Surg 1963;4:152-174. 



RE.qxd 4/16/04 3:19 PM Page 579 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 579 

Haimovici H: Iatrogenic acute arterial thrombosis: In Vascular Emergencies. 
New York, Appleton-Century-Crofts, 1982, pp 225-232. 

Haimovici H, Hoffert PW, Zinicola N, Steinman C: An experimental and clin- 
ical evaluation of grafts in the venous system. Surg Gynecol Obstet 
1970;131:1173-1186. 

Hall RL, Anderson CA, Bickerstaff LK: Isolated vertebral artery injuries. Contemp 
Surg 1986;29:57-62. 

Haller JA: Bullet transection of both carotid arteries: Immediate repair with 
recovery. Am J Surg 1962;103:532-535. 

Hallowell. Extract of a letter from Mr. Lambert, surgeon at Newcastle upon 
Tyne, to Dr. Hunter, giving an account of new method of treating an 
aneurysm. Med Obser Inq 1762;30:360. 

Halpern AA, Nagel DA: Anterior compartment pressures in patients with tibial 
fractures. J Trauma 1980;20:786-790. 

Halpern NB, Aldrete JS: Factors influencing mortality and morbidity from injuries 
to the abdominal aorta and inferior vena cava. Am J Surg 1979;137:384- 
388. 

Halsted W: Ligation of the first portion of the left subclavian artery and exci- 
sion of subclavico-axillary aneurysm. Bull Johns Hopkins Hosp 1892;3:93. 

Halsted W: Discussion in Bernheim, BM. Bull Johns Hopkins Hosp 1916;27:93. 

Halsted WS: The effect of ligation of the common iliac artery on the circula- 
tion and function of lower extremity. Report of a cure of iliofemoral 
aneurysm by their application of an aluminum band to the vessel. Bull 
Johns Hopkins Hosp 1912;23:191-220. 

Halsted WS: Ligation of the left subclavian artery in its first portion. Bull Johns 
Hopkins Hosp 1924;21:1. 

Hamerlijnck RP, Rutsaert RR, De Geest R, et al: Surgical correction of descend- 
ing thoracic aortic aneurysms under simple aortic cross-clamping. J Vase 
Surg 1989;9:568-573. 

Hammond SL, Gomez ER, Coffey JA, et al: Involvement of the lymphatic system 
in chronic venous insufficiency. In Bergan JJ, Yao JST (eds): Venous Dis- 
orders. Philadelphia, WB Saunders, 1991. 

Hamza N, Marath A, Al-Fakhry MR: The management of aneurysms and arte- 
riovenous fistulae of the popliteal artery arising from war trauma: Empha- 
sis on sigmoid operative approach. J Cardiovasc Surg 1990;31:457-461. 

Hanobrugh JF, Narrod JA: Arteriovenous fistulas following central venous 
catheterization. Care Med 1983;9:287-289. 

Hansaker G: An ordeal in a snowdrift. The Evening Star 14 June 1971. 

Harbison SP: Major vascular complications of intervertebral disc surgery. Ann 
Surg 1954;140:342-348. 

Hardin WD Jr, Adinolfi MF, O'Connell RC, Kerstein MD: Management of trau- 
matic peripheral vein injuries: Primary repair or vein ligation. Am J Surg 
1982;144:235-238. 

Hardin CA: Bypass saphenous grafts for the relief of venous obstruction of the 
extremity. Surg Gynecol Obstet 1962;115:709-712. 

Hardin WD, O'Connell RC, Adinolfi MF, Kerstein MD: Traumatic arterial injuries 
of the upper extremity: Determinants of disability. Am J Surg 1985; 150:266- 
270. 

Hardy JD, Raju S, Neely WA, Berry DW: Aortic and other arterial injuries. Ann 
Surg 1975;181:640-653. 

Hardy JD, Timmis HH: Repair of intracardiac gunshot injuries: Report of three 
cases. Ann Surg 1969;169:906-913. 



RE.qxd 4/16/04 3:19 PM Page 580 



580 VI • THE RICH REFERENCE COLLECTION 



Hardy JD, Williams RD: Penetrating heart wounds: Analysis of 12 consecutive 
cases without morality. Ann Surg 1967;166:228-231. 

Hare RR, Caspar MR: The intimal flap. Arch Surg 1971;102:552-555. 

Harken DE: Foreign bodies in, and in relation to, the thoracic blood vessels 
and heart: Techniques for approaching and removing foreign bodies from 
the chambers of the heart. Surg Gynecol Obstet 1946;83:117-125. 

Harken DE, Williams AC: Foreign bodies in and in relation to the thoracic 
blood vessels and heart: Migratory foreign bodies within the blood vascu- 
lar system. Am J Surg 1946;72:80-90. 

Harken DE, Zoll PM: Foreign bodies in and in relation to the thoracic blood 
vessels and heart: Indications for the removal of intracardiac foreign bodies 
and the behavior of the heart during manipulation. Am HeartJ 1946;32:1- 
19. 

Harley DP, Mena I: Cardiac and vascular sequelae of sternal fractures. J Trauma 
1986;26:553-555. 

Harrington DP, Barth KH, Whit Rljr, Brawley RK: Traumatic pseudoaneurysm 
of the thoracic aorta in close proximity ot the anterior spinal artery: A ther- 
apeutic dilemma. Surgery 1980;87:153-156. 

Harrington EB, Schwartz M, Jacobson JH II, et al: Aortocaval fistula: A clinical 
spectrum. J Cardiovasc Surg 1989;30:579-583. 

Harris JD: A case of arteriovenous fistula following closed fracture of tibia and 
fibula. Br J Surg 1963;50:774-776. 

Hart RJJr, Gregoratos G: Ventricular septal defects caused by stab wounds: Report 
of two cases. Milit Med 1974;139:289-292. 

Hartford JM, Fayer RL, Shaver TE, et al: Transection of the thoracic aorta: 
Assessment of a trauma system. Am J Surg 1986;151:224-229. 

Harding RP, McGahan JP, Lindfors KK: Letter. Radiology 1987;165:291-292. 

Harding RP, McGahan JP, Blaisdell FW, Lindfors KK: Stab wounds to the extrem- 
ities: Indications for arteriography. Radiology 1987;162:465-467. 

Hartsuck JM, Moreland HJ, Williams GR: Surgical management of vascular 
trauma distal to the popliteal artery. Arch Surg 1972;105:937-940. 

Harvey EN, Butler EG, McMillenJH, Puckett WO: Mechanism of wounding. 
War Med 1945;8:91-104. 

Harvey EN, Korr IM, Oster G, McMillenJH: Secondary damage in wounding 
due to pressure changes accompanying the passage of high velocity mis- 
siles. Surgery 1947;21:218-239. 

Harvey EN, McMillenJH: An experimental study of shock waves resulting from 
the impact of high velocity missiles on animal tissues. J Exp Med 1947; 
85:321-328. 

Hashizume M, Yang Y, Gait S: Intimal response of saphenous vein to intralu- 
minal trauma simulated angioscopic insertion. J Vase Surg 1987;5:862-868. 

Hashmonai M, Schramek KA, Abrahamson J, et al: Acute vascular injuries of 
the upper extremity. Isr J Med Sci 1974;10:599. 

Haskell RJ, French WJ, Harley DP: Traumatic aorto-right ventricular fistula pre- 
senting with a diastolic murmur. Am HeartJ 1985;109:1110-1112. 

Hass GM: Types of internal injuries of personnel involved in aircraft accidents. 
JAviatMedl944;15:77. 

Hass L, Staple T: Arterial injures associated with fractures of the proximal tibia 
following blunt trauma. South Med J 1969;62:1439. 

Hassantash SA, Mock C, Maier RV: Traumatic visceral artery aneurysm: Pre- 
sentation as massive hemorrhage from perforation into an adjacent hollow 
viscus. J Trauma 1995;38:357-360. 



RE.qxd 4/16/04 3:19 PM Page 581 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 581 

Hasselgren PO, Eriksson B, Lukes P, Seeman T: False popliteal aneurysm caused 
by exostosis of the femur. J Cardiovasc Surg 1983;24:540-542. 

Hassett A, Moran J, Sabiston DC, Kisslo J: Utility of echocardiography in the 
management of patients with penetrating missile wounds of the heart. Am 
J Cardiol 1987;7:1151-1156. 

Hawkes SZ: Traumatic rupture of the heart and intrapericardial structures. Am 
J Surg 1935;27:503-507. 

Hawkins ML, Carraway RP, Ross SE: Pulmonary artery disruption from blunt 
thoracic trauma. Am Surg 1988;54:148-152. 

Hayes JM, Vanwickle GN: Axillary artery injury with minimally displaced frac- 
ture of the neck of the humerus. J Trauma 1983;23:431-433. 

Heaton LD, Hughes CW, Rosegay H, et al: Military surgical practices of the 
United States Army in Vietnam. In Current Problems in Surgery. Chicago, 
Year Book Medical Publishers, 1966. 

Heberer G: Ruptures and aneurysms of the thoracic aorta after blunt chest 
trauma.J Cardiac Surg 1971;12:115-120. 

Heberer G, Becker HM, Stelter WJ: Vascular injuries in polytrauma. World J 
Surg 1983;7:68-79. 

Hebra A, Roison JG, Elliott BM: Traumatic aneurysm associated with fibroin- 
timal proliferation of the common carotid artery following blunt trauma: 
Case report. J Trauma 1993;34:297-299. 

Hegarty MM, Angorn IB, Gollogy J, Baker LW: Traumatic arteriovenous fistu- 
lae. Injury 1975;7:20-28. 

Heggtveit HA, Campbell JS, Hooper GD: Innominate arterial aneurysms occur- 
ring after blunt trauma. Am J Clin Pathol 1974;42:69-74. 

Heiberg E, Wolverson MK, Sundaram M, Shields JB: CT in aortic trauma. AJR 
Am J Roentgenol 1983;140:1119-1124. 

Heidenhain L: Uber naht von arterienwunden. Centralbl Chir 1895;22:1113- 
1115. 

Helfet DL, Howey T, Sanders R, Johansen K: Limb salvage versus amputation. 
Preliminary results of the Mangled Extremity Severity Score. Clin Orthop 
1990;256:80. 

Helvie MA, Rubin JM, Silver TM, Kresowik TF: The distinction between femoral 
artery pseudoaneurysms and other causes of groin masses: Value of duplex 
doppler sonography. AJR 1988;150:1177-1180. 

Henderson V, Nambisan R, Smith ME, et al: Angiographic yield in penetrating 
extremity trauma. WesJ Med 1991;155:253. 

Hennessy OF, Gibson RN, Allison DJ: Use of giant steel coils in the therapeu- 
tic embolization of a superior mesenteric artery-portal vein fistula. Car- 
diovasc Intervent Radiol 1986;9:42. 

Henroteaux D, Thoorens P, Trotteur G: Rupture of the external iliac artery 
after total hip replacement. J Beige Radiol 1989;72:518-519. 

Heppenstall RB, Sapega AA, Izant T, et al: Compartment syndrome: A quanti- 
tative study of high-energy phosphorus compounds using 31P-magnetic 
resonance spectroscopy. J Trauma 1989;29:1113-1119. 

HergetCH: Wound ballistics. In Bowers WB (ed): Surgery of Trauma. Philadel- 
phia, JB Lippincott Co, 1956. 

Herlyn KE: Erfahrungen auf dem gebiete von aneurysmen und arteriovenosen 
fisteln. Atti della prima Riunione Internacionale de Angio-cardio- 
Chirurgia. Milano: Casa Edetrice "Mellon," 1951. 

Hermreck AS, Sifers TM, Reckling FW, et al: Traumatic vascular injuries: Methods 
and results of repair. AmJ Surg 1974;128:813-817. 



RE.qxd 4/16/04 3:19 PM Page 582 



582 VI • THE RICH REFERENCE COLLECTION 



Hershey FB. Secondary repair of arterial injuries. Am Surg 1961;27:33-41. 

Hershey FB, Spencer AD: Surgical repair of civilian arterial injuries. Arch Surg 
1960;80:953-962. 

Hershey FB, Spencer AD: Autogenous vein grafts for repairs of arterial injuries. 
Arch Surg 1963;86:836-845. 

Herskowitz K, Sclafani S: Acute traumatic dissection of the common iliac arter- 
ies with spontaneous healing: Case report. Cardiovasc Intervent Radiol 
1990; 13:364. 

Hertzer NR: Peripheral atheromatous embolization following blunt abdomi- 
nal trauma. Surgery 1977;82:244-247. 

Hess PJ, Howe HR, Robicsek F, et al: Traumatic tears of the thoracic aorta: 
Improved results using the Bio-Medicus pump. Ann Thorac Surg 1989;48:6- 
9. 

Hessel SJ, Adams DF: Complication of angiography. Radiology 1981;138:273- 
281. 

Hewes RC, Smith DC, Lavine MH: Iatrogenic hydromediastinum simulating 
aortic laceration. AJR Am J Roentgenol 1979;133:817-820. 

Hewitt RL: Technical considerations in acute military vascular injuries of the 
extremities. Milit Med 1969;134:617-621. 

Hewitt RL, Collins DJ: Acute arteriovenous fistulas in war injuries. Ann Surg 
1969;169:447-449. 

Hewitt RL, Collins DJ,HamitHF: Arterial injuries at a surgical hospital in Vietnam. 
Arch Surg 1969;98:313-316. 

Hewitt RL, Grablowsky OM: Acute traumatic dissecting aneurysm of the abdom- 
inal aorta. Ann Surg 1970;171:160-164. 

Hewitt RL, Smith AD, Becker ML, et al: Penetrating injuries of the thoracic 
outlet. Surgery 1974;76:715-722. 

Hewitt RL, Smith AD, Drapanas T: Acute traumatic arteriovenous fistulas. 
J Trauma 1973;13:901-906. 

Hewitt RL, Smith AD Jr, Weichert RF, Drapanas T: Penetrating cardiac injuries: 
Current trends in management. Arch Surg 1970;101:683-688. 

Heyes FLP, Aukland A: Occlusion of the common femoral artery complicating 
total hip arthroplasty. J Bone Joint Surg 1985;67:533-535. 

HiattJR, Busuttil RW, Wilson SE: Impact of routine arteriography on manage- 
ment of penetrating neck injuries. J Vase Surg 1984;1:860-866. 

HiattJR, Yeatman LA, Child JB: The value of echocardiography in blunt chest 
trauma. J Trauma 1988;28:914-922. 

HiattJR, Martin NA, Machleder HI: The natural history of a traumatic verte- 
bral artery aneurysm: Case report. J Trauma 1989;29:1592-1603. 

Hiebert CA, Gregory FJ: Bullet embolism from the head to the heart. JAMA 
1974;229:442-443. 

Hierholzer C, Harbercht BG, Billiar TR, Tweardy DJ: Hypoxia-inducible factor- 
1 activation and cyclo-oxygenase-2 induction are early reperfusion inde- 
pendent inflammatory events in hemorrhagic shock. Arch Orthop Trauma 
Surg 2001;121:219-222. 

Hiertonn T, Lindberg K, Rob CG: Cystic degeneration of the popliteal artery. 
Br J Surg 1957;44:348. 

Hiertonn T, RybeckB: Traumatic Arterial Lesions. Stockholm, ForsvaretsForskn- 
ingsanstalt, 1968. 

Higgins RSD, SanchezJA, DeGuidis L: Mechanical circulatory support decreases 
neurologic complications in the treatment of traumatic injuries of the tho- 
racic aorta. Arch Surg 1992;127:516-519. 



RE.qxd 4/16/04 3:19 PM Page 583 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 583 

Highbloom RY, Koolpe H, Morris MC: Traumatic occlusion of two radiocephalic 
fistulas: Case report and their management. J Trauma 1990;30:364-365. 

Hilgenberg AD, Logan DL, Adkins CW, et al: Blunt injuries of the thoracic 
aorta. Ann Thorac Surg 1992;53:233-239. 

Hill LL: A report of a case of successful suturing of the heart, and a table of 37 
other cases of suturing by different operators with various terminations 
and conclusions drawn. Med Rec N Y2001;62:846. 

Hillborn M, Downey D: Deep venous thrombosis complicating sonographically 
guided compression repair of pseudoaneurysm of the common femoral 
artery. AJR Am J Roentgenol 1993;161:1334-1335. 

Hills MW, Delprado AM, Deane SA: Sternal fractures: Associated injuries and 
management. J Trauma 1993;35:55-60. 

Hills MW, Thomas SG, McDougall PA, et al: Traumatic thoracic aortic rupture: 
Investigation determines outcome. Aust N Z J Surg 1994;64:312-318. 

Hiratzka LF, Wright CB: Experimental and clinical results of grafts in the venous 
system: A current review. J Surg Res 1978;25:542-561. 

Hirsch B: Blood Vessel Surgery. London, Edward Arnold & Company, 1912. 

Hirsch EF: Internal versus external fixation of fractures with concomitant vas- 
cular injuries in Vietnam. J Trauma 1971;11:463-473. 

Hirshberg A, Wall MJ, Allen MR: Double jeopardy: Thoracoabdominal injuries 
requiring surgical intervention in both chest and abdomen. J Trauma 
1995;39:225-231. 

HixWR, Mills M: Management of esophageal wounds. Ann Surg 1970;1 72:1002- 
1006. 

Ho RM, Freed MM: Persistent hypertension in young spinal cord injured indi- 
viduals resulting from aortic repair. Arch Phys Med Rehabil 1 991; 72: 743- 
746. 

Hobson RW II, Croom RD III, Rich NM: Influence of heparin and low molec- 
ular weight dextran on the patency of autogenous vein grafts: Vein grafts 
in the venous system. Ann Surg 1973;178:773-776. 

Hobson RW II, Croom RD III, Swan KG: Hemodynamics of the distal arteri- 
ovenous fistula in venous reconstruction. J Surg Res 1973;14:483-489. 

Hobson RW II, Israel MR, Lynch TG: Axillosubclavian arterial aneurysms. New 
York, Grune & Stratton, 1982. 

Hobson RW II, Lee BC, Lynch TG, et al: Use of intermittent pneumatic com- 
pression of the calf in femoral venous reconstruction. Surg Gynecol Obstet 
1984;159:284-286. 

Hobson RW II, Rich NM: Traumatismes veineux des membres inferieurs. In 
Kieffer E (ed): Traumatismes Arteriels. Paris, AERCV, 1995. 

Hobson RW II, Rich NM, Wright CB: Venous Trauma: Pathophysiology, Diag- 
nosis and Surgical Management. Mt. Kisco, NY, Futura Publishing, 1983. 

Hobson RW II, Rich NM, Wright CB: Complications of direct venous recon- 
struction. In Complications of Vascular Surgery. Orlando, Grune & 
Stratton, 1985, pp 351-367. 

Hobson RW II, Howard EW, Wright CB, et al: Pathophysiology of venous liga- 
tion: Significance in arterial/venous injuries. Surgery 1972;74:824. 

Hobson RWII, Howard EW, Wright CB, et al: Hemodynamics of canine femoral 
venous ligation: Significance in combined arterial and venous injuries. 
Surgery 1973;74:824-829. 

Hobson RW II, Wright CB: Peripheral side to side arteriovenous fistula: Hemo- 
dynamics and application in venous reconstruction. Am J Surg 1973;126:411- 
414. 



RE.qxd 4/16/04 3:19 PM Page 584 



584 VI • THE RICH REFERENCE COLLECTION 



Hobson RWII, Wright CB, Rich NM, Collins GJJr: Assessment of colonic ischemia 
during aortic surgery by Doppler ultrasound. J Surg Res 1976;20:231-235. 

Hobson RW II, Wright CB, Swan KG, et al: Current status of venous injury and 
reconstruction in the lower extremities. In BerganJJ, YaoJST (eds) : Venous 
Problems. Chicago, Year Book Medical Publishers, 1978, pp 469-484. 

Hobson RWII, Yeager RA, Lynch TG, et al: Femoral venous trauma: Techniques 
for surgical management and early results. Am J Surg 1983;146:220-224. 

Hoff SJ, Reilly MK, Merrill WH, et al: Analysis of blunt and penetrating injury 
of the innominate and subclavian arteries. Am Surg 1994;60:151-154. 

Hohf RP: Arterial injuries occurring during orthopaedic operations. Clin Orthop 
1963;28:21. 

Holcomb GW, Meacham PW, Dean RH: Penetrating popliteal artery injuries 
in children. J Pediatr Surg 1988;23:859-861. 

HollemanJH, Killebrew LH: Tibial artery injuries. Am J Surg 1982;144:362-364. 

HollermanJH, Killebrew LH: Injury to the popliteal artery. Surg Gynecol Obstet 
1981;153:392-394. 

Hollier LH: Protecting the brain and spinal cord. J Vase Surg 1987;5:524-528. 

Hollingsworth RK, Johnston WW, McCooeyJF: Traumatic saccular aneurysm 
of the thoracic aorta. J Thorac Surg 1952;24:325-345. 

Holman E: Arteriovenous Aneurysms: Abnormal Communication between the 
Arterial and Venous Circulations. New York, Macmillan, 1937. 

Holman E: Clinical and experimental observations on arteriovenous fistulae. 
Ann Surg 1940;112:840-875. 

Holman E: War injuries to arteries and their treatment. Surg Gynecol Obstet 
1942;75:183-192. 

Holman E: Contributions to cardiovascular physiology gleaned from clinical 
and experimental observations of abnormal arteriovenous communica- 
tions. J Cardiovasc Surg 1962;3:48-63. 

Holman E: Abnormal arteriovenous communications: Great variability of effects 
with particular reference to delayed development of cardiac failure. Cir- 
culation 1965;32:1001-1009. 

Holman E: Abnormal Arteriovenous Communications: Peripheral and Intrac- 
ardiac, Acquired and Congenital, 2nd ed. Springfield, 111, Charles C Thomas, 
1968. 

Holman E, Sir William Osier, Halsted WS: Two contrasting personalities. Pharos 
AOA 1971;34:134. 

Holman EF: Physiology of an arteriovenous fistula. Arch Surg 1971;7:64-82. 

Holmes TWJr, Netterville RE: Complications of first rib fracture including one 
case each of tracheoesophageal fistula and aortic arch aneurysm. J Tho- 
racic Surg 1956;32:74-91. 

Holzer CE Jr: Gunshot wound involving the abdominal aorta. Surgery 
1948;23:645-652. 

Honigman B: Mast increase mortality 7 times with heart injury. JAMA 1991;266. 

Hoover NW: Injuries of the popliteal artery associated with fractures and dis- 
locations. Surg Clin North Am 1961;41:1099-1112. 

Horowitz MD, Schultz CS, Stinson EB: Sensitivity and specificity of echocar- 
diogram diagnosis of pericardial effusion. Circulation 1974;50:239-247. 

Horsely V: The destructive effects of small projectiles. Nature 1894;1:104-108. 

HorsleyJS: Surgery of the Blood Vessels. St. Louis, Mosby, 1915. 

Horwitz JR, Black CT, Lally KP, Andrassy RJ: Venous bypass as an adjunct for 
the management of a retrohepatic venous injury in a child. J Trauma 
1995;39:584-585. 



RE.qxd 4/16/04 3:19 PM Page 585 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 585 

Hoshida S, Yamashita N, Otsu K, et al: Cholesterol feeding exacerbates myocar- 
dial injury in Zucker diabetic fatty rats. Am J Physiol Heart Circ Physiol 

2000;278:H256-H262. 
Houck WS: Blunt trauma to the popliteal artery: Review and presentation of 

four cases. Am Surg 1977;43:434-437. 
Howanitz EP, Murray KD, Galbraith TA, Myerowitz PD: Peripheral venous bullet 

embolization to the heart: Case report and review of the literature. J Vase 

Surg 1988;8:55-58. 
Howanitz EP, Buckley D, Galbraith TA, et al: Combine blunt traumatic rupture 

of the heart and aorta: Two case reports and review of the literature. 

J Trauma 1990;30:506-508. 
Howard C, Thai ER, Redman HC, Gibson P: Intra-arterial digital subtraction 

angiography in the evaluation of peripheral vascular trauma. Ann Surg 

1989;210:108-111. 
Howe HRJr, Poole GVJr, Hansen KJ, et al: Salvage of lower extremities fol- 
lowing combined orthopaedic and vascular trauma: A predictive salvage 

index. Am Surg 1987;53:205-208. 
Howe HR, Pennell PC: A superior temporary shunt for management of vascu- 
lar trauma of extremity. Arch Surg 1986;121:1212. 
Hoyt DB, Shackford SR, Davis JW, et al: Thoracotomy during trauma resusci- 
tations: An appraisal by board certified general surgeons. J Trauma 

1989;29:1318-1321. 
Huang MS, Liu M, Wu J, et al: Ultrasonography for the evaluation of hemo- 

peritoneum during resuscitation: A simple scoring system. J Trauma 

1994;36:173-177. 
Huang P, Fong C, Radmaker A: Prediction of traumatic aortic rupture from 

plain chest film findings using stepwise logistic regression. Ann Emerg Med 

1987;16:1330-1333. 
Hudson HM II, Woodson J, Hirsch E: The management of traumatic aortic tear 

in the multiply-injured patient. Ann Vase Surg 1991;5:445-448. 
Hughes CW: Acute vascular trauma in Korean War casualties: An analysis of 

180 cases. Surg Gynec Obstet 1954;99:91-100. 
Hughes CW: Use of intra-aortic balloon catheter tamponade for controlling 

intra-abdominal hemorrhage in man. Surgery 1954;36:65-68. 
Hughes CW: The primary repair of wounds of major arteries: An analysis of 

experience in Korea in 1953. Ann Surg 1955;141:297-303. 
Hughes CW: Arterial repair during the Korean War. Ann Surg 1958;147:555- 

561. 
Hughes CW: Vascular injuries in the orthopaedic patient. J Bone Joint Surg 

1958;40A:1271. 
Hughes CW: Vascular surgery in the armed forces. Milit Med 1959;124:30-46. 
Hughes CW: Acute vascular injuries: Civilian and military. J Trauma 1971 ;1 1:1 89- 

190. 
Hughes CW, Bowers WF: Traumatic Lesions of Peripheral Vessels. Springfield, 

111, Charles C Thomas, 1961. 
Hughes CW, Cohen A: The repair of injured blood vessels. Surg Clin North Am 

1958;38:1529-1543. 
Hughes CW, Jahnke EJJr: The surgery of traumatic arteriovenous fistulas and 

aneurysms: A five-year follow up study of 215 lesions. Surg Clin North Am 

1958;148:790-797. 
Hughes CW, Rich NM. The management of vascular injuries. South Med Bull 

1969;57:36. 



RE.qxd 4/16/04 3:19 PM Page 586 



586 VI • THE RICH REFERENCE COLLECTION 



Hugier: Anevrisme raterioso-veineux de l'artere femorale gauche. BullSoc Chir 

Paris 1852;2:106. 
Hugier (1848). Cited by Horsely. The destructive effects of small projectiles. 

Nature 1894;1:104-108. 
Huk I, Brovkovych V, Nanobashvili J, et al: Prostaglandin Ei reduces 

ischemia/reperfusion injury by normalizing nitric oxide and superoxide 

release. Shock 2000;14:234-242. 
Hull DA, Hyde CL: Arterial injuries in civilian practice. J Kentucky Med Assoc 

1967;65:975-978. 
Hunink MG, BosJJ: Triage of patients to angiography for detection of aortic 

rupture after blunt chest trauma: Cost-effectiveness analysis of using CT. 

AJR 1995;165:27-36. 
Hunt JP, Baker CC, Lentrz CW, et al: Thoracic aorta injuries: Management and 

outcome of 144 patients. J Trauma 1996;40:547-556. 
Hunt TK, Blaisdell FW, Okimoto J: Vascular injuries of the base of the neck. 

Arch Surg 1969;98:586-590. 
Hunt TK, Leeds FH, Wanebo HJ, Blaisdell FW: Arteriovenous fistulas of major 

vessels of the abdomen. J Trauma 1971;11:483-493. 
Hunter J: A Treatise on the Blood, Inflammation and Gunshot Wounds. London, 

George Nicol, 1974. 
Hunter JI: Cited in Power, D-Arcy. Hunter's operation for the cure of aneurysm. 

Br J Surg 1929;17:193-196. 
Hunter W: The history of an aneurysm of the aorta, with some remarks on 

aneurysms in general. Med Obs Soc Phys Lond 1757;1:323. 
Hunter W: Further observations upon a particular species of aneurysm. Med 

Obs Soc Phys Lond 1762;2:390. 
Hurwitt ES, Seidenberg B: The non operative management of two cases of catheter 

perforation of the aorta. Am J Surg 1965;110:452-455. 
Hurwitt E, Seidenberg B: Rupture of the heart during cardiac massage. Ann 

Surg 1953;137:115-119. 
HuttonJE Jr, Rich NM: Pathophysiology of wounding. In McAninch JW (ed): 

Traumatic and Reconstructive Urology. Philadelphia, WB Saunders, 1995. 
Iannettoni MD, McCurry KR, Rodriguez JL, et al: Simultaneous traumatic ascend- 
ing and descending thoracic aortic rupture. Ann Thoracic Surg 1994;57:481- 

484. 
Ikard R, Merendino KA: Accidental excision of the superior mesenteric artery: 

Technical and physiological considerations. Surg Clin North Am 1970; 

50:1075-1085. 
Ikeda Y, Young LH, Scalia R, Lefer AM: Cardioprotective effects of citrulline in 

ischemia/reperfusion injury via a non-nitric oxide-mediated mechanism. 

Methods Find Exp Clin Pharmacol 2000;22:563-571. 
Imamoglou K, Read RC, Heubl HC: Cervicomediastinal vascular surgery. Surgery 

1967;61:274-80. 
Inahara T: Arterial injuries of the upper extremity. Surgery 1962;51:605-610. 
Inberg MC, Laaksonen V, Scheinin TM, et al: Early repair of traumatic rupture 

of the thoracic aorta: Report of two cases. ScandJ Thorac Cardiovasc Surg 

1972;6:287. 
Inoue T, Kawada K, Tanaka S, et al: Clinical application of the temporary long 

external bypass method for cross-clamping of the descending thoracic aorta. 

J Thorac Cardiovasc Surg 1972;63:787-793. 
Inui FK, Shannon J, Howard JM: Arterial injuries in the Korean conflict: Expe- 
riences with 111 consecutive injuries. Surgery 1955;37:850-857. 



RE.qxd 4/16/04 3:19 PM Page 587 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 587 

Isaacs JP: Sixty penetrating wounds of the heart: Clinical and experimental 
observations. In Blalock A (ed): Recent Advances in Surgery. 1959, 
p 696. 

Isaacs JP, Swanson HS, Smith RA: Transient childhood strokes from internal 
carotid stenosis. JAMA 1969;207:1859-1862. 

Iskecelli OK: Bullet embolus of left femoral artery: Report of a case which 
occurred after abdominal gunshot wound. Arch Surg 1962;85:184-185. 

Iskersky V, Reines HD, Vujic I: Left-sided vena cava mimicking traumatic rup- 
tured thoracic aorta. J Trauma 1987;27:797-799. 

Israel. Cited in Murphy JB: Resection of arteries and veins injured in continu- 
ity-end-to-end suture: Experimental clinical research. Med Rec 1897;51: 
73. 

Itani KM, BurchJM, Spjut-Patrinely V, et al: Emergency center arteriography. 
J Trauma 1992;32:302-307. 

Ivatury RR, Shah PM, Ho K, et al: Emergency room thoracotomy for the resus- 
citation of patients with fatal penetrating injuries of the heart. Ann Thorac 
Surg 1981;32:377-385. 

Ivatury RR, Rohman M, Lankin DH, Stahl WM: Ultrasonography in the diag- 
nosis and management of post-venorrhaphy thrombosis of the protal vein. 
J Trauma 1985;25:362-365. 

Ivatury RR, Nallanthambi M, Roberge RJ: Penetrating thoracic injuries: Stabi- 
lization vs. prompt transport. J Trauma 1987;27:1066. 

Ivatury RR, Nallanthambi M, Lankin DH, et al: Portal vein injuries: Noninva- 
sive follow-up of venorrhaphy. Ann Surg 1987;206:733-737. 

Jackson FE, Mazur J: Traumatic aortic aneurysms in neurological patients. Milit 
Med 1965;130:878-886. 

Jackson MR, Olson DW, Beckett WC Jr, et al: Abdominal vascular trauma: A 
review of 106 injuries. Am Surg 1992;58:622-626. 

Jackson MR, Brengman ML, Rich NM: Delayed presentation of 50 years after 
a World War II vascular injury with intraoperative localization by duplex 
ultrasound of a traumatic false aneurysm. J Trauma 1997;43:159-161. 

Jacobs JP, Horowitz MD, Ladden DA: Intra-aortic balloon counterpulsation in 
penetrating cardiac trauma. J Cardiovasc Surg 1992;33:38-40. 

JacobsonJH, HaimovJ: Venous revascularization of the arm: Report of three 
cases. Surgery 1977;81:599-604. 

Jaeschke H: Mechanisms of reperfusion injury after warm ischemia of the liver. 
Hepatobiliary Pancreat Surg 1998;5:402-408. 

Jaggers RC, Feliciano DV, Mattox KL, et al: Injury to popliteal vessels. Arch Surg 
1982;117:657-661. 

Jahnke EJ Jr: The surgery of acute vascular injuries: A report of 77 cases. Milit 
Surg 1953;112:249-251. 

Jahnke EJJr: Late structural and functional results of arterial injuries primar- 
ily repaired. Surgery 1958;43:175-183. 

Jahnke EJJr, Fisher GW, Jones C: Acute traumatic rupture of the thoracic aorta: 
Report of six consecutive cases of successful early repair. J Thorac Car- 
diovasc Surg 1964;48:63-77. 

Jahnke EJJr, Howard JM: Primary repair of major arterial wounds: Report of 
58 battle casualties. Arch Surg 1953;66:646-649. 

Jahnke EJJr, Hughes CW, Howard JM: The rationale of arterial repair on the 
battlefield. Am J Surg 1954;87:396-401. 

Jahnke EJJr, Seeley SF: Acute vascular injuries in the Korean War: An analysis 
of 77 consecutive cases. Ann Surg 1953;138:158-177. 



RE.qxd 4/16/04 3:19 PM Page 588 



588 VI • THE RICH REFERENCE COLLECTION 

Jain SP, Roubin GS, Iyer SS: Closure of an iatrogenic femoral artery pseudoa- 
neurysm by transcutaneous coil embolization. Cathet Cardiovasc Diagn 
1996;39:317-319. 

Jamieson NV, Watson CJ, Dunn DC: Dissection of the abdominal aorta associ- 
ated with traumatic aneurysms of the iliac vessels. Case report. Acta Chir 
Scand 1986;152:473-475. 

JamousMA, Silver JR, Baker JHE: Paraplegia and traumatic rupture of the aorta: 
A disease process or surgical complication. Injury 1992;23:475-478. 

Jarrar D, Chaudry IH, Wang P: Organ dysfunction following hemorrhage and 
sepsis: Mechanisms and therapeutic approaches. IntJ Mol Med 1999;57:368- 
374. 

Jarrar D, Wang P, Chaudry IH: Hepatocellular dysfunction: Basic considera- 
tions. In Holzheimer RG, MannickJA (eds) : Surgical Treatment Evidence- 
based and Problem-oriented. Munich, Zuckschwerdt, 2001, pp 763-767. 

Jarrar D, Wang P, Song GY, et al: Inhibition of tyrosine kinase signaling after 
trauma-hemorrhage: A novel approach for improving organ function and 
decreasing susceptibility to subsequent sepsis. Ann Surg 2000;231 :399-407. 

Jarstfer BS, Rich NM: The challenge of arteriovenous fistula formation follow- 
ing disk surgery: A collective review. J Trauma 1976;16:726-733. 

JassinowskyA: Die arteriennhat: Eine experimentelle studie. InaugDissDorpat 
1889. 

Javid H: Vascular injuries of the neck. Clin Orthop 1963;28:70. 

JayJB, French SW III: Traumatic rupture of the thoracic aorta: Review of lit- 
erature and case report. Arch Surg 1954;68:657-662. 

Jensen: Ueber circulare gefassutur. Arch Klin Chir 1903;69:938-998. 

Jensen AR: Bullet wound of the abdominal aorta with survival. J Fla Med Assoc 
1963;49:656-657. 

Jensen BT: Fourteen years' survival with an untreated traumatic rupture of the 
thoracal aorta. Am J Forens Med Pathol 1988;9:58-59. 

Jeresaty RM, LissJP: Effects of brachial artery catheterization on arterial pulse 
and blood pressure in 203 patients. Am Heart J 1968;76:481-485. 

Jernigan WR, Gardner WC: Carotid artery injuries due to closed cervical trauma. 
J Trauma 1971;11:429-435. 

Jimenez E, Martin M, Krukenkamp I, Barrett J: Subxiphoid pericardiotomy 
versus echocardiography: A prospective evaluation of the diagnosis of occult 
penetrating cardiac injuries. Surgery 1990;108:676-680. 

Jimenez F, Utrilla A, Cuesta C, et al: Popliteal artery and venous aneurysm as 
a complication of arthroscopic meniscectomy.J Trauma 1988;28:1404-1405. 

JohansenK, BandykD, ThieleB, Hansen STJr: Temporary intraluminal shunts: 
Resolution of a management dilemma in complex vascular injuries.J Trauma 
1982;22:395-402. 

Johansen K, Daines M, Howey T, et al: Objective criteria accurately predict ampu- 
tation following lower extremity trauma. J Trauma 1990;30:568-573. 

Johansen K, Lynch K, Paun M, Copass M: Non-invasive vascular tests reliably 
exclude occult arterial trauma in injured extremities.J Trauma 1991;31:515- 
522. 

Johns TNP: A comparison of suture and non-suture methods for the anasto- 
mosis of veins. Surg Gynecol Obstet 1947;84:939-942. 

Johnson GJr, Peters RM, Dart CH Jr: A study of cardiac vein negative pressure 
in arterio-venous fistula. Surg Gynecol Obstet 1967;124:82-86. 

Johnson GJr, Blythe WB: Hemodynamic effects of arteriovenous shunts used 
for hemodialysis. Ann Surg 1970;171:715-723. 



RE.qxd 4/16/04 3:19 PM Page 589 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 589 

Johnson B, Thursby P: Subclavian artery injury caused by a screw in a clavicu- 
lar compression plate. Cardiovasc Surg 1996;4:414-415. 

Johnson GW, LowryJH: Rupture of the axillary artery complicating anterior 
dislocation of the shoulder. J Bone Joint Surg 1962;44B:116. 

Johnson RH, Wall MJ, Mattox KL: Innominate artery trauma: A thirty-year expe- 
rience. J Vase Surg 1993;17:134-140. 

Johnson SB, Kearney PA, Smith MD: Echocardiography in the evaluation of 
thoracic aorta. Surg Clin North Am 1995;75:193-205. 

Johnson V, Eiseman B: Evaluation of arteriovenous shunts to maintain patency 
of venous autograft. Am J Surg 1969;1 18:915. 

Johnston R, Wall M, Mattox K: Innominate artery trauma: A 30-year experi- 
ence. J Vase Surg 1993;17:134-139. 

Jones AM, Graham NJ, Looney JR: Arterial embolism of a high-velocity rifle 
bullet after a hunting accident: Case report and literature review. Am J 
Forsensic Med Pathol 1983;4:259-264. 

Jones EW, HelmsworthJ: Penetrating wounds of the heart: Thirty years' expe- 
rience. Arch Surg 1968;96:671-682. 

Jones JC: Traumatic rupture of ascending aorta and left main bronchus. Ann 
Thorac Surg 1989;47:484. 

Jones JW, Hewitt RL, Drapanas T: Cardiac contusion: A capricious syndrome. 
Ann Surg 1975;181:567-574. 

Jones RE, Smith EC, Bone GE: Vascular and orthopaedic complications of knee 
dislocation. Surg Gynecol Obstet 1979;149:554-558. 

Jones RF, Terrell JC, Walyer FE: Penetrating wounds of the neck: An analysis of 
274 cases. J Trauma 1967;7:228-234. 

Jousi M, Leppaninemi A: Management and outcome of traumatic aortic injuries. 
Ann Chirurgiae Gynaecol 2000:89. 

Jurkovich GJ, Zingarelli W, Wallace J, Curreri PW: Penetrating neck trauma: 
Diagnostic studies in the asymptomatic patients. J Trauma 1985;25:819- 
822. 

Jurkovich GJ, Hoyt DB, Moore FA, et al: Portal triad injuries. J Trauma 1995; 
39:426-434. 

Kadambi A, Skalak TC: Role of leukocytes and tissue-derived oxidants in short- 
term skeletal muscle ischemia reperfusion injury. Am J Physiol Heart Circ 
Physiol 2000;278:H435-H443. 

Kaeffer N, Richard V, Thuillez C: Delayed coronary endothelial protection 24 
hours after preconditioning: Role of free radicals. Circulation 1997;96:231 1- 
2316. 

Kahn AM, Joseph WL, Hughes RK: Traumatic aneurysms of the thoracic aorta: 
Excision and repair without graft. Ann Thorac Surg 1967;4:175-181. 

Kakkar W: The cephalic vein as a peripheral vascular graft. Surg Gynecol Obstet 
1969;128:551-556. 

Kakos GS, Williams TE Jr, Kilman JW, Kalssen KP: Traumatic left ventricular 
aneurysms after penetrating chest injury. Ann Surg 1971;174:202-206. 

Kalmar P, Otto CB, Rodewald G: Selection of proper time of operation of trau- 
matic thoracic aortic aneurysms (TTA) .J Thorac Cardiovasc Surg 1982;36- 
37. 

Kam J, Jackson H, Ben-Menachem Y: Vascular injuries in blunt pelvic trauma. 
Radiol Clin North Am 1981;19:171-186. 

Kamiya H, Hanaki Y, Kojima S: Fistula between noncoronary sinus of Valsalva 
and right atrium after blunt chest trauma. Am Heart J 1987;114:429- 
431. 



RE.qxd 4/16/04 3:19 PM Page 590 



590 VI • THE RICH REFERENCE COLLECTION 



Kang SS, Labropoulos N, Mansour MA, Baker WH: Percutaneous ultrasound 
guided thrombin injection: A new method for treating postcatheterization 
femoral pseudoaneurysms. J Vase Surg 1998;27:1032-1038. 

Kang SS, Labropoulos N: Postcatheterization femoral pseudoaneurysms: Treat- 
ment with ultrasound-guided thrombin injection. Vase Ultrasound Today 
1998;3:125-140. 

Kang SS, Labropoulos N, Mansour MA: Expanded indications for ultrasound- 
guided thrombin injection of pseudoaneurysms. J Vase Surg 2000;31:289- 
298. 

Kapp JP, Gielchinsky I, Jelsma R: Metallic fragment embolization to the cere- 
bral circulation. J Trauma 1973;13:256-261. 

Kappes S, Towne J, Adams M: Perforation of the superior vena cava: A com- 
plication of subclavian dialysis. JAMA 1983;249:2232-2233. 

Karalis DE, Victor MF, Davis GA, et al: The role of echocardiography in blunt 
chest trauma: A transthoracic and transesophageal echocardiographic study. 
J Trauma 1994;36:53-58. 

Karmody AM, Lempert N, Jarmolych J: The pathology of post-catheterization 
brachial artery occlusion. J Surg Res 1976;20:601-606. 

Karrell R, Shaffer MA, Franaszek JB: Emergency diagnosis, resuscitation, and 
treatment of acute penetrating cardiac trauma. Ann Emerg Med 
1982;11:504-517. 

KashukJL, Moore EE, MillikanJS, MooreJB: Major abdominal vascular trauma: 
A unified approach. J Trauma 1982;22:672-679. 

Kassel BA, Pugkhem T, Schechter LS, ChassinJL: Traumatic rupture of distal 
thoracic aorta. N YStateJ Med 1980;80:1615-1617. 

Kato M, Matsuda T, Kaneko M: Experimental assessment of newly devised tran- 
scatheter stent-graft for aortic dissection. Ann Thorac Surg 1995;59:908- 
915. 

Katz NM, Blackstone EH, Kirklin JW, Karp RM: Incremental risk factors for 
spinal cord injury following operation for acute traumatic aortic transec- 
tion. J Thorac Cardiovasc Surg 1981;81:669-674. 

Katz S, Mullin R, Berger RL: Traumatic transection associated with retrograde 
dissection and rupture of the aorta: Recognition and management. Ann 
Thorac Surg 1974;17:273-276. 

Kaufman JL, Dinerstein CR, Shah DM, et al: Renal artery intimal flaps after 
blunt trauma: Indications for non-operative therapy. J Vase Surg 1988;8:33- 
37. 

Kaushik VS, Mandel AK, Awariefe OA, et al: Early thoracotomy for stab wounds 
of the heart. J Cardiovasc Surg 1979;20:423-426. 

Kavic SM, Atweh N, Ivy ME, et al: Celiac axis ligation after gunshot wound to 
the abdomen: Case report and literature review. J Trauma 2001;50:738- 
739. 

Kawada T, Mieda T, Abe H, et al: Surgical experience with traumatic rupture 
of the thoracic aorta. J Cardiovasc Surg 1990;31:359-363. 

Kay JK, Dykstra PC, Tsuji HK: Retrograde ilioaortic dissection: A complication 
of common femoral artery perfusion during open-heart surgery. Am J 
Surgery 1966;111:464-468. 

Kazui T, Komatsu S, Yokoyama H: Surgical treatment of aneurysms of the tho- 
racic aorta with the aid of partial cardiopulmonary bypass: An analysis of 
95 patients. Ann Thorac Surg 1987;43:622-627. 

Kearney P, Smith D, Johnson S, et al: Use of transesophageal echocardiogra- 
phy in the evaluation of traumatic aortic injury.J Trauma 1993;34:696-703. 



RE.qxd 4/16/04 3:19 PM Page 591 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 591 

KeeleyJL: A bullet embolus to the left femoral artery following a thoracic gunshot 

wound: Probable entrance through thoracic aorta: Case report and resume 

of peripheral arterial bullet emboli. J Thorac Surg 1951;21:608-620. 
Keeley SB, Snyder WH III, Weigelt JA: Arterial injuries below the knee: Fifty- 
one patients with eighty-two injuries. J Trauma 1983;23:285-290. 
Kelly GL, Eiseman B: Civilian vascular injuries. J Trauma 1975;15:507-514. 
Kelly GL, Eiseman B: Management of small arterial injuries: Clinical and exper- 
imental studies. J Trauma 1976;16:681-685. 
Kelly JJ, Reuter KL, Waite RJ: Vascular injury complicating lumbar diskectomy: 

CT diagnosis. AJR Am J Roentgenol 1989;153:1233-1234. 
Kemmerer WT, Eckert WG, Gathright JB, et al: Patterns of thoracic injuries in 

fatal traffic accidents. J Trauma 1961;1:595-599. 
Kennedy FR, Cornwell EE, Camel J: Aortoesophageal fistula due to gunshot 

wounds: Report of two cases with one survivor. J Trauma 1995;38:971- 

974. 
KennedyJC: Complete dislocations of the knee. J Bone Joint Surg 1959;41:878. 
Kennedy JC: Complete dislocation of the knee joint. J Bone Joint Surg 

1963;45:889-904. 
Keynes G: The Apologie and Treatise of Ambroise Pare. Chicago, University of 

Chicago, 1952. 
Khalil IM, Livingston DH: Intravascular shunts in complex lower limb trauma. 

J Vase Surg 1986;4:582-587. 
Khoury G, Sfeir R, Khalifeh M, et al: Penetrating trauma to the abdominal 

vessels. Cardiovasc Surg 1996;4:405-407. 
Kieny R, Charpentier A: Traumatic lesions of the thoracic aorta: A report of 73 

cases. J Cardiovasc Surg 1991;32:613-619. 
Kikta MJ, Meyer JP, Bishara RA, et al: Crush syndrome due to limb compres- 
sion. Arch Surg 1987;122:1078-1081. 
Kilburn P, Sweeney JG, Silk FF: Three cases of compound posterior dislocation 

of the elbow with rupture of the brachial artery. J Bone Joint Surg 

1962;44:119-121. 
Killen DA: Injury of the superior mesenteric vessel secondary to non-penetrating 

abdominal trauma. Am Surg 1964;30:306-312. 
KillewichLA, Bedford GR, Beach KW, StrandnessDE: Diagnosis of deep venous 

thrombosis: A prospective study comparing duplex scanning to contrast 

venography. Circulation 1989;79:810-814. 
Kim H, Hwan KK: Role of nitric oxide and mucus in ischemia/reperfusion- 

induced gastric mucosal injury in rats. Pharmacology 2001;62:200-207. 
KinmonthJB: A report on the physiology and relief of traumatic arterial spasm. 

BrMedJ1952;l:59. 
Kipfer B, Leupi F, Schuepbach P, et al: Acute traumatic rupture of the thoracic 

aorta: Immediate or delayed surgical repair? Eur J Cardiothorac Surg 

1994;8:30-33. 
Kirkup JR: Major arterial injury complicating fracture of the femoral shaft. 

J Bone Joint Surg 1963;45:337-343. 
Kirsh MM, Behrendt DM, Orringer MB, et al: The treatment of acute traumatic 

rupture of the aorta: A 10-year experience. Ann Surg 1976;184:308. 
Kirsh MM, Crane J, Kahn DG, et al: Roentgenographic evaluation of trauma 

rupture of the aorta. Surg Gynecol Obstet 1970;131:900-904. 
Kirsh MM, Kahn DR, Crane JD, et al: Repair of acute traumatic rupture of the 

aorta without extracorporeal circulation. Ann Thorac Surg 1970;10:227- 

236. 



RE.qxd 4/16/04 3:19 PM Page 592 



592 VI • THE RICH REFERENCE COLLECTION 



Kirshner R, Seltzer S, D'Orsi C, DeWeese JA: Upper rib fractures and medi- 
astinal widening: Indications for aortography. Ann Thorac Surg 1983;35:450- 
454. 

Kitzmiller JW, Hertzer NR, Beven EG: Routine surgical management of brachial 
artery occlusion after cardiac catheterization. ArchSurg 1982; 11 7:1066-1071. 

Kizer KW, Boone HA, Heneveld E, Orozco JR: Nail gun injury to the heart. 
J Trauma 1995;38:382-383. 

Kjellstrom T, Risberg B: Vascular trauma: Review of 10 years' experience. Acta 
Chir Scand 1980;146:261. 

Klein MD, Coran AG, Whitehouse WMJr, et al: Management of iatrogenic arte- 
rial injuries in infants and children. J Pediatr Surg 1982;17:933-939. 

Klein SR, Bongard FS, White RA: Neurovascular injuries of the thoracic outlet 
and axilla. Am J Surg 1988;156:115-118. 

Klein SR, Baumgartner FJ, Bongard FS: Contemporary management strategy 
of major inferior vena caval injuries. J Trauma 1991;31:1032. 

Kleinert HE: Homograft patch repair of bullet wounds of the aorta: Experi- 
mental study and report of a case. Arch Surg 1958;76:811-820. 

Kleinert HE, Romero J: Blunt abdominal trauma. J Trauma 1961;1:226-240. 

Kleinert HE, Kasdan ML: Restoration of blood flow in upper extremity injuries. 
J Trauma 1963;3:461. 

Kleinert HE, Kasdan ML, Romero JL: Small bloodvessel anastomosis for salvage 
of severely injured upper extremity. J Bone Joint Surg 1963;45:788- 
796. 

Kleinert HE, Volianitis GJ: Thrombosis of the Palmar Arch and its tributaries: 
Etiology and newer concepts in treatment. J Trauma 1965;5:447-457. 

Kleinert HE: Vascular diseases. 1972 (unpublished paper). 

Kleinert HE, Burget GC, Morgan JA, et al: Aneurysms of the hand. Arch Surg 
1973;106:554-557. 

Kleinsasser LJ: The removal of a wire lodged in the interventricular septum of 
the heart. Surgery 1961;50:500-503. 

Kline DG, Hackett ER: Reappraisal of timing for exploration of civilian peri- 
pheral nerve injuries. Surgery 1975;78:54-65. 

Kline RM, Hertzer NR, Beven EG, et al: Surgical treatment of brachial artery 
injuries after cardiac catheterization. J Vase Surg 1990;12:20-24. 

Klingensmith W, Oles P, Martinez H: Arterial injuries associated with disloca- 
tion of the knee or fracture of the lower femur. Surg Gynecol Obstet 
1965;120:961-964. 

Klingensmith W, Oles P, Martinez H: Fractureswith associated bloodvessel injury. 
Am J Surg 1965;110:849-852. 

Kluger Y, Sagie B, Soffer D, et al: The use of hemorrhage occluder pins for con- 
trolling paravertebral intercostal artery bleeding: Case report. J Trauma 
1997;43:687. 

Knott-Craig CJ, PrzybojewskiJZ, Barnard PM: Penetrating wounds of the heart 
and great vessels: A new therapeutic approach. S Afr Med J 1982;62:316- 
320. 

Knott LH, Crawford FA, Grogan JB: Comparison of autogenous vein, Dacron 
and Gore-Tex in infected wounds. J Surg Res 1978;24:288-293. 

Knudson MM, Lewis FR, Atkinson K, Neuhaus A: The role of duplex ultrasound 
arterial imaging in patients with penetrating extremity trauma. Arch Surg 
1993;128:1033. 

Kollmeyer KR, Hunt JL: Aortoazygous fistula from gunshot wounds to the 
suprarenal abdominal aorta. J Trauma 1985;25:257-259. 



RE.qxd 4/16/04 3:19 PM Page 593 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 593 

Kootstra G, Schipper JJ, Klasen HJ, Binnendijk B: Femoral shaft fracture with 

injury of the superficial femoral artery in civilian accidents. Surg Gynecol 

Obstet 1976;142:399-403. 
Kornblith BA: Gunshot wound through the abdominal aorta. Ann Surg 

1941;113:637-640. 
Kowalenko T, Stern S, Wang X, Dronen S: Improved outcome with "hypoten- 
sive" resuscitation of uncontrolled hemorrhagic shock in the swine model. 

J Trauma 1991;31:1032. 
Kozloff L, Rich NM, BrottWH, et al: Vascular trauma secondary to diagnostic 

and therapeutic procedures: Cardiopulmonary bypass and intra-aortic 

balloon assist. Am J Surg 1980;140:302-305. 
Kozlov AV, Sobhian B, Duvigneau C, et al: Organ specific formation of nitrosyl 

complexes under intestinal ischemia/reperfusion in rats involves NOS- 

independent mechanism (s). Shock 2001;15:366-371. 
RrajewskiLP, Hertzer NR: Blunt carotid artery trauma. Ann Surg 1980;19 1:341- 

346. 
Rram HB, Appel PL, Shoemaker WC: Increased incidence of cardiac contu- 
sion in patients with traumatic thoracic aortic rupture. Ann Surg 

1988;208:615-618. 
Rram HB, Appel PL, Wohlmuth DA, Shoemaker WC: Diagnosis of traumatic 

thoracic aortic rupture: A ten-year retrospective analysis. Ann Thorac Surg 

1989;47:282-286. 
Rram HB, Wohlmuth DA, Appel PL, Shoemaker WC: Clinical and radiographic 

indications for aortography in blunt chest trauma. J Vase Surg 1987;6:168- 

176. 
Rraus TW, Paetz B, Richter GM, Allenberg JR: The isolated posttraumatic 

aneurysm of the brachiocephalic artery after blunt thoracic contusion. 

Ann Vase Surg 1993;7:275-281. 
Rrauss M: Studies in wound ballistics: Temporary cavity effects in soft tissues. 

MilitMed 1957;120:221. 
Rrige JEJ, Spence RAJ: Popliteal artery trauma: A high risk injury. Br J Surg 

1987;74:91-94. 
Rrishnasastry RV, Friedman SG, Deckoff SL, Doscher W: Traumatic juxtarenal 

aortocaval fistula and pseudoaneurysm. Ann Vase Surg 1990;4:378-380. 
Rronzon I: Diagnosis and treatment of iatrogenic femoral artery pseudoa- 
neurysm: A review. J Am Soc Echocardiogr 1997;10:236-245. 
Rropilak M, Satiani B: Combined superficial femoral artery and vein injury with 

deep venous thrombosis: Elements of proper management. Contemp Surg 

1988;32:24-28. 
Rrosnick A: Death due to migration of the ball from an aortic-valve prosthesis. 

JAMA 1965;191:1083-1084. 
Rruse-Andersen S, LorentzenJE, Rohr N: Arterial injuries of the upper extrem- 
ities. Acta Chir Scand 1983;149:473-477. 
Rudok RA, Bongard F: Determinants of survival after vena caval injury. Arch 

Surg 1984;119:1009-1012. 
Rudsk R, Sheldon G, Lim R: Atrial-caval shunting (ACS) after trauma. J Trauma 

1982;22:81-85. 
Rudsk RA, Bongard F, Lim RC Jr: Determinants of survival after vena 

caval injury: Analysis of a 14-year experience. Arch Surg 1984;119:1009- 

1012. 
Ruiper DH: Cardiac tamponade and death in a patient receiving total parenteral 

nutrition. JAMA 1974;230:877. 



RE.qxd 4/16/04 3:19 PM Page 594 



594 VI • THE RICH REFERENCE COLLECTION 



Kulick DL, Kotlewski A, Hurvitz RJ, et al: Aortic rupture following percutaneous 
catheter balloon coarctoplasty in an adult. Am Heart J 1990;119:190-193. 

Kulshrestha P, Iyer KS, Das B, et al: Chest injuries: A clinical and autopsy profile. 
J Trauma 1988;28:844-847. 

Kurtoglu M, Ertekin C, Bulut T, et al: Management of vascular injuries of the 
extremities. One hundred and fifteen cases. Int Angiol 1991;10:95. 

Kurzel RB, Edinger DD: Injury to the great vessels: A hazard of transabdomi- 
nal endoscopy. South Med J 1983;76:656-657. 

Kurzweg FT: Vascular injuries associated with penetrating wounds of the groin. 
J Trauma 1980;20:214-217. 

Kuzman M, Tomic B, Stevanovic R, et al: Fatalities in the war in Croatia, 1991 
and 1992: Underlying and external causes of death. JAMA 1993;270:626- 
628. 

Kummel: Uber circulare naht der gefasse. Munch Med Wschr 1899;46:1398. 

KwaanJHM, Connolly JE: Successful management of prosthetic graft infection 
with continuous povidone-iodine irrigation. Arch Surg 1981;116:716-720. 

Kyosola K, Jarvinen A: Abdominal aortic aneurysm and dissection after blunt 
trauma. J Cardiovasc Surg 1987;28:737-739. 

La BergeJM, Jeffrey RB: Aortic lacerations: Fatal complications of thoracic aor- 
tography. Radiology 1987;165:367-369. 

La Garde LA: Report of the Surgeon General of the Army to the Secretary of 
War. Washington, DC, Government Printing Office, 1893. 

La Garde LA: Gunshot Injuries (How They Are Inflicted, Their Complications 
and Treatment). New York, William Wood & Company, 1916. 

LacyJH, BoxJM, Connors D, et al: Pseudoaneurysm: Diagnosis with color Doppler 
ultrasound. J Cardiovasc Surg 1990;31:727-730. 

Lafani SJA, Shatzkes D, Scalea T: The removal of intravascular bullets by inter- 
ventional radiology: The prevention of central migration by balloon occlu- 
sion—Case report. J Trauma 1991;31:1423-1425. 

Lagerstrom CF, Reed RL, Rowlands BJ, Fisher R: Early fasciotomy for acute clin- 
ically evident posttraumatic compartment syndrome. AmJ Surg 1989;158:36- 
39. 

Lai MD, Hoffman HB, AdamkiewiczJJ: Dissecting aneurysm of internal carotid 
artery after non-penetrating neck injury. Acta Radiol Diag 1966;5:290-295. 

Lain KC, Williams GR: Arteriography in acute peripheral arterial injuries: An 
experimental study. Surg Forum 1970;21:179-181. 

Lam CR, Mclntyre R: Air-pistol injury of pulmonary artery and aorta. Report 
of a case with peripheral embolization of pellet and residual aorticopul- 
monary fistula. J Thorac Cardiovasc Surg 1970;59:729-732. 

Lamb RK, Pawade A, Prior AL: Intravascular missile: Apparent retrograde course 
from the left ventricle. Thorax 1988;43:499-500. 

Lancey RA, Davliakos GP, Vander Salm TJ: Simultaneous repair of multiple trau- 
matic aortic tears. Ann Thorac Surg 1995;60:1120-1121. 

Landreneau R, Mitchum P, Fry W: Iliac arterial transposition. Arch Surg 
1989;124:978-981. 

Lange R, Bach A, Hansen S, Johansen K: Open tibial fractures with associated 
vascular injuries: Prognosis of limb salvage. J Trauma 1985;25:203-207. 

Lange R: Limb reconstruction versus amputation decision making in massive 
lower extremity trauma. Clin Orthop 1989;243:92-99. 

Langenbeck B: Beitrage zur chirurgischen pathologie der venen. Arch Klin 
Chirl861;l:l. 

Langley G: Gunshotwound of the innominate artery. Br MedJ 1943;2:711-712. 



RE.qxd 4/16/04 3:19 PM Page 595 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 595 

LaRoque G: Penetrating bullet wound of the thoracic aorta followed by lodg- 
ment of the bullet in the femoral artery. Ann Surg 1931;4:16. 

Larrey D: Sur une blessure du pericorde suivie d'hydropericarde. Bull Sci Med 
1810;6:1. 

Larrey D: Clin Chir Paris 1829;2:284. 

Lassonde J, Laurendeau F: Blunt injury of the abdominal aorta. Am Surg 
1981;194:745-748. 

Lassonde J, Morin M, Laurendeau F: Arterial injuries. Can J Surg 1984;27:343- 
345. 

Lau J, Mattox KL, Beall AC Jr, DeBakey ME: Use of substitute conduits in trau- 
matic vascular injury. J Trauma 1977;17:541-546. 

Launois B, Chateaubriant P, Rosat P, Kiroff G: Repair of suprahepatic caval 
lesions under extracorporeal circulation in major trauma. J Trauma 
1989;29:127-128. 

Lavenson GJ, Rich NM, BaughJH: Value of ultrasonic flow detector in the man- 
agement of peripheral vascular disease. Am J Surg 1970;120:522. 

Lavenson GJ, Rich NM, Strandness DE Jr: Ultrasonic flow detector value in 
combat vascular injuries. Arch Surg 1971;103:644-647. 

Laverick MD, D'sa Barros, Kirk SJ, Mollan RAB: Management of blunt injuries 
of the axillary and the neck of the humerus: Case report. J Trauma 
1990;30:360-361. 

Lawrence K, Shefts L, McDanielJ: Wounds of common carotid arteries: Report 
of 17 cases from World War II. Am J Surg 1948;76:29-37. 

Lawton R, Rossi N, Funk D: Intracardiac perforation. Arch Surg 1969;98:213- 
216. 

Layton T, Stroh A, Villella E: Missile embolus of the portal vein. J Trauma 
1985;25:1111-1112. 

Le Bret F, Ruel P, Rosier H, et al: Diagnosis of traumatic mediastinal hematoma 
with transesophageal echocardiography. Chest 1994;105:373-376. 

Learmonth J: Combined neuro-vascular lesions. Acta Chir Scand 1952;104:93- 
99. 

Learmonth J: An unusual type arteriovenous communication. Br J Surg 
1945;32:321-323. 

Learmonth J: Vascular injuries in war. Royal Soc Med 1946;39:488. 

Leavitt B, Meyer J, Morton JR, et al: Survival following nonpenetrating trau- 
matic rupture of cardiac chambers. Ann Thorac Surg 1987;44:532-535. 

Leblanc J, Wood A, O'Shea M, Williams W: Peripheral arterial trauma in chil- 
dren. J Cardiovasc Surg 1985;26:325-331. 

Ledgerwood A, Lucas C: Survival following superior mesenteric artery occlu- 
sion from trauma. J Trauma 1974;14:622-626. 

Ledgerwood A, Lucas C: Biological dressings for exposed vascular grafts: A rea- 
sonable alternative. J Trauma 1975;15:567-574. 

Ledgerwood A, Kazmers M, Lucas C: The role of thoracic aortic occlusion for 
massive hemoperitoneum. J Trauma 1976;16:610-615. 

Ledgerwood A: The wandering bullet. Surg Clin North Am 1977;57:97-109. 

Ledgerwood A, Mullins R, Lucas C: Primary repair vs ligation for carotid artery 
injuries. Arch Surg 1980;115:488-493. 

Lee F, Katzberg R, Gutierrez O, et al: Reevaluation of plain radiographic find- 
ings in the diagnosis of aortic rupture: The role of inspiration and posi- 
tioning on mediastinal width. J Emerg Med 1993;11:289-296. 

Lee H, Beale LS: On the repair of arteries and veins after injury. Trans Med 
Chir Soc 1865;50:477. 



RE.qxd 4/16/04 3:19 PM Page 596 



596 VI • THE RICH REFERENCE COLLECTION 



Lee R, Stahlman G, Sharp K: Treatment priorities in patients with traumatic 

rupture of the thoracic aorta. Am Surg 1992;58:37-43. 
Lefer A, Lefer D: Pharmacology of the endothelium in ischemia-reperfusion 

and circulatory shock. Ann Rev Pharmacol Toxicol 1993;33:71-90. 
Lefer A: Endotoxin, Cytokines, and nitric oxide in shock. Shock 1994;1:79-80. 
Lefer A: Role of the beta2-integrins and immunoglobulin superfamily members 

in myocardial ischemia-reperfusion. Ann Thorac Surg 1999;68:1920-1923. 
Lehr HA, Guhlmann A, Nolte D, et al: Leukotrienes as mediators in ischemia- 
reperfusion injury in a microcirculation model in the hamster. J Clin Invest 

1991;87:2036-2041. 
LemaireJ, Dondelinger R: Percutaneous coil embolization of iatrogenic femoral 

arteriovenous fistula or pseudo-aneurysm. Eur J Radiol 1994;18:96-100. 
Lemos P, Okumura M, Azevedo AC, et al: Cardiac wounds: Experience based 

on a series of 121 operated cases. J Cardiovasc Surg 1976;17:1-8. 
Lennox A, Griffin M, Nicolaides A, Mansfield A: Percutaneous ultrasound guided 

thrombin injection: A new method for treating postcatheterization femoral 

pseudoaneurysms. J Vase Surg 1998;28:1120-1121. 
Lennox.AF, Griffin M, Cheshire N: Treatment of an iatrogenic femoral artery 

pseudoaneurysm with percutaneous duplex-guided injection of thrombin. 

Circulation 1999;100:39-41. 
Lennox.AF, Delis K, Szendro G: Duplex-guided thrombin injection for iatro- 
genic femoral artery pseudoaneurysm is effective even in anticoagulated 

patients. Br J Surg 2001;87:796-801. 
Leppaniemi A, Wherry D, Pikoulis E, et al: Arterial and venous repair with vas- 
cular clips: Comparison with suture closure. J Vase Surg 1997;26:24-28. 
Leppaniemi A, Wherry D, Pikoulis E, et al: Common bile duct repair with tita- 
nium staples: Comparison with suture closure. Surg Endosc 1997;11:714- 

717. 
Leppaniemi AK, Salo J, Haapiainen R: Civilian low velocity gunshot wounds of 

the liver. Eur J Surg 1994;160:663-668. 
Leppaniemi AK, Savolainen H, Salo J: Traumatic inferior vena caval injuries. 

Scand J Thorac Cardiovasc Surg 1994;28:103-108. 
Leppaniemi AK, Rich NM, Pikoulis E, et al: Sutureless vascular reconstruction 

with titanium clips. Int Angiol 2000;19:69-74. 
Lester J: Arteriovenous fistula after percutaneous vertebral angiography. Acta 

Radiol Diag 1966;5:337-340. 
Letsou G, GertlerJ, Baker CC, Hammond G: Blunt innominate injury: A report 

of three cases. J Trauma 1989;29:104-108. 
Letsou G, Gusberg R: Isolated bilateral renal artery thrombosis: An unusual 

consequence of blunt abdominal trauma — Case report. J Trauma 

1990;30:509-511. 
Lev El A, Adar R, Rubinstein Z: Axillary artery injury in erect dislocation of the 

shoulder. J Trauma 1981;21:323-324. 
LeVeen H, Cerruti M: Surgery of large inaccessible arteriovenous fistulas. Ann 

Surg 1963;158:285-289. 
Levin A, Gover P, Nance F: Surgical restraint in the management of hepatic 

injury: A review of the Charity Hospital experience. J Trauma 1978;18:399- 

404. 
Levin L, Goldner R, Urbaniak J, et al: Management of severe musculoskeletal 

injuries of the upper extremity. J Orthop Trauma 1990;4:432. 
Levin P, Rich NM, Hutton JE Jr, et al: Role of arteriovenous shunts in venous 

reconstruction. Am J Surg 1971;122:183-191. 



RE.qxd 4/16/04 3:19 PM Page 597 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 597 

Levin P, Rich NM, Hutton JE Jr: The role of collateral circulation in arterial 

injuries. Arch Surg 1971;102:392-398. 
Levin P, Rich NM, Hutton JE Jr: Patency of venous grafts in the venous system. 

J Cardiovasc Surg (Torino) 1972;13:421-427. 
Levine E, Alverdy JC: Carotid-esophageal fistula following a penetrating neck 

injury: Case report. J Trauma 1990;30:1588-1590. 
Levitsky S, James P, Anderson RW, Hardeway RI: Vascular trauma in Vietnam 

battle casualties: An analysis of 55 consecutive cases. Ann Surg 1968;1 68:831- 

836. 
Lewis D, Davies A, Irvine C: Compression ultrasonography for false femoral 

artery aneurysms: Hypocoagulability is a cause of failure. Eur J Vase Surg 

1998;16:427-428. 
Lewis T: The adjustment of blood flow to the affected limb in arteriovenous 

fistula. Clin Sci 1940;4:277-285. 
LewtasJ: Traumatic subclavian aneurysm: Ligature of innominate and carotid 

arteries: Recovery. Br Med J 1889;2:312. 
Lexer E: Die ideale operation des arteriellen und des arteriell-venosen 

aneurysma. Arch Klin Chir 1907;83:459-477. 
Liau C, Ho F, Chen M, Lee Y: Treatment of iatrogenic femoral artery pseudoa- 

neurysm with percutaneous thrombin injection. J Vase Surg 1997;26:18- 

23. 
Lichtenstein M: Acute injuries involving the large blood vessels in the neck. 

Surg Gynecol Obstet 1947;85:165-175. 
Lichti E, Erickson T: Traumatic arteriovenous fistula: Clinical evaluation and 

intraoperative monitoring with the Doppler ultrasonic flowmeter. Am J 

Surg 1974;127:333-335. 
Liddicoat J, Bekassy SM, Daniell M, DeBakey ME: Inadvertent femoral artery 

"stripping": Surgical management. Surgery 1975;77:318-320. 
LiekwegW, Greenfield L: Management of penetrating carotid arterial injuries. 

Ann Surg 1966;188:587-592. 
Lilienthal H: Thoracic surgery: The surgical treatment of thoracic disease. 

Philadelphia: WB Saunders Company, 1926. 
Lillehei K, Robinson M: A critical analysis of the fatal injuries resulting from 

the Continental flight 1713 airline disaster: Evidence in favor of improved 

passenger restraint system. J Trauma 1994;37:826-830. 
Lim LT, Michuda M, Flanigan DP, Pankovich A: Popliteal artery trauma: 31 con- 
secutive cases without amputation. Arch Surg 1980;115:1307-1313. 
Lim LT, SalettaJ, Flanigan DP: Subclavian and innominate artery trauma. Surgery 

1979;86:890-897. 
Lim RC, Glickman M, Hunt TK: Angiography in patients with bunt trauma to 

the chest and abdomen. Surg Clin North Am 1972;52:551-565. 
Lim RC, Miller S: Management of acute civilian vascular injuries. Surg Clin 

North Am 1982;62:113-119. 
Lim RC, Trunkey DD, Blaisdell FW: Acute abdominal aortic injury: An analy- 
sis of operative and post-operative management. Arch Surg 1974;109:706- 

712. 
Linberg E: Bullet wound of the thoracic aorta with survival. Maryland State 

MedJ 1959;8:285-286. 
LindenJ: Molecular approach to adenosine receptors: Receptor-mediated mech- 
anisms of tissue protection. Ann Rev Pharmacol Toxicol 2001;41:775-787. 
Lindenauer S, Thompson N, Kraft R: Late complications of traumatic arteri- 
ovenous fistulas. Surg Gynecol Obstet 1969;129:525-532. 



RE.qxd 4/16/04 3:19 PM Page 598 



598 VI • THE RICH REFERENCE COLLECTION 



Lindenbaum B: Complications of knee joint arthroscopy. Clin Orthop Rel Res 

1981;160:157. 
Lindenbaum G, Jacobs L, Morris M: Perioperative surface and transesophageal 

color-flow Doppler evaluation of posttraumatic intracardiac shunt. Am Heart 

J 1990;119:193-196. 
Lindskog G: The surgery of the innominate artery. N Engl J Med 1946;235:71- 

76. 
Lindskog G, Liebow A, Glenn W: Thoracic and Cardiovascular Surgery with 

Related Pathology. New York, Appleton-Century-Crofts, 1962. 
Linker R, Crawford FJr, Rittenbury M, Barton M: Traumatic aortocaval fistula: 

case report. J Trauma 1989;29:255-257. 
Linson M: Axillary artery thrombosis after fracture of the humerus. J Bonejoint 

Surg 1980;62-A:1214-1215. 
Linton R: Injuries to major arteries and their treatment. N YJ Med 1949;49:2039. 
Linton R: Arterial injuries associated with fractures of the extremity. J Bone 

Joint Surg 1964;46:575-576. 
Lipscomb P, Burleson R: Vascular and neural complications in the supracondylar 

fractures of the humerus in children. J Bonejoint Surg 1955;37:487-497. 
Litchford B, Okies JE, Sugimura S, Starr A: Acute aortic dissection from cross 

clamp injury. J Thorac Cardiovasc Surg 1976;72:709-713. 
Little J, Ferguson D: The incidence of hypothenar hammer syndrome. Arch 

Surg 1972;105:684. 
Lizama V, Zerbini M, Gagliardi R, Howell L: Popliteal vein thrombosis and 

popliteal artery pseudoaneurysm complicating osteochondroma of the 

femur. AJR Am J Roentgenol 1987;148:783-784. 
Lloyd J: Traumatic peripheral aneurysms. Am J Surg 1987;93:755-764. 
LockJ, Huffman A, Johnson R: Blunt trauma to the abdominal aorta. J Trauma 

1987;27:674-677. 
Loello F, Nunn D: False aneurysm of the inferior epigastric artery as a com- 
plication of abdominal retention sutures. Surgery 1973;74:460. 
LohseJ, Botham RJ, Waters R: Traumatic bilateral renal artery thrombosis. J 

Urol 1982;127:522. 
Loose H, Haslam P: The management of peripheral arterial aneurysms using 

percutaneous injection of fibrin adhesive. BrJ Radial 1998;17:1255-1259. 
Lopez-Viegro M, SynderWH, Clagett GP: Penetrating abdominal aortic trauma: 

A review of 129 patients. J Vase Surg 1992;15:247-248. 
Lord JW, Stone P, Clouthier W, Breidenbach L: Major blood vessel injury during 

elective surgery. Arch Surg 1958;77:282. 
Lord R, Ehrenfeld W, Wylie EJ: Arterial injury from the Fogarty catheter. Med 

J Aust 1968;2:70-71. 
Lord R, Irana C: Assessment of arterial injury in limb trauma. J Trauma 

1974;14:1042-1053. 
Lorenz P, Steinmetz B, Lieberman J: Emergency thoracotomy: Survival corre- 
lates with physiologic status. J Trauma 1992;32:780. 
Louridas G, Perry MO: Basic data related to vascular trauma. Ann Vase Surg 

1989;3:397. 
Love C, Evans S: Gunshot wound of the abdominal aorta and anoxic cardiac 

arrest: Report of a survival. Ann Surg 1963;158:131-132. 
Love L, Braun T: Arteriography of peripheral vascular trauma. AJR Am J 

Roentgenol 1968;102:431-440. 
Lovric Z: Reconstruction of major arteries of extremities after war injuries. 

J Cardiovasc Surg (Turin) 1993;34:33-37. 



RE.qxd 4/16/04 3:19 PM Page 599 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 599 

Lovric Z, Wertheimer B, Candric K, et al: Reconstruction of the popliteal artery 

after war injury. Unfallchirurg 1993;97:375-377. 
Lovric Z, Wertheimer B, Candric K, et al: War injuries of major extremity vessels. 

J Trauma 1994;36:248-251. 
Lowen H, Fink S, Helpern M: Transfixion of the heart by embedded ice pick 

blade with eight months' survival. Circulation 1950;2:426-433. 
Lozman H, Beaufils AT, Rossi G, et al: Vascular trauma observed at an urban 

hospital center. Surg Gynecol Obstet 1978;146:237-240. 
Lozman H, Robbins H: Injury to the superior gluteal artery as a complication 

of total hip replacement arthroplasty: A case report. J Bone Joint Surg 

1983;65:268-269. 
Lucas A, Richardson JD, Flint LM, Polk H: Traumatic injury of the proximal 

superior mesenteric artery. Ann Surg 1981;193:30-34. 
Lucas R, Tumacder O, Wilson G: Hepatic artery occlusion following hepatic 

artery catheterization. Ann Surg 1971;173:238-243. 
Ludewig RM, Wangensteen S: Aortic bleeding and the effect of external counter 

pressure. Surg Gynecol Obstet 1969;128:252-258. 
Luetic V, Sosa T, Tonkovic I, et al: Military vascular injuries in Croatia. Cardio- 

vasc Surg 1993;1:3-6. 
Luke J: Arterial trauma. J Cardiovasc Surg 1962;3:165-168. 
Lumpkin M, Logan W, Couves C, Howard JM: Arteriography as an aid in the 

diagnosis and localization of acute arterial injuries. Ann Surg 1958; 147:353- 

358. 
Lumsden A, Miller J, Kosinski A: A prospective evaluation of surgically treated 

groin complications following percutaneous cardiac procedures. Am Surg 

1994;60:132-137. 
Lundell C, Quinn M, Finck E: Traumatic laceration of the ascending aorta: 

Angiographic assessment. AJR Am J Roentgenol 1985;145:715-719. 
Lupetin A, Beckman I, Daffner R: CT diagnosis of traumatic abdominal aortic 

rupture. J Comput Assist Tomogr 1990;14:313-314. 
Lynch K, Johansen K: Can Doppler pressure measurement replace "exclusion" 

arteriography in extremity trauma? Ann Surg 1991;214:737-741. 
Lynch K, Johansen K: Can non-invasive vascular tests replace screening arteri- 
ography in extremity trauma? J Vase Surg 9999. 
Lyons C, Perkins R: Cardiac stab wounds. Am Surg 1957;23:507. 
Macbeth A, Malone J, Norton L, Peltier L: Paralysis and aortic thrombosis fol- 
lowing blunt abdominal trauma. J Trauma 1982;22:591-594. 
MacDonell J: Traumatic aortic aneurysm. Can Med Assoc J 1956;75:581- 

584. 
MacGowan W: Acute ischemia complicating limb trauma. J Bone Joint Surg 

1968;50-B:472-484. 
Machiedo G, Jain K, Swan KG, et al: Traumatic aortocaval fistula. J Trauma 

1983;23:243-247. 
Machle W: Lead absorption from bullets lodged in tissues: Report of two cases. 

JAMA1940;115:1536. 
Machleder HI, SweeneyJ, Barker WF: Pulseless arm after brachial artery catheter- 
ization. Lancet 1972;1:407. 
MacLachlin A, Carroll S, Meades G, Amacher AL: Valve replacement in the 

recanalized incompetent superficial femoral vein in dogs. Ann Surg 

1965;162:446. 
MacLean L: The diagnosis and treatment of arterial injuries. Can Med Assoc J 

1963;88:1091-1101. 



RE.qxd 4/16/04 3:19PM PageSOO 



600 VI • THE RICH REFERENCE COLLECTION 



MacLean L, Flam R, Petersen D: Diagnosis and treatment of arterial injuries. 

Minn Med 1961;44:133-142. 
Maggisano R, Nathans A, Alexandrova NA, et al: Traumatic rupture of the tho- 
racic aorta: Should one always operate immediately? Ann Vase Surg 1995;9: 

44-52. 
Magilligan D, DavilaJ: Innominate artery disruption due to blunt trauma. Arch 

Surg 1979;114:307-309. 
Mahoney B, Gerdes D, Roller B, Ruiz E: Aortic compressor for aortic occlusion 

in hemorrhagic shock. Ann Emerg Med 1984;13:11-16. 
MajeskiJA, Gants A: A management of peripheral arterial vascular injuries with 

Javid shunt. Am J Surg 1979;138:324-345. 
Makins G: Blessures des vaisseaux. Comptes-endus conference Chirurgicale 

Interalliee pour l'etude des Plaies de Guerre. Arch Med Pharm Mil 

1917;68:341. 
Makins G: Influence exerted by military experience of John Hunter himself 

and the military surgeon of today. Lancet 1917;1:249-254. 
Makins G: Gunshot injuries to the blood vessels. Bristol, England, John Wright 

and Sons, 1919. 
Makins G: Injuries to the blood vessels. Official History of the Great War Medical 

Service. Surgery of the War, vol 2. London, His Majesty's Stationary Office, 

1922, pp 170-296. 
Makins G: Specimen showing the effects of gunshot injury on the heart and 

blood vessels. Now on exhibit in the museum of the Royal College of Sur- 
geons of England. Br J Surg 1977;18:141-146. 
Makins G, Howard JM, Green R: Arterial injuries complicating fractures and 

dislocations: The necessity for a more aggressive approach. Surgery 

1966;59:203-209. 
Maleux G, Soula P, Otal P, et al: Traumatic aortobiliac dissection treated by 

kissing-stent placement. J Trauma 1997;43:706-708. 
Malt R, McKhann C: A report on replantation of severed arms. JAMA 

1964;189:716-722. 
Man B, Kraus L, Shachor D: Self-inflicted stab wound of the abdominal aorta. 

J Cardiovasc Surg 1978;19:503-505. 
Mandal A, Boitano MA: Reappraisal of low-velocity gunshot wounds of the 

aorta and inferior vena cava in civilian practice. J Trauma 1978;18:580- 

585. 
Mandal A, Oparah S: Unusually low mortality of penetrating wounds of 

the chest: Twelve years experience. J Thorac Cardiovasc Surg 1989;1: 

119. 
Mandelbaum I, Kalsbeck J: Extrinsic compression of internal carotid artery. 

Ann Surg 1970;171:434-437. 
Manlove G, Quattlebaum F, Flom R, LaFaveJ: Gunshot wounds of the abdom- 
inal aorta. Am J Surg 1960;99:941-944. 
Mansberger A, Linberg E: First rib resection for distal exposure of subclavian 

vessels. Surg Gynecol Obstet 1965;120:579. 
Mansour MA, Moore FA, Moore EE: Hypogastric arterial embolization in pelvic 

fracture hemorrhage: Case report. J Trauma 1990;30:1417-1418. 
Marcove R, Lindeque B, Silane M: Pseudoaneurysm of the popliteal artery with 

an unusual arteriographic presentation. Clin Orthop Related Res 

1988;234:142-144. 
Margolies M, Ring E, Waltman A: Arteriography in the management of hem- 
orrhage from pelvic fractures. N Engl J Med 1972;287:317-321. 



RE.qxd 4/16/04 3:19PM Page SOI 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 601 

Marin M, Veith F, CynamonJ, et al: Initial experience with transluminally placed 

endovascular grafts for the treatment of complex vascular lesions. J Vase 

Surg 1995;222:449-465. 
Marin M, Veith F, Panetta T: Transluminally placed endovascular stented graft 

repair for arterial trauma. J Vase Surg 1994;20:466-473. 
Marin M, Veith F, Panetta T, et al: Percutaneous transfemoral insertion of a 

stented graft to repair a traumatic femoral arteriovenous fistula. J Vase Surg 

1993;18:299-302. 
Markey JJ, Hines J, Nance C: Penetrating neck wounds: A review of 218 cases. 

Am Surg 1975;41:77-83. 
Marnocha K, Maglinte D: Plain-film criteria for excluding aortic rupture in 

blunt chest trauma. AJR Am J Roentgenol 1985;144:19-21. 
Marnocha K, Maglinte D, Woods J, et al: Blunt chest trauma and suspected 

aortic rupture: Reliability of chest radiographs finds. Ann Emerg Med 

1985;14:644-649. 
Marsh C, Moore R: Deceleration trauma. Am J Surg 1957;93:623. 
Marsh D, Sturm J: Traumatic aortic rupture: Roentgenographic indications for 

angiography. Ann Thorac Surg 1976;21:337-340. 
Martin L, McKenney M, SosaJ, et al: Management of lower extremity arterial 

trauma. J Trauma 1994;37:591-599. 
Martin RR, Mattox KL, Burch JM, Richardson R: Advances in treatment of vas- 
cular injuries from blunt and penetrating limb trauma. World J Surg 

1992;16:930-937. 
Martin R, Flynn T, Rowlands G: Blunt cardiac rupture. J Trauma 1984;24:287- 

290. 
Marts B, Durham R, Shapiro M, et al: Computed tomography in the diagnosis 

of blunt thoracic injury. Am J Surg 1994;168:688-692. 
Marty-Ane C, Alric P, Prudhomme M, et al: Intravascular stenting of traumatic 

abdominal dissection. J Vase Surg 1996;23:156-161. 
Marty-Ane C, Serres-Cousine O, Laborde J, et al: Endovascular stent for use in 

acute aortic dissection: An experimental study. Ann Vase Surg 1994;8:434- 

442. 
Marvasti M, Meyer J, Ford B, Parker FJ: Spinal cord ischemia following opera- 
tion for traumatic aortic transection. Ann Thorac Surg 1986;42:425-428. 
Marvasti M, Parker FJ, Bredenberg CE: Injury to arterial branches of aortic 

arch. J Thorac Cardiovasc Surg 1984;32:293-298. 
Marymont J, Coder H, Harris JJ, et al: Posterior hip dislocation associated with 

acute traumatic injury of the thoracic aorta: A previously unrecognized 

injury complex. J Orthop Trauma 1990;4:383-387. 
MarzelleJ, Nottin R, Dartevelle P, et al: Combined ascending aorta rupture and 

left main bronchus disruption from blunt chest trauma. Ann Thorac Surg 

1989;1989:769-771. 
Massberg S, Enders G, Matos F, et al: Fibrinogen deposition at the postischemic 

vessel wall promotes platelet adhesion during ischemia-reperfusion in vivo. 

Blood 1999;94:3829-838. 
Massberg S, Messmer K: The nature of ischemia/reperfusion injury. Transplant 

Proc 1998;30:4217-4223. 
Massberg S, Sausbier M, Klatt P, et al: Increased adhesion and aggregation of 

platelets lacking cyclic guanosine 3',5'-monophosphate kinase I.J Exp Med 

1999;189:1255-1264. 
Matas R: Traumatic aneurysm of the left brachial artery — Incision and partial 

excision of sac: Recovery. Phil Med News 1888;53:462-466. 



RE.qxd 4/16/04 3:19PM PageS02 



602 VI • THE RICH REFERENCE COLLECTION 



Matas R: Traumatisms and traumatic aneurysms of the vertebral artery and 

their surgical treatment. Ann Surg 1893;18:477-521. 
Matas R: Traumatic arteriovenous aneurysms of the subclavian vessels, with an 

analytical study of fifteen reported cases, including one operated. Trans 

Am Surg Assoc 1901;19:237. 
Matas R: An operation for radical cure of aneurysm based on arteriography. 

Ann Surg 1903;37:161-196. 
Matas R: Recent advances in the technique of thoracotomy and pericardiotomy 

for wounds of the heart. South Med J 1908;1:75-81. 
Matas R: Surgery of the vascular system. In Keen WW, DaCosta JC (eds) : 

Philadelphia, WB Saunders, 1909, p 67. 
Matas R: Testing the efficiency of the collateral circulation as a preliminary to 

the occlusion of the great surgical arteries. Ann Surg 1911;53:1-43. 
Matas R: Testing the efficiency of the collateral circulation as a preliminary to 

the occlusion of the great surgical arteries. JAMA 1914;68:1441-1447. 
Matas R: Endoaneurysmorrhaphy, I: Statistics of operation of endoaneurysm- 

orrhaphy, II: Personal experiences and observations on the treatment of 

arteriovenous aneurysms by the intravascular method of suture: With special 

references to the transvenous route. Surg Gynecol Obstet 1920;30:456-458. 
Matas R: Military Surgery of the Vascular System. Philadelphia, WB Saunders, 

1921. 
Mathieu D, Wattel F, Bouachour G, et al: Post-traumatic limb ischemia: Pre- 
diction of final outcome by transcutaneous oxygen measurements in hyper- 
baric oxygen. J Trauma 1990;30:307-314. 
Mathur A, Pochaczevsky R, Levotitz B, Feraru F: Fogarty balloon catheter for 

removal of catheter fragment in subclavian vein. JAMA 1971;217:481. 
Matloff D, Morton J: Acute trauma to the subclavian arteries. Am J Surg 

1968;115:675-680. 
Matolo M, Danto L, Wolfman E. Traumatic aneurysm of the abdominal aorta: 

Report of two cases and review of the literature. Arch Surg 1974;108:867- 

869. 
MatsubaraJ, Seko T, Ohta T, et al: Traumatic aneurysm of the abdominal aorta 

with acute thrombosis of bilateral iliac arteries. Arch Surg 1983;1 18:1337- 

1339. 
Mattox KL: Comparison of techniques of autotransfusion. Surgery 1978;84:700- 

702. 
Mattox KL: Emergency department thoracotomy. JACEP 1978;7:455. 
Mattox KL: Abdominal venous injuries. Surgery 1982;91:497-501. 
Mattox KL: CPR: Not for everyone. Emerg Med 1983;15:147-150. 
Mattox KL: Thoracic great vessel injury. Surg Clin North Am 1988;68:693- 

703. 
Mattox KL: Thoracic vascular trauma. J Vase Surg 1988;7:725-729. 
Mattox KL: Approaches to trauma involving the major vessels of the thorax. 

Surg Clin North Am 1989;69:77-92. 
Mattox KL: Fact and Fiction about management of aortic transection. Ann Thorac 

Surg 1989;48:1-3. 
Mattox KL: Indications for thoracotomy: Deciding to operate. Surg Clin North 

Am 1989;1:47. 
Mattox KL: Invited editorial comment: What drives the need for new technol- 
ogy? J Trauma 1992;32:761-768. 
Mattox KL: The abbreviated injury scale, 1985 revision: A condensed chart for 

clinical use. J Trauma 1997;42:353-368. 



RE.qxd 4/16/04 3:19PM PageS03 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 603 

Mattox KL, Allen MK: Penetrating wounds of the thorax. Injury 1986;174:313- 

317. 
Mattox KL, Beall AC Jr, Ennix C, DeBakey ME: Intravascular migratory bullets. 

Am J Surg 1979;137:192-195. 
Mattox KL, Beall AC Jr, Jordan GLJr, DeBakey ME: Cardiorrhaphy in the emer- 
gency center. J Thorac Cardiovasc Surg 1974;68:886-895. 
Mattox KL, Bickell W, Pepe PE, et al: Prospective MAST study in 911 patients. 

J Trauma 1989;20:1104. 
Mattox KL, Bickell WH, Pepe PE, Mangelsdorff A: Prospective randomized 

evaluation of antishock MAST in posttraumatic hypotension. J Trauma 

1986;26:779-786. 
Mattox KL, BurchJM, Richardson R, Martin RR: Retroperitoneal vascular injury. 

Surg Clin North Am 1990;70:635-653. 
Mattox KL, Espada R, Beall AC Jr: Performing thoracotomy in the emergency 

center. J Am Coll Emerg Phys 1974;3:13-17. 
Mattox KL, Espada R, Beall ACJr: Traumatic injury to the portal vein. Ann Surg 

1975;181:519-522. 
Mattox KL, Feliciano DV: Role of external cardiac compression in truncal trauma. 

J Trauma 1982;22:934-935. 
Mattox KL, Feliciano DV, Burch J, et al: Five thousand seven hundred sixty car- 
diovascular injuries in 4459 patients: Epidemiologic evolution 1958 to 1987. 

Ann Surg 1989;209:698-707. 
Mattox KL, Holzman M, Pickard LR, et al: Clamp /repair: A safe technique for 

treatment of blunt injury to the descending thoracic aorta. Ann Thorac 

Surg 1985;40:456-465. 
Mattox KL, Limacher M, Feliciano D: Cardiac evaluation following heart injury. 

J Trauma 1985;25:758-765. 
Mattox KL, Maningas P, Moore EE, et al: Prehospital hypertonic saline/dextran 

infusion for post-traumatic hypotension. The USA multicenter trial. Ann 

Surg 1991;213:482-491. 
Mattox KL, McCollum W, Beall ACJr, et al: Management of penetrating injuries 

of the suprarenal aorta. J Trauma 1975;15:808-815. 
Mattox KL, McCollum W, Jordan GLJr, et al: Management of upper abdomi- 
nal vascular trauma. Am Surg 1974;128:823-828. 
Mattox KL, Moore EE, Feliciano DV: Trauma. East Norwalk, Conn, Appleton 

& Lange, 1988, pp 519-536. 
Mattox KL, Pickard L, Allen MK: Emergency thoracotomy for injury. Injury 

1986;17:327-331. 
Mattox KL, Pickard L, Allen MK, Garcia-Rinaldi R: Suspecting thoracic aortic 

transection. JACEP 1978;7:12-15. 
Mattox KL, Rea J, Ennix C, et al: Penetrating injuries to the iliac arteries. Am 

J Surg 1978;136:663-667. 
Mattox KL, Von Koch L, Beall AC Jr, DeBakey ME: Logistic and technical 

considerations in the treatment of the wounded heart. Circulation 

1975;51(suppl I) and 52:210-214. 
Mattox KL, Whisennand H, Espada R, Beall ACJr: Management of acute com- 
bined injuries to the aorta and inferior vena cava. AmJ Surg 1975;130:720- 

724. 
Maughon J: An inquiry into the nature of wounds resulting in killed in action 

in Vietnam. Milit Med 1970;135:8-13. 
Mavroudis C, Roon A, Baker CC, Thomas AN: Management of acute cervi- 

cothoracic vascular injuries. J Thorac Cardiovasc Surg 1980;80:342-349. 



RE.qxd 4/16/04 3:19PM PageS04 



604 VI • THE RICH REFERENCE COLLECTION 



May A, Lipchik E, DeWeeseJA: Repair of hepatic and superior mesenteric artery 
injury: Patency demonstrated by aortography. Ann Surg 1965;162:869-872. 

McBride L, Tidik S, StothertJ, et al: Primary repair of traumatic aortic disrup- 
tion. Ann Thorac Surg 1987;43:65-67. 

McBurney R, Vaughn R: Rupture of the aorta due to nonpenetrating trauma. 
Ann Surg 1961;153:670-679. 

McBurney R, Gegan E: Blunt trauma to the aorta and major arteries. JAMA 
1962;180:330. 

McCabe C, Ferguson C, Ottinger L: Improved limb salvage in popliteal artery 
injuries. J Trauma 1983;23:982-985. 

McCann W: Successful wound repair of a stab wound of the ascending aorta. 
NYStateJMed 1958;58:3177-3178. 

McCarthy W, Yao JST, Schafer M, et al: Upper extremity arterial injury in ath- 
letes. J Vase Surg 1989;9:317-327. 

McCollum C, Mavor E: Brachial artery injury after cardiac catheterization. 
J Vase Surg 1986;4:355-359. 

McCordJ: Oxygen-derived free radicals in postischemic tissue injury. N Engl J 
Med 1985;312:159-163. 

McCorkell S, Harley J, Morishima M, Cummings D: Indications for angiogra- 
phy in extremity trauma. AJR 1985;145:1245-1247. 

McCormack L, Cauldwell E, Anson BJ: Brachial and antebrachial arterial pat- 
terns: a study of 750 extremities. Surg Gynecol Obstet 1953;96:43-54. 

McCormick T, Burch B: Routine angiographic evaluation of neck and extrem- 
ity injuries. J Trauma 1979;19:384-387. 

McCreadyR: Upper extremity vascular injuries. Surg Clin North Am 1988;68:725- 
740. 

McCready R, Logan N, Daugherty M: Long-term results with autogenous tissue 
repair of traumatic extremity vascular injuries. Ann Surg 1987;206:804- 
808. 

McCready R, Procter C, Hyde GL: Subclavian-axillary vascular trauma. J Vase 
Surg 1986;3:24-31. 

McCroskey B, Moore EE, Moore FA, Abernathy CM: A unified approach to the 
torn thoracic aorta. Am J Surg 1991;162:473-476. 

McCroskey B, Moore EE, Pearce W, et al: Traumatic arterial injuries of the 
upper extremity: Determinants of disability. Am J Surg 1988;156:553- 
555. 

McCroskey B, Moore EE, Rutherford R: Vascular Trauma. The Surgical Clinics 
of North America. Philadelphia, WB Saunders, 1988. 

McCullough J, Hollier L, Nugent M: Paraplegia after thoracic aorta occlusion: 
Influence of cerebrospinal fluid drainage. Experimental and early clinical 
results. J Vase Surg 1988;7:153-160. 

McCutchan J, Gillham N: Injury to the popliteal artery associated with dislo- 
cation of the knee: Palpable distal pulses do not negate the requirement 
for arteriography. Injury 1989;20:307-310. 

McDonald E, Goodman P, Weinstock D: Clinical indications for arteriography 
in trauma to the extremity: A review of 114 cases. Radiology 1975;166:45- 
47. 

McDonald M, Mota-Filipe H, Paul A, et al: Calpain inhibitor I reduces the acti- 
vation of nuclear factor-kappaB and organ injury/dysfunction in hemor- 
rhagic shock. FASEBJ 2001;15:171-186. 

McDonough J, Altemeier WA: Subclavian venous thrombosis secondary to 
indwelling catheters. Surg Gynecol Obstet 1971;133:397-400. 



RE.qxd 4/16/04 3:19PM PageS05 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 605 

McFadden P, Jones J, Ochsner JL: The fuzzy foreign body fragment: A subtle 

roentgenograph^ clue to mediastinal vascular injury. Am J Surg 1985; 

149:809-811. 
McFadden D, Lawelor B, Ali I: Portal vein injury. Can J Surg 1987;30:91. 
McGough E, Helfrich L, Hughes R: Traumatic in timal prolapse of the common 

carotid artery. Am J Surg 1972;123:724-75. 
Mcllduff J, Foster E, Alley RD : Traumatic aortic rupture: An additional roentgen o- 

graphic sign. Ann Thorac Surg 1977;24:77-79. 
Mclntyre RJ, Moore EE, Read R, et al: Transesophageal echocardiography in 

the evolution of a transmediastinal gunshot wound: Case report. J Trauma 

1994;36:125-127. 
McKenzie A, Sinclair A: Axillary artery occlusion complicating shoulder dislo- 
cation: A report of two cases. Ann Surg 1958;148:139-144. 
McKnightJ, Meyer J, Neville JJ: Nonpenetrating traumatic rupture of the tho- 
racic aorta. Ann Surg 1964;160:1069-1072. 
McLaughlin JS, Suherlis H, Yeager G: Sterile pericarditis from foreign body: 

Acute tamponade one month following gunshot wound. Ann Thorac Surg 

1967;3:52-56. 
McLean T, McManus R: Penetrating trauma involving the innominate artery. 

Ann Thorac Surg 1991;51:113-115. 
McMillan R, Landreneau M, McCormic G, McDonald J: Major vascular injuries 

of the torso. South Med J 1992;85:375-377. 
McNab A, Fabinyi G, Miline P: Blunt trauma to the carotid artery. Aust N Z J 

Surg 1988;58:651-656. 
McNally W: Lead poisoning caused by a bullet embedded for twenty-seven years. 

IndustMedl949;18:77. 
McNalley M, Sugg W: Traumatic communication between the aorta, right atrium, 

and left atrium. J Thorac Cardiovasc Surg 1967;54:150-152. 
McNamaraJJ, Brief DK, Beasley W, WrightJ: Vascular injury in Vietnam combat 

casualties: Results of treatment at the 24th Evacuation Hospital, ljuly 1967 

to 12 August 1969. Ann Surg 1973;178:143-147. 
McNamaraJJ, Brief DK, Stremple J, WrightJ: Management of fractures with 

associated arterial injury in combat casualties. J Trauma 1973;13:17-19. 
McNutt R, Seabrook G, Schmitt D, Aprahamian C: Blunt tibial artery trauma: 

predicting the irretrievable extremity. J Trauma 1989;29:1624. 
McQuillan W, Nolan B: Ischaemia complicating injury: A report of thirty-seven 

cases. J Bone Joint Surg 1968;50-B:482-492. 
McSwain NE Jr: Pneumatic trousers and the management of shock. J Trauma 

1977;17:719-724. 
McSwain NE Jr: PASG—Holding on for dear life. Emergency 1990. 
Meagher DJ, Defore W, Mattox KL, Harberg F: Vascular trauma in infants and 

children. J Trauma 1979;19:532-536. 
Meek A, Robbs J: Vascular injury with associated bone and joint trauma. Br J 

Surg 1984;71:341-344. 
Meissner M, Paun M, Johansen K: Duplex sonography for arterial trauma. Am 

JSurgl991;161:552. 
MelkiJ, Tabley A, BessouJP, Soyer R: Chronic false aneurysm of the sub-renal 

abdominal aorta after abdominal contusion (French) . J Maladies Vascu- 

laires 1990;15:377-379. 
Melton S, Croce MA, Patton JJ, et al: Popliteal artery trauma: Systemic antico- 
agulation and intraoperative thrombolysis improve limb salvage. Ann Surg 

1997;225:518-527. 



RE.qxd 4/16/04 3:19PM PageS06 



606 VI • THE RICH REFERENCE COLLECTION 



Menger MD, Kerger H, Geisweid A, et al: Leukocyte-endothelium interaction 
in the microvasculature of postischemic striated muscle. Adv Exp Med Biol 
1994;361:541-545. 

Menger MD, Pelikan S, Steiner D, Messmer K: Microvascular ischemia- 
reperfusion injury in striated muscle: Significance of "reflow paradox." 
Am J Physiol 1992;263:H1901-H1906. 

Menger MD, Rucker M, Vollmar B: Capillary dysfunction in striated muscle 
ischemia/reperfusion: On the mechanisms of capillary "no reflow." Shock 
1997;8:2-7. 

Menger MD, Vollmar B: In vivo analysis of microvascular reperfusion injury in 
striated muscle and skin. Microsurgery 1994;15:383-389. 

Mengoli L: Aneurysmorrhaphy of the internal carotid artery utilizing intralu- 
minal distal control. Am J Surg 1969;117:397-399. 

Menzoian J, Doyle J, Cantelmo N, et al: A comprehensive approach to extrem- 
ity vascular trauma. Arch Surg 1985;120:801-805. 

Menzoian J, Doyle J, Logerfo F, et al: Evaluation and management of vascular 
injuries of the extremities. Arch Surg 1983;118:93-95. 

Menzoian J, Logerfo F, Doyle J, et al: Management of vascular injuries to the 
leg. Am J Surg 1981;144:231-234. 

Meredith J, O'Neil E, Snow D, Hansen K: Femoral vein catheter related deep 
venous thrombosis: A perspective evaluation with venous duplex sonog- 
raphy. J Trauma 1991;31:1034. 

Merin G, Bitran D, Donchin Y, et al: Traumatic rupture of the thoracic aorta 
during pregnancy. Surgical considerations. Chest 1981;79:99-100. 

Merine D, Brody W: Role of CT in excluding major arterial injury after blunt 
thoracic trauma. Invest Radiol 1989;24:733-734. 

Merrill W, Lee R, HammonJJ, et al: Surgical treatment of acute traumatic tear 
of the thoracic aorta. Ann Surg 1988;207:699-706. 

Messina L, Brothers T, Wakefield T: Clinical characteristics and surgical man- 
agement of vascular complications in patients undergoing cardiac catheter- 
ization: Interventional versus diagnostic procedures. J Vase Surg 1991; 
13:593-600. 

Metzdroff M, Hill J, Matar A, et al: Use of sutureless intraluminal aortic pros- 
theses in traumatic rupture of the aorta. J Trauma 1986;26:691-694. 

Metzger D, Hamilton R, Stephenson D: Mesenteric arteriovenous fistula: 
Unusual cause of distal gastrointestinal bleeding. Am J Surg 1972;124: 
767. 

Meyer D, Jessen M, Grayburn P: Use of echocardiography to detect occult cardiac 
injury after penetrating thoracic trauma: A prospective study. J Trauma 
1995;39:902-909. 

Meyer JP, Lim LT, Schuler J, et al: Peripheral vascular trauma from close-range 
shotgun injuries. Arch Surg 1985;120:1126-1131. 

Meyer JP, Walsh J, Barrett J, et al: Analysis of 18 recent cases of penetrating 
injuries to common and internal carotid arteries. Am J Surg 1988;156:96- 
99. 

Meyer JP, Walsh J, Schuler J: The early fate of venous repair following civilian 
vascular trauma: A clinical hemodynamic and venographic assessment. Ann 
Surg 1987;206:458-464. 

Meyer JP, Goldfaden D, BarrettJ, et al: Subclavian and innominate artery trauma: 
A recent experience with nine patients. J Cardiovasc Surg 1988;29:283-289. 

Meyer J, Neville JJ, Hansen W: Traumatic rupture of the aorta in a child. JAMA 
1969;208:527-529. 



RE.qxd 4/16/04 3:19PM PageS07 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 607 

Meyer TJ, Slager R: False aneurysm following subtrochanteric osteotomy. J Bone 
Joint Surg 1964;46:581-582. 

Michaels A, Gerndt S, Taheri P, et al: Blunt force injury to the abdominal aorta. 
J Trauma 1996;41:105-109. 

Michaels J: Choice of material for above-knee femoropopliteal bypass graft. Br 
J Surg 1989;76:7-14. 

Michaud P, ChassignolleJ, Termet H, et al: Traumatic aneurysms of the aorta. 
Analysis of 11 observations. J Cardiovasc Surg 1971;12:121-130. 

Michelassi F, Pietrabissa A, Ferrari M, et al: Bullet emboli to the systemic and 
venous circulation. Surgery 1990;107:239-245. 

Midgley F, Behrendt DM: Surgical repair of chronic post-traumatic aneurysm 
of the aortic arch. J Thorac Cardiovasc Surg 1974;67:229-232. 

Mikulin T, Walker E: False aneurysm following blunt trauma. Injury 1984; 15:309- 
310. 

Miller D: Gangrene from arterial injuries associated with fractures and dislo- 
cations of the leg in the young and in adults with normal circulation. Am 
J Surg 1957;93:367-375. 

Miller D, Freeark R: Injuries to the popliteal artery among the young. Am J 
Surg 1962;104:633-639. 

Miller F, Bond SJ, Shumate C: Diagnostic pericardial window: A safe alterna- 
tive to exploratory thoracotomy for suspected heart injuries. Arch Surg 
1987;122:605-609. 

Miller F, Richardson JD, Thomas H, et al: Role of CT in diagnosis of major arte- 
rial injury after blunt thoracic trauma. Surgery 1988;106:596-602. 

Miller F, Seward J, Gersh B, Tajik A: Two-dimensional echocardiographic find- 
ings in cardiac trauma. Am J Cardiol 1982;50:1022-1027. 

Miller F, Shumate C, Richardson JD: Myocardial contusion: when can the diag- 
nosis be eliminated? Arch Surg 1989;124:805-808. 

Miller H, Welch C: Quantitative studies on time factor in arterial injuries. Ann 
Surg 1949;130:428-438. 

Millham F, Grindlinger G: Survival determinants in patients undergoing emer- 
gency room thoracotomy for penetrating chest injury. J Trauma 1993;34:332- 
336. 

Millikan JS, Moore EE, Cogbill TH: Inferior vena cava injuries: A continuing 
challenge. J Trauma 1983;23:207-212. 

Millikan JS, Moore EE, Van Way G, Kelly GL: Vascular trauma in the groin: Con- 
trast between iliac and femoral injuries. Am J Surg 1981;142:695-698. 

Millikan JS, Moore EE: Outcome of resuscitative thoracotomy and descending 
aortic occlusion. J Trauma 1984;24:387-392. 

Millikan JS, Moore EE: Critical factors in determining mortality from abdom- 
inal aortic trauma. Surg Gynecol Obstet 1985;160:313-316. 

Mills J, Wiedeman J, Robison JG, Hallett JJ: Minimizing mortality and morbid- 
ity from iatrogenic arterial injuries: The need for early recognition and 
prompt repair. J Vase Surg 1986;4:22-27. 

Mirvis S, Kostrubiak I, Whitley N, et al: Role of CT in excluding major arterial 
injury after blunt thoracic trauma. AJR Am J Roentgenol 1987;149:601- 
605. 

Mitchell D, Needlemon L, Bezzi M, et al: Femoral artery pseudoaneurysm: Diag- 
nosis with conventional duplex and color Doppler. Radiology 1987;165:687- 
690. 

Mitchell FI, Thai E: Results of venous interposition grafts in arterial injuries. 
J Trauma 1990;30:336-339. 



RE.qxd 4/16/04 3:19 PM Page 608 



608 VI • THE RICH REFERENCE COLLECTION 



Mitchell R, Enright L: The surgical management of acute and chronic injuries 
of the thoracic aorta. Surg Gynecol Obstet 1983;157:1-4. 

Mittal A, May I, Samson P: Traumatic aneurysm of the aortic arch. Ann Thorac 
Surg 1972;13:494-498. 

Corrected 

Monson D, Saletta J, Freeark R: Carotid vertebral trauma. J Trauma 1969;9:987- 
999. 

Montales E, Yao S, Silva Y: Management of traumatic peripheral arteriovenous 
fistulas. J Trauma 1973;13:161-165. 

Montgomery M: Effect of therapeutic venous ligation on blood flow in cases of 
arterial occlusion. Proc Soc Exp Biol Med 1929;27:178. 

Montgomery M: Therapeutic venous occlusion: Its effect on blood flow in the 
extremity in acute obstruction. Arch Surg 1932;24:1016-1027. 

Moon M, Dake M, Pelc L, et al: Intravascular stenting of acute experimental 
type B dissections. J Surg Res 1993;54:381-388. 

Moore C, Cohen A: Combined arterial, venous and ureteral injury complicat- 
ing lumbar disc surgery. Am J Surg 1968;115:574-577. 

Moore C, Wolma F, Brown R, Derrick J: Vascular trauma: A review of 250 cases. 
Am J Surg 1971;122:576-578. 

Moore EE, Burch JM, Franciose RJ, et al: Staged physiologic restoration and 
damage control surgery. World J Surg 1998;22:1184. 

Moore EE, Cogbill TH, Jurkovich GJ: Organ injury scaling III: Chest wall, abdom- 
inal vascular, ureter, bladder and urethra. J Trauma 1992;33:337-339. 

Moore EE, Cogbill TH, Malangoni M, et al: Organ injury scaling. Surg Clin 
North Am 1995;75:293-303. 

Moore EE, Malangoni M, Cogbill TH: Organ Injury Scaling IV: Thoracic, vas- 
cular, lung, cardiac, and diaphragm. J Trauma 1994;36:229-300. 

Moore E, Webb W, Verrier E, et al: MRI of chronic posttraumatic aneurysms of 
the thoracic aorta. AJR Am J Roentgenol 1984;143:1195-1196. 

Moore HJ, Nyhus L, Kanar E, Harkins H: Gunshot wounds of the major arter- 
ies: An experimental study with clinical implications. Surg Gynecol Obstet 
1954;98:129-147. 

Moore T: Acute arterial obstruction due to the traumatic circumferential intimal 
fracture. Ann Surg 1958;148:111-114. 

Moore T, Peters M: Thru-and-thru gunshot penetration of the abdominal aorta 
in a 4-year-old child managed by aortic transection, debridement and reanas- 
tomosis with survival. J Trauma 1979;19:537-539. 

Moore W: Vascular Surgery. Orlando, Grune & Stratton, 1986. 

Morano J, Burkhalter J, Daniel CJr: Bilateral popliteal arteriovenous fistulas. 
J Trauma 1987;27:577-578. 

Moreno C, Moore EE, Majune J: Pericardial tamponade. A critical determinant 
for survival following penetrating cardiac wounds. J Trauma 1986;26: 
821. 

Moreno C, Moore EE, Rosenberger A, Cleveland H: Hemorrhage associated 
with major pelvic fracture: Amultispecialty challenge. J Trauma 1986;26:987- 
994. 

Morgan P, Goodman L, Aprahamian C, et al: Evaluation of traumatic aortic 
injury: Does dynamic contrast-enhanced CT ply a role? Radiology 
1992;182:661-666. 

Morin J, Provan J, Jewett M, Ameli FM: Vascular injury and repair associated 
with retroperitoneal lymphadenectomy for nonseminomatous germinal 
cell tumours of the testis. Can J Surg 1992;35:253-256. 



RE.qxd 4/16/04 3:19PM PageS09 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 609 

Morin R, Wright CB, Dunn E, et al: Vascular trauma. In James E, Corry R, Perry 
JFJr (eds): Basic Surgical practice. St. Louis, Hanley & Belfus, 1987, pp 
521-523. 

Morris GC Jr, Beall AC Jr, Berry WB, et al: Anatomical studies of the distal 
popliteal artery and its branches. Surg Forum 1960;10:498-502. 

Morris GC Jr, Beall AC Jr, Roof WR, DeBakey ME: Surgical experience with 220 
acute arterial injuries in civilian practice. Am J Surg 1960;99:775-781. 

Morris GCJr, Creech O Jr, DeBakey ME: Acute arterial injuries in civilian prac- 
tice. Am J Surg 1957;93:565-572. 

Morton J, Southgate W, DeWeeseJA: Arterial injuries of the extremities. Surg 
Gynecol Obstet 1966;123:611-627. 

Morton JR, Crawford ES: Bilateral traumatic renal artery thrombosis. Ann Surg 
1972;176:62-67. 

Morton JR, Reul GJ, Arbegast NR, et al: Bullet embolus to the right ventricle: 
Report of three cases. Am J Surg 1971;122:584-590. 

Moss A, Bruhn F: The echocardiogram. An ultrasound technic for the detec- 
tion of pericardial effusion. N Engl J Med 1996;274:380. 

Moss C, Veith F, Jason R, Rudavsky A: Screening isotope angiography in arter- 
ial trauma. Surgery 1969;86:881-889. 

Motsay G, Manlove C, PerryJF: Major venous injury with pelvic fracture.J Trauma 
1969;9:343-346. 

Mott V: Reflections on securing in a ligature the arteria innominata. Med Surg 
1918;1:9. 

Moure P: Les Greffes Vasculaires. Paris, Doin, 1914. 

Moya J, de Pablo C, Sanchez M, et al: Diagnosis by Doppler echocardiography. 
Chest 1990;98:1016-1017. 

Mozingo J, Denton I: The neurologic deficit associated with sudden occlusion 
of the abdominal aorta due to blunt trauma. Surgery 1975;77:118-125. 

Mubarak S, Owen C: Double-incision fasciotomy of the leg for decompression 
in compartment syndromes. J Bone Joint Surg 1977;59A:184. 

Mucha PJ, Olivier HJ, Pairolero P, Farnell M: Blunt thoracic aortic injury: A 
reappraisal. J Trauma 1983;23:652. 

Mucha PJ, Schaff H, Pairolero P: Clamp/repair of traumatic transection of 
descending aorta. Ann Thorac Surg 1987;43:351-352. 

Mufti M, LaGuerre J, Pochaczevsky R, et al: Diagnostic value of hematoma in 
penetrating arterial wounds of the extremities. Arch Surg 1970;101:562- 
569. 

Mulder D, GrollmanJJ: Traumatic disruption of the thoracic aorta. Am J Surg 
1969;118:311-316. 

Muller WJ, Goodwin W: Renal arteriovenous fistula following nephrectomy. 
Ann Surg 1956;144:240-244. 

Mullins R, Lucas C, Ledgerwood A: The natural history following venous liga- 
tion for civilian injuries. J Trauma 1980;20:737-743. 

Munda R, Bole PV, Lande A, Clauss RH: The use of an external Dacron shunt 
in repair of a supraceliac abdominal aortic traumatic aneurysm. J Cardio- 
vasc Surg 1977;18:141-146. 

Murdock C: Traumatic rupture of the thoracic aorta. Arch Surg 1957;74:589- 
592. 

MurphyJ: Resection of arteries and veins injured in continuity end-to-end suture. 
Exp Clin Res Med Rec 1897;51:73-104. 

MurphyJ: Myositis; ischemic myositis; infiltration myositis; cicatricial muscular 
or tendon fixation in forearm; internal, external and combined compression 



RE.qxd 4/16/04 3:19 PM Page 610 



610 VI • THE RICH REFERENCE COLLECTION 



myositis, with subsequent musculotendinous shortening. JAMA 1914; 

63:1249-1255. 
Murphy T, Dorfman G, Segall M: Iatrogenic arterial dissection: Treatment by 

percutaneous transluminal angioplasty. Cardiovasc Intervent Radiol 

1991;14:302. 
Murphy T, Piper C, Anderson CL: Complications of left heart catheterization. 

Am Surg 1971;37:472-475. 
Murray D: Posttraumatic thrombosis of the internal carotid and vertebral 

arteries after non-penetrating injuries of the neck. Br J Surg 1957;44: 

556. 
Murray E, Minami K, Kortke H, et al: Traumatic sinus of Valsalva fistula and 

aortic valve rupture. Ann Thorac Surg 1993;55:760-761. 
Murray G: Heparin in thrombosis and embolism. Br J Surg 1940;27:567- 

598. 
Murray G: Surgical repair of injuries to main arteries. Am J Surg 1952;83:480. 
Murray G, Brawley RK, Gott V: Reconstruction of the innominate artery by 

means of a temporary heparin-coated shunt bypass. J Thorac Cardiovasc 

Surg 1971;62:34-41. 
Murray G, Janes J: Prevention of acute failure of circulation following injuries 

to large arteries: Experiments with glass cannulae kept patent by admin- 
istration of heparin. Br Med J 1940;2:6-7. 
Mustard W, Bull C: A reliable method for relief of traumatic vascular spasm. 

Ann Surg 1962;155:339-344. 
Myers S, Reed M, Black CT, et al: Noniatrogenic pediatric vascular trauma. J 

Vase Surg 1989;10:258-265. 
Myers S, Harward T, Cagle L: Isolated subclavian artery dissection after blunt 

trauma. Surgery 1991;109:336. 
Myers SI, Harward TRS, Maher DP, et al: Complex upper extremity vascular 

trauma in an urban population. J Vase Surg 1990;12:305-309. 
Myers W, Lawton B, Sautter R: An operation for tracheal-innominate artery 

fistula. Arch Surg 1972;105:269-274. 
Myles R, Yellin AE: Traumatic injuries of the abdominal aorta. Am J Surg 

1979;138:273-277. 
Nabatoff R, Blum L, Touroff A: Long-term sequelae of common femoral vein 

ligation in the treatment of thromboembolic disease. Surgery 1970;67:272- 

276. 
Nachbur B, Meyer R, Verkkala K, Zurcher R: The mechanisms of severe arter- 
ial injury in surgery of the hip joint. Clin Orthop 1979;141:122-133. 
Naclerio E: Penetrating wounds of the heart: Experience with 249 patients. Dis 

Chest 1964;46:1-22. 
Nakano J, DeSchryver C: Effects of arteriovenous fistula on systemic and pul- 
monary circulations. Am J Physiol 1964;207:1319. 
Napoli P, Meade P, Adams CW: Primary aortoenteric fistula from a post trau- 
matic pseudoaneurysm. J Trauma 1996;41:149-152. 
Nashef S, Talwalkar N, Janieson M: Aortic arch rupture in a child following 

minor blunt trauma. J Thorac Cardiovasc Surg 1987;35:240-241. 
NataliJ, Lacombe M, Bruchou P, Vinardi G: Les traumatismes arteriels vus tar- 

divement conduite a tenir en leur presence. Presse Med 1964;72:2273. 
NataliJ, Maraval M, Kieffer E, Petrovic P: Fractures of a clavicle and injuries of 

the subclavian artery: Report of 10 cases. J Cardiovasc Surg 1975;16:541. 
Natali J, Benhemori AC: Iatrogenic vascular injuries. J Cardiovasc Surg 

1979;20:169-176. 



RE.qxd 4/16/04 3:19 PM Page 611 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 61 1 

Naude G, Back M, Perry MO, Bongard FS: Blunt disruption of the abdominal 

aorta: Report of a case and review of the literature. J Vase Surg 1997;25:931- 

935. 
Navarre J, Cardillo P: Vascular trauma in children and adolescents. Am J Surg 

1982;143:229-231. 
Neagu S: Abdomino-thoracic shooting wound with multivisceral involvement 

including injury of abdominal aorta in the supra-mesocolic segment. Acta 

Chir Belg 1993;93:107-109. 
NeelS: Army aeromedical evacuation procedures in Vietnam.JAMA 1968;204:99- 

103. 
Neely W, Hardy JD, Artz CP: Arterial injuries in civilian practice: A current reap- 
praisal with analysis of 43 cases. J Trauma 1961;1:424-439. 
Neville R, Franco C, Anderson RJ, et al: Popliteal artery agenesis: a new anatomic 

variant. J Vase Surg 1990;12:573-576. 
Neville R, Hobson R, Wantanabe B: A prospective evaluation of arterial intimal 

injuries in an experimental model. J Trauma 1991;31:669-675. 
Neville R, Yasuhara H, Watanabe B: Endovascular management of arterial intimal 

defects: An experimental comparison by arteriography, angioscopy, and 

intravascular ultrasonography. J Vase Surg 1991;13:496-502. 
Nghiem D, Boland JP: Four-compartment fasciotomy of the lower extremity 

without fibulectomy: A new approach. Am Surg 1980;46:414-417. 
Ngu V, Konstam PG: Traumatic dissecting aneurysm of the abdominal aorta. 

Br J Surg 1965;52:981-982. 
Nguyen L, LewinJ: Angiographic demonstration of fistula between abdominal 

aorta and thoracic duct. JAMA 1970;211:499-500. 
Nicholas G, Lane F: Traumatic superior mesenteric artery-superior mesenteric 

vein fistula. J Trauma 1974;14:344. 
Nicholas G, DeMuth WE Jr: Long-term results of brachial thrombectomy fol- 
lowing cardiac catheterization. Ann Surg 1976;183:436-438. 
Nichols J, Svoboda J, Parks S: Use of temporary intravascular shunts in selected 

peripheral arterial injuries. J Trauma 1986;26:1094-1096. 
Nichols J, Lillehei K: Nerve injury associated with acute vascular trauma. Surg 

Clin North Am 1988;68:837-852. 
Nicholson J, Foster R, Heath R: Bryant's traction: A provocative cause of cir- 
culatory complication. JAMA 1955;157:415-418. 
Nicoladoni C: Phlebarteriectasie der rechten oberen extremitat. Arch Klin Chir 

1875;18:252. 
Nizzero A, Miles J: Blunt trauma of the abdominal aorta. Can Med Assoc J 

1986;135:219-220. 
Nolan B, McQuillan N: A study of acute traumatic limb ischaemia. Br J Surg 

1965;52:559-565. 
Nolan B: Vascular injuries. J Royal Coll Surg 1968;13:72. 
Nolte D, Lehr HA, Messmer K: Adenosine inhibits postischemic leukocyte- 

endothelium interaction in postcapillary venules of the hamster. Am J 

Physiol 1991;261:H651-H655. 
Nolte D, Hecht R, Schmid P, et al: Role of Mac-1 and ICAM-1 in ischemia- 

reperfusion injury in a microcirculation model of BALB/C mice. Am J 

Physiol 1994;267:H295-H930. 
Noon G, Boulafendis D, Beall AC Jr: Rupture of the heart secondary to blunt 

trauma.J Trauma 1971;11:122-128. 
Nordestgaard A, Bodily KC, Osborne RJ, ButtorffJ: Major vascular injuries during 

laparoscopic procedures. Am J Surg 1995;169:543-545. 



RE.qxd 4/16/04 3:19 PM Page 612 



612 VI • THE RICH REFERENCE COLLECTION 



Norotsky M, Rogers F, Shackford S: Delayed presentation of splenic artery 
pseudoaneurysms following blunt abdominal trauma: Case reports. J Trauma 
1995;38:444-447. 

Norris C, Zlotnick R, Silva W, Wheeler H: Traumatic pseudoaneurysm follow- 
ing blunt trauma. J Trauma 1986;28:480-482. 

Norris G: Varicose aneurysm at the bend of the arm: Ligature of the artery 
above and below the sac; Secondary hemorrhages with a return of the 
aneurysm thrill on the tenth day; Cure. Am J Med Sci 1843;5:17. 

North C, Ahmadi J, Segall H, Zee C: Penetrating vascular injuries of the face 
and neck: Clinical and angiographic correlation. AJR Am J Roentgenol 
1986;147:995-999. 

Norton L, Spencer FC: Long-term comparison of vein patch with direct suture. 
Arch Surg 1964;89:1083. 

Noth P: Electrocardiographic patterns in penetrating wounds of the heart. Am 
HeartJ 1946;32:713. 

NunleyD: Radiographic diagnosis of aortic injury. Ann Thorac Surg 1984;38:424- 
425. 

NunleyJ, Roman L, UrbaniakJ: Arterial shunting as an adjunct to major limb 
revascularization. Ann Surg 1981;193:271-273. 

Nunn D: Abdominal aortic dissection following non-penetrating abdominal 
trauma. Am Surg 1973;39:117-179. 

Nypaver T, SchulerJ, McDonnell P: Long-term results of venous reconstruc- 
tion after trauma in civilian practice. J Vase Surg 1992;16:762-768. 

O'DonnellV, AtikM, Pick Revaluation and management of penetrating wounds 
of the neck. Am J Surg 1979;138:309-314. 

O'Donnell TJ, Brewster DC, Darling RC, et al: Arterial injuries associated with 
fractures and/or dislocations of the knee. J Trauma 1977;17:775-784. 

O'Gorman R, Feliciano DV: Arteriography performed in the emergency center. 
Am J Surg 1986;152:323-325. 

O'Gorman R, Feliciano DV, Bitondo CG, et al: Emergency center arteriogra- 
phy in the evaluation of suspected peripheral vascular injuries. Arch Surg 
1984;119:568-573. 

O'Neill JJ: Traumatic vascular lesions in infants and children. In Dean RH, 
O'Neill ]J (eds): Vascular Disorders of Childhood. Philadelphia, Lea & 
Febiger, 1983, pp 181-193. 

O'Neill JJ, Killen DA: Autogenous vein graft for repair of acute tibial artery 
injury. Ann Surg 1965;162:218-220. 

O'Neill MJ, MyersJ, Brown G, et al: Avulsion of the innominate artery from the 
aortic arch associated with a posterior tracheal tear. J Trauma 1982;22:56- 
59. 

O'Reilly M, Hood J, Livingston RH, IrwinJ: Penetrating injuries of the popliteal 
vein: A report on 34 cases. Br J Surg 1980;67:337-340. 

O'Sullivan M, Folkerth T, Morgan J, Fosburg R: Posttraumatic thoracic aortic 
aneurysm: Recognition and treatment. Arch Surg 1972;105:14-18. 

Ochsner JL, Crawford ES, DeBakey ME: Injuries of the vena cava caused by 
external trauma. Surgery 1961;49:397-405. 

Ochsner M, Harviel J, Stafford P, et al: Development and organization for casu- 
alty management on a 1 ,000-bed hospital ship in the Persian Gulf. J Trauma 
1992;32:501-513. 

Ochsner M, Hoffman A, DiPasquale D, et al: Associated aortic rupture-pelvic 
fracture: An alert for orthopaedic and general surgeons. J Trauma 1992;33: 
429-434. 



RE.qxd 4/16/04 3:19 PM Page 613 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 613 

Ochsner MJ, Champion HR, Chambers R, Harviel J: Pelvic fracture as an indi- 
cator of increased risk of thoracic rupture. J Trauma 1980;29:1376-1379. 

Odekwu P, Oiler D: Hematemesis as initial presentation of traumatic paraceliac 
pseudoaneurysm of the abdominal aorta. Cardiovasc Surg 1994;2:781-782. 

Odom C: Causes of amputations of battle injuries with emphasis on vascular 
injuries. Surgery 1946;19:562. 

Ogilvie W: War surgery in Africa. Br J Surg 1944;31:313. 

Ohkawa M, Clemens M, Chaudry IH: Studies on the mechanism of beneficial 
effects of ATP-MgC12 following hepatic ischemia. Am J Physiol 
1983;244:R695-R702. 

Ohkawa M, Chaudry IH, Clemens M, Baue AE: ATP-MgCL produces sustained 
improvementin hepatic mitochondrial function and blood flow after hepatic 
ischemia. J Surg Res 1984;37:226-234. 

Okubo S, Bernardo NL, Elliot G, et al: Tyrosine kinase signaling in action poten- 
tial shortening and expression of HSP72 in late preconditioning. Physiol 
Heart Circ Physiol 2000;279:H2269-H2276. 

Olinde A: Letters to the editors: Traumatic subclavian-axillary aneurysm. J Vase 
Surg 1990;11:848-849. 

Oiler D, Rutledge R, Thomason M, et al: Vascular injuries in a rural state: A 
review of 940 patients from state trauma registry. J Trauma 1991;31:1036. 

Oiler D, Rutledge R, Clancey T, et al: Vascular injuries in a rural state: A review 
of 978 patients from a state trauma registry. J Trauma 1992;32:740-746. 

Orcutt M, Levine B, Gaskill H III, Sirinek K: Iatrogenic vascular injury: A reducible 
problem. Arch Surg 1985;120:384-385. 

Orcutt M, Levine B, Gaskill H III, Sirinek K: Civilian vascular trauma of the 
upper extremity. J Trauma 1986;26:63-67. 

Orcutt M, Levine B, Root H, Sirinek K: The continuing challenge of popliteal 
vascular injuries. Am J Surg 1983;146:758-761. 

Ordog G, Albin D, Wasserberger J, Balasubramanium S: Shotgun "birdshot" 
wounds to the neck. J Trauma 1988;28:491-497. 

Orringer M, Kirsh MM: Primary repair of acute traumatic aortic disruption. 
Ann Thorac Surg 1983;35:672-675. 

Ortner A, Berg HF, Ledbendiger A: Limb salvage through small-vessel surgery. 
Arch Surg 1961;83:414-421. 

Osier T, Baker SP, Long W: A modification of the injury severity score that both 
improves accuracy and simplifies scoring. J Trauma 1997;43:922-926. 

Osier W: Case of arteriovenous aneurysm of the axillary artery and vein of 14 
years duration. Ann Surg 1893;17:37-40. 

Osier W: Report of a case of arteriovenous aneurysm of the thigh.Johns Hopkins 
HospBulll905;16:119. 

Osier W: An arteriovenous aneurysm of the axillary vessels of 30 years dura- 
tion. Lancet 1913;2:1248. 

Osier W: Remarks on arteriovenous aneurysm. Lancet 1915;1:949. 

Ottolenghi C: Vascular complications in injuries about the kneejoint. Clin Orthop 
1982;165:148-156. 

Ouchi H, Ohara I, Kijima M: Intraluminal protrusion of completely disrupted 
intima: An unusual form of acute arterial injury. Surgery 1965;57:220-224. 

Overbeck W, Gruenagel N, Krauss H: Eisensplitterverletzung der intraprikar- 
dialen aorta. Thoraxchirurgie 1968;16:274. 

Oweida S, Roubin G, Smith RI, Salam A: Postcatheterization vascular compli- 
cations associated with percutaneous transluminal coronary angioplasty. 
J Vase Surg 1990;12:310-315. 



RE.qxd 4/16/04 3:19 PM Page 614 



614 VI • THE RICH REFERENCE COLLECTION 



Owen D, Hodgson P: Control of hemorrhage following missile wound to the 
pelvis. J Trauma 1980;20:906-908. 

OwensJ: The management of arterial trauma. Surg Clin North Am 1963;43:371- 
385. 

Owens T, Watson W, Prough D, et al: Limiting initial resuscitation of uncon- 
trolled hemorrhage reduces internal bleeding and subsequent volume 
requirements. J Trauma 1995;39:200-207. 

Pachter H, Spencer F, Hofsetter S: Experience with the finger fracture tech- 
nique to achieve intra-hepatic hemostasis in 75 patients with severe injuries 
of the liver. Ann Surg 1978;197:771-777. 

Pachter H, Spencer FC, Hofsetter S, et al: The management of juxtahepatic 
venous injuries without an atriocaval shunt: Preliminary clinical observa- 
tions. Surgery 1986;99:569-575. 

Padar S: Air gun pellet embolizing the intracranial internal carotid artery: Case 
report. J Neurosurg 1975;43:222-224. 

Padberg FT, RubelowskyJ, Hernandez-Maldonado J, et al: Infrapopliteal arte- 
rial injury: Prompt revascularization affords optimal limb salvage. J Vase 
Surg 1992;16:877-886. 

Page C, Hagood C, Kremmerer W: Management of catheterization brachial 
artery thrombosis. Surgery 1982;72:619-623. 

Paget S: The Surgery of the Chest. London, John Wright and Company, 1896. 

Pairolero P, Walls J, Payne W, et al: Subclavian-axillary artery aneurysms. Surgery 
1981;90:757-763. 

Palma E, Esperon R: Vein transplants and graft in the surgical treatment of the 
post-phlebitic syndrome. J Cardiovasc Surg 1960;1:94-107. 

Pan M, Medina A, Suarez de Lezo J: Obliteration of femoral pseudoaneurysm 
complicating coronary intervention by direct puncture and permanent or 
removable coil insertion. Am J Cardiol 1997;80:786-788. 

Panes J, Kurose I, Rodriguez-Vaca D, et al: Diabetes exacerbates inflammatory 
responses to ischemia-reperfusion. Circulation 1996;93:161-167. 

Panetta T, Sclafani S, Goldstein A, et al: Percutaneous transcatheter emboliza- 
tion for massive bleeding from pelvic fractures. J Trauma 1985;25:1021- 
1029. 

Panetta T, Bottiurai VS, Batson RC: Nonreversed translocated saphenous vein 
bypass for arterial trauma. J Trauma 1988;28:1065-1070. 

Panetta T, Hunt J, Buechter K: A blind, randomized canine study of duplex 
scanning and arteriography for the diagnosis of arterial injury. J Trauma 
9999. 

Panetta TF, Hunt JP, Buechter KJ, et al: Duplex ultrasonography versus arteri- 
ography in the diagnosis of arterial injury: An experimental study.J Trauma 
1992;33:627-636. 

Panetta T, Hunt J, Buechter K, et al: Duplex ultrasonography versus arteriog- 
raphy in the diagnosis of arterial injury: An experimental study.J Trauma 
1992;33:627-636. 

Pappas P, Haser P, Teehan E, et al: Outcome of Complex Venous Reconstruc- 
tions in Trauma Patients: Proceedings of the Eight American Venous Forum, 
1996 February 23-25: San Diego. San Diego, American Venous Forum, 
1996. 

Parkinson D, West M: Traumatic intracranial aneurysms.J Neurosurg 1980;52:11- 
20. 

Parks T: Surgical management of gunshot injures of the large intestine. J R Soc 
Med 1979;72:412-416. 



RE.qxd 4/16/04 3:19 PM Page 615 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 615 

Parmley L, Mattingly T, Manion W, Jahnke EJ Jr: Non-penetrating traumatic 
injury of the aorta. Circulation 1958;17:1086-1101. 

Parmley L, Mattingly T, Manion W: Penetrating wounds of the heart and aorta. 
Circulation 1958;17:953-973. 

Parmley LJ, Orbison J, Hughes CW, Mattingly T: Acquired arteriovenous fistu- 
las complicated by endarteritis and endocarditis lenta due to streptococ- 
cus faecalis. N Engl J Med 1954;250:305-309. 

ParodiJ: Endovascular repair of abdominal aortic aneurysms and other arter- 
ial lesions. J Vase Surg 1995;21:556-557. 

ParodiJ: Endovascular repair of aortic aneurysms, arteriovenous fistulas, and 
false aneurysms. World J Surg 1996;20:655-663. 

Partap V, Cassoff J: Ultrasound-guided percutaneous thrombin injection for 
treatment of femoral pseudoaneurysms: Technical note. Can Assoc Radiol 
J 1999;50:182-184. 

Pasch AR, Bishara RA, Lim LT, et al: Optimal limb salvage in penetrating civil- 
ian vascular trauma. J Vase Surg 1986;3:189-195. 

Pasch AR, Rashad A, Bishara RA, et al: Results of venous reconstruction after 
civilian vascular trauma. Arch Surg 1986;121:607-611. 

Passaro EJ, Pace W: Traumatic rupture of the aorta. Surgery 1959;46:787-791. 

Pate J: Traumatic rupture of the aorta: Emergency operation. Ann Thorac Surg 
1985;39:531-537. 

Pate J, Butterick O, Richardson R: Traumatic rupture of the thoracic aorta. 
JAMA 1968;203:1022-1024. 

Pate J, Cole F, Walker W, Fabian TC: Penetrating injuries of the aorta arch and 
its branches. Ann Thorac Surg 1993;55:586-592. 

Pate J, Fabian TC, Walker W: Acute traumatic rupture of the aortic isthmus: 
Repair with cardiopulmonary bypass. Ann Thorac Surg 1995;59:90-99. 

Pate J, Fabian TC, Walker W: Traumatic rupture of the aortic isthmus: An emer- 
gency? World J Surg 1995;19:119-125. 

PateJ, Richardson RJ: Penetratingwounds of cardiac valves.JAMA 1969;207:309- 
311. 

PateJ, Sherman R, Jackson T, Wilson H: Cardiac failure following traumatic 
arteriovenous fistula: A report of 14 cases. J Trauma 1965;5:398-403. 

PateJ, Wilson H: Arterial injuries of the base of the neck. Arch Surg 1964;89:1 106- 
1110. 

Patel A, Marin M, Veith F, et al: Endovascular graft repair of penetrating sub- 
clavian artery injuries. J Endovasc Surg 1996;3:383-388. 

Patel K, Kulkarni S, Babu S, Clauss RH: Abdominal vascular trauma. Dis Chest 
1987;30:13-23. 

Patel K, Cortes LE, Semel L, et al: Bullet embolism.J Cardiovasc Surg 1989;30:584- 
590. 

Patel N, Mann F, Jurkovich GJ: Penetrating ulcer of the descending aorta 
mimicking a traumatic aortic laceration. AJR Am J Roentgenol 1996;166: 
20. 

Pathria M, Zlatkin M, Sartoris D, et al: Ultrasonography of the popliteal fossa 
and lower extremities. Radiol Clin North Am 1988;26:77-85. 

Patman R, Poulos E, Shires G: The management of civilian arterial injuries. 
Surg Gynecol Obstet 1964;118:725-738. 

Patman R, Thompson J: Fasciotomy in peripheral vascular surgery: Report of 
164 patients. Arch Surg 1970;101:633-672. 

Patman R: Indications and technique. In Rutherford R (ed): Vascular Surgery. 
Philadelphia, WB Saunders, 1984. 



RE.qxd 4/16/04 3:19PM PageS16 



616 VI • THE RICH REFERENCE COLLECTION 



Paton B, Elliot D, Taubman J, Owens J: Acute treatment of traumatic aortic 

rupture. J Trauma 1971;11:1-14. 
Patt A, McCroskey B, Moore EE: Hypothermia-induced coagulopathies in trauma. 

Surg Clin North Am 1988;64:775-785. 
Patterson F, Morton K: The cause of death on fractures of the pelvis: With a 

note on treatment by ligation of the hypogastric (internal iliac) artery. 

J Trauma 1973;13:849-856. 
Patterson R, Burns W, Jannotta F: Rupture of the thoracic aorta: Complication 

of resuscitation. JAMA 1973;226:197. 
Patton A, Guyton S, Lawson D: Treatment of severe arterial injuries. Am J Surg 

1981;141:465-471. 
Payne S, Snell M: Traumatic renal artery dissection. Urology 1988;16:335-337. 
Peacock J, Proctor HJ: Factors limiting extremity function following vascular 

injury. J Trauma 1977;17:532-534. 
Pearlman A: Transesophageal echocardiography: Sound diagnostic technique 

or two edged sword? N Engl J Med 1991;324:841-843. 
Peck C: The operative treatment of heart wounds. Ann Surg 1909;50:100-134. 
Peck J, Eastmen A, Bergan JJ, Sedwitz M: Popliteal vascular trauma: A commu- 
nity experience. Arch Surg 1990;120:1339-1344. 
Pecunia R, Raves J: A technique for evaluation of the injured extremity with a 

single film exclusion arteriography. Surg Gynecol Obstet 1990;1 70:448. 
Peiper H: Significance of traumatology in abdominal and vascular surgery. Jpn 

J Surg 1985;15:93-102. 
Pemberton J, Seefeld P, Barker NW: Traumatic arteriovenous fistula involving 

the abdominal aorta and the inferior vena cava. Ann Surg 1946;123:580- 

586. 
Penn I: The vascular complications of fractures of the clavicle. J Trauma 

1964;4:819-831. 
Pennington D, Dranpanas T: Acute post-traumatic coarctation of the abdomi- 
nal aorta. Surgery 1975;78:538-542. 
Pepe PE, Bass RR, Mattox KL: Clinical trail of the pneumatic antishock garment 

in the urban prehospital setting. Ann Emerg Med 1986;15:1407-1410. 
Perchinsky M, LongWB, HillJ, et al: Extracorporeal cardiopulmonary life support 

with heparin-bonded circuitry in the resuscitation of massively injured 

trauma patients. Am J Surg 1995;169:488-491. 
Perdue GJ, Smith RI: Intra-abdominal vascular injury. Surgery 1968;64:562- 

568. 
Perkins R, Elchos T: Stab wound of the aortic arch. Ann Surg 1958;147:83-86. 
Perler B, McCabe C, Abbott WM, Buckley MJ: Vascular complications of intra- 

aortic balloon counterpulsation. Arch Surg 1983;118:957-9562. 
Perry MO: The Management of Acute Vascular Injuries. Baltimore, Williams & 

Wilkins, 1981. 
Perry M: Metabolic consequences of vascular trauma. Contemp Surg 1981 ;1 8:39- 

46. 
Perry MO: Iatrogenic injuries of arteries in infants. Surg Gynecol Obstet 

1983;157:415-418. 
Perry MO: Compartment syndromes and reperfusion injury. Surg Clin North 

Am 1988;68:853-864. 
Perry MO, Synder W, Thai E: Carotid artery injuries caused by blunt trauma. 

Ann Surg 1980;192:74-77. 
Perry MO, Thai E, Shires G: Management of arterial injuries. Ann Surg 

1971;173:403-408. 



RE.qxd 4/16/04 3:19 PM Page 617 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 617 

Peterson A, Williams D, Rodriguez J, Francis I: Percutaneous treatment of a 

traumatic aortic dissection by balloon fenestration and stent placement. 

AJR Am J Roentgenol 1996;164:1274-1276. 
Peterson H, Greenspan J, Ory H: Death following puncture of the aorta during 

laparoscopic sterilization. Obstet Gynecol 1982;59:133-134. 
Peterson N: Traumatic bilateral renal infraction. J Trauma 1989;29:158-161. 
Petit-Dutaillis D, Janet H, Thiebaut F, Guillaumat L: Effets d'une inversion cir- 

culatoire par anastomose carotido-jugulaire sure une hemiplegie droit avec 

aphasie due a una thrombose de la carotide interne d'Origine inconnue 

chez tin adolescent de 13 ans. Rev Neurol 1949;81:75. 
Petrovsky B, Milinov O: "Arterialization" and "venization" of vessels involved in 

traumatic arteriovenous fistulae: Aetiology and pathogenesis (an experi- 
mental study). J Cardiovasc Surg 1967;8:396-407. 
Petty S, Parker L, Mauro M, et al: Chronic posttraumatic aortic pseudoaneurysm: 

Recognition before rupture. Postgrad Med 1991;89:177-178. 
Pezzella A, Todd E, Dillon M, et al: Early diagnosis and individualized treat- 
ment of blunt thoracic aortic trauma. Am Surg 1978;44:699-703. 
Pezzella A, Griffen W, Ernst CB: Superior mesenteric artery injury following 

blunt abdominal trauma: Case report with successful primary repair. 

J Trauma 1978;18:472-474. 
Phifer T, Gerlock A, Vekovius W, et al: Amputation risk factors in concomitant 

superficial femoral artery and vein injuries. Ann Surg 1984;199:241- 

243. 
Phifer T, Gerlock A, Rich NM, McDonaldJ: Long-term patency of venous repairs 

demonstrated by venography. J Trauma 1985;25:342-346. 
Phillips C, Jacobsen D, Brayton DF, Bloch JH: Central vessel trauma. Am Surg 

1979;45:517-530. 
Phillips E, Rogers W, Caspar MR: First rib fractures: Incidence of vascular injury 

and indications for angiography. Surgery 1981;89:42-47. 
Pick T: On partial rupture of arteries from external violence. St George's Hosp 

Rep (Lond) 1873;6:161. 
Pick T: A clinical lecture of a case of arteriovenous aneurysm. Lond Med Times 

Gaz 1883;2:677. 
Pickard L, Mattox KL, Espada R, et al: Transection of the descending thoracic 

aorta secondary to blunt trauma. J Trauma 1977;17:749-754. 
Pickelmann S, Nolte D, Leiderer R, et al: Attenuation of postischemic reper- 

fusion injury in striated skin muscle by diaspirin-cross-linked Hb. Am J 

Physiol 1998;275:H361-H368. 
Pietri P, Alagni G, Settembrini P: Iatrogenic vascular lesions. Int Surg 

1981;66:213-216. 
Pikoulis E, Rhee P, Nishibe T, et al: Arterial repair with synthetic patch by using 

titanium clips. J Trauma 2000;48:292-295. 
Pisters P, Heslin M, Riles T: Abdominal aortic pseudoaneurysm after blunt trauma. 

J Vase Surg 1993;18:307-309. 
Pitner S: Carotid thrombosis due to intraoral trauma: An unusual complica- 
tion of a common childhood accident. N Engl J Med 1966;274:764- 

767. 
Piwnica A, Chetochine F, Soyer R, Winckler C: Traumatic rupture of the aortic 

arch with disinsertion of the innominate artery. J Thorac Cardiovasc Surg 

1971;61:246-252. 
Plume S, DeWeese JA: Traumatic rupture of the thoracic aorta. Arch Surg 

1979;114:240-243. 



RE.qxd 4/16/04 3:19PM PageS18 



618 VI • THE RICH REFERENCE COLLECTION 



Plummer D, Bunette D, Asinger R, Ruiz E: Emergency department echocar- 
diography improves outcome in penetrating cardiac injury. Ann Emerg 

Med 1992;21:709-712. 
Pochaczevsky R, Mufti M, LaGuerre J: Arteriography of penetrating wound of 

the extremities: Help or hindrance? J Canad Assoc Radiol 1973;24:354- 

361. 
Pomerantz M, Hutchison D: Traumaticwounds of the heart.J Trauma 1969;9:135- 

139. 
Pontius G, Kilbourne B, Paul E: Non-penetrating abdominal trauma. Arch Surg 

1957;72:800. 
Pool E: Treatment of heart wounds. Ann Surg 1912;55:485-512. 
Poole G: Fracture of the upper ribs and injury to the greatvessels. Surg Gynecol 

Obstet 1989;169:275-282. 
Pope M, Johnston K: Anaphylaxis after thrombin injection of a femoral pseudoa- 

neurysm: Recommendations for prevention. J Vase Surg 2000;32:190-191. 
Porter W, Bigger IA: Stab wounds of the heart and great vessels: A study of 

seventeen cases. Trans Am Clin Climatol Asoc 1939;54:96-104. 
Posner M, Moore EE, Greenholz S, et al: Natural history of untreated inferior 

vena cava injury and assessment of venous access. J Trauma 1986;26:698- 

701. 
PostempskiP: La sutura deivasi sanguigni. Arch Socltal Chir Roma 1886;3:391. 
Potts R, Alguire PC: Pseudoaneurysm of the abdominal aorta: A case report 

and review of the literature. Am J Med Sci 1991;301:265-268. 
Pouyanne H, Arne L, Loiseau P, Mouton L: Considerations sur deux cas de 

thrombose de la carotide interne chez l'enfant. Rev Neurol 1957;97:525. 
Power D-A: Hunter's operation for the cure of aneurysm. BrJ Surg 1929;17:193. 
Pradham D, Juanteguy J, Wilder R: Arterial injuries of extremities associated 

with fractures. Arch Surg 1972;105:582-585. 
Prat A, Warembourg HJ, Watel A, et al: Chronic traumatic aneurysm of the 

descending thoracic aorta (19 cases). J Cardiovasc Surg 1986;27:268-272. 
Pratt G: Importance of a knowledge of vascular surgery in World War II. Am J 

Surg 1942;56:335. 
Prean G, Aragon GE: Arterial embolism from a bullet: A case report. Am Surg 

1976;42:863-865. 
Preston A: Arterial injuries of warfare: Complications and management. Surgery 

1946;20:786. 
Pretre R, Kursteiner K, Khatchatourian G, Faidutti B: Traumatic occlusion of 

the left anterior descending artery and rupture of the aortic isthmus. J 

Trauma 1995;39:388-390. 
Pretre R, LaHarpe R, Cheretakis A: Blunt injury to the ascending aorta: Three 

patterns of presentation. Surgery 1996;119:603-610. 
Pridgen W, Jacobs J: Postoperative arteriovenous fistula. Surgery 1962;51:205. 
Pritchard D, MaloneyJ, Barnhorst DA, Spittel JJ: Traumatic popliteal arteri- 
ovenous fistulas. Arch Surg 1977;112:849-852. 
Proudfoot R, Harris M, Victor D, Bennett S: Arterial bullet embolism. J Ky Med 

Assoc 1989;87:17-18. 
Puckett W: The wounding effect of small high velocity fragments as revealed 

by high speed radiography. J Elisha Mitchell Sci Soc 1946;62:59. 
Puijlaert C: Roentgen diagnosis of traumatic rupture of the aorta. Radiol Clin 

North Am 1976;45:217-235. 
Puri R, Clark J, Corkey P: Axillary artery damage following a closed fracture of 

the neck of the humerus: A case report. Injury 1985;16:426-427. 



RE.qxd 4/16/04 3:19 PM Page 619 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 619 

Qinyao W, Weijin S, Youren Z: New concepts in severe presacral hemorrhage 

during proctectomy. Arch Surg 1982;19:1013-1020. 
Quast D, Shirkey AL, Fitzgerald J, et al: Surgical correction of injuries to the 

vena cava: An analysis of sixty-one cases. J Trauma 1965;5:1-10. 
Queral L, Flinn W, Yao JST, Bergan JJ: Management of peripheral arterial 

aneurysms. Surg Clin North Am 1979;59:693-706. 
Quigley M, Bret PM: Occult pseudoaneurysm of the abdominal aorta follow- 
ing gunshot wound: The importance of plain film findings. J Trauma 

1995;38:269-273. 
Rabhan N, Guillebeau J, Brackney EL: Arteriovenous fistula of the superior 

mesenteric vessels after a gunshot wound. N Engl J Med 1962;266:603. 
Rabinowitz R, Goldfarb D: Surgical treatment of axillosubclavian venous throm- 
bosis: A case report. Surgery 1971;70:703. 
Rabinsky I, Sidhu G, Wagner R: Mid-descending aortic traumatic aneurysms. 

Ann Thorac Surg 1990;50:155-160. 
Raffa H, Sorefan A: Repair of torn aortic root with the left atrial appendage. 

Jpn Heart J 1992;33:265-269. 
Rahat M, Lahat N, Smollar J, et al: Divergent effects of ischemia/reperfusion 

and nitric oxide donor on TNF-alpha mRNA accumulation in rat organs. 

Shock 2001;15:312-317. 
Ramakantan R, Shah P: False aneurysm secondary to aortic cannulation: Rupture 

into lung with fatal hemoptysis during aortography. J Thorac Cardiovasc 

Surg 1989;37:322-323. 
Ramanathan T, Somasundaram K, Yong N: Successful repair of penetrating 

wound of the thoracic aorta. Thorax 1975;30:348-351. 
Rana M, Singh G, Wang P, et al: Protective effects of preheparinization on the 

microvasculature during and after hemorrhagic shock. J Trauma 

1992;32:420-426. 
Randhawa MJ, Menzoian J: Seat belt aorta. Ann Vase Surg 1990;4:370-377. 
Ransdell H, Glass HJ: Gunshot wounds of the heart: A review of 20 cases. Am J 

Surg 1960;999:788-797. 
Ranshoff J: Arteriovenous aneurysm of the superior thyroid artery and vein. 

Surg Gynecol Obstet 1935;61:816. 
Rapoport S, Sniderman U, Morse S, et al: Pseudoaneurysm: A complication of 

faulty technique in femoral artery puncture. Radiology 1985;154:529-530. 
Rau S, Carner D: Brachial plexus compression. Arch Surg 1981;116:175-178. 
Rautio J, Paavolainen P: Afghan war wounded: Experience with 200 cases. 

J Trauma 1988;28:523. 
Ravikumar S, Stahl WM: Intraluminal balloon catheter occlusion for major vena 

cava injuries. J Trauma 1985;25:458-460. 
Razek M, Mnaymneh W, Yacoubian HD: Injuries of peripheral arteries with 

associated bone and soft tissue injuries. J Trauma 1973;13:907. 
RazzoukA, Gundry S, WangN, etal: Pseudoaneurysms of the aorta after cardiac 

surgery or chest trauma. Am Surg 1993;59:818-823. 
Rea W, Sugg W, Wilson L, et al: Coronary artery lacerations: An analysis of 22 

patients. Ann Thorac Surg 1969;7:518-528. 
Read R, Moore EE, Moore FA, et al: Intravascular ultrasound for the diagnosis 

of traumatic aortic disruption: A case report. Surgery 1993;114:624-628. 
Read R, Moore EE, Moore FA, Haenel J: Partial left heart bypass for thoracic 

aorta repair. Arch Surg 1993;128:746-752. 
Reckling F, Peltier L: Acute knee dislocations and their complications. J Trauma 

1969;9:181-191. 



RE.qxd 4/16/04 3:19PM PageS20 



620 VI • THE RICH REFERENCE COLLECTION 



Redman H: Thoracic, abdominal, and peripheral trauma: Evaluation with angio- 
graphy. JAMA 1977;237:2415-2418. 

Reed J, McGinn R, Gorman JF: Traumatic mesenteric arteriovenous fistula 
presenting as the superior mesenteric artery syndrome. Arch Surg 
1986;121:1209. 

Rees R, Bonneval M, Batson R, Hollier L: Angiography in extremity trauma: A 
prospective study. Am Surg 1978;44:661-663. 

Rehn L: Ueber penetrirende herzwunden und hernalt. Arch Klin Chir 
1897;55:56-60. 

Rehr R, Mack M, Firth B: Aortic regurgitation and sinus of Valsalva-right atrial 
fistula after blunt thoracic trauma. Br Heart J 1982;48:410-412. 

Reichle F, Golsorkhi M: Diagnosis and management of penetrating arterial and 
venous injuries in the extremities. Am J Surg 1980;140:365-367. 

Reid C, Kawanishi D, Rahimtoola SH, Chandraratna P: Chest trauma: Evalua- 
tion by two-dimensional echocardiography. Am Heart J 1987;113:971- 
976. 

Reid J, Redman H, WeigeltJA, et al: Wounds of the extremities in proximity to 
major arteries: Value of angiography in the detection of arterial injury. AJR 
1988;151:1053-1059. 

Reid J, WeigeltJA: Forty-three cases of vertebral trauma. J Trauma 1988;28:1007- 
1012. 

Reid J, WeigeltJA, Thai E, Francis H III: Assessment of proximity of a wound 
to major vascular structures as an indication for arteriography. Arch Surg 
1988;123:942-946. 

Reid M: The effect of arteriovenous fistula upon the heart and blood vessels: 
An experiment and clinical study. Bull Johns Hopkins Hosp 1920;31:43- 
50. 

Reid M: Abnormal arteriovenous communications. Acquired and congenital, 
II. The origin and nature of arteriovenous aneurysms, cirsoid aneurysms 
and simple angiomas. Arch Surg (Chicago) 1925;10:996-1009. 

Reid M, McGuire J: Arteriovenous aneurysm. Ann Surg 1938;108:643-693. 

Reiley M, Bond D, Branick RI, Wilson E: Vascular complications following total 
hip arthroplasty: A review of the literature and a report of two cases. Clin 
Orthop 1984;186:23-28. 

Reilly PM, Rotondo MF, Carpenter JP, et al: Temporary vascular continuity during 
damage control: Intraluminal shunting for proximal superior mesenteric 
artery injury. J Trauma 1996;39:757-760. 

Reilly L, Ramos T, Murray S, et al: Optimal exposure of the proximal abdomi- 
nal aorta: A critical appraisal of transabdominal medial visceral rotation. 
J Vase Surg 1994;19:375-389. 

Reines HD, Dill L, Saad S: Neurogenic pulmonary edema and missile emboli. 
J Trauma 1980;20:698-701. 

Reisman J, Morgan A: Analysis of 46 intra-abdominal aortic injuries from blunt 
trauma: Case reports and literature review. J Trauma 1990;30:1294- 
1297. 

Reissman P, Rivkind A, Jurim O, Simon D: Case report: The management of 
penetrating cardiac trauma with major coronary artery injury — Is car- 
diopulmonary bypass essential? J Trauma 1992;33:773-775. 

Requena R, Cherukuri R, Lerner R: A logical approach in the management of 
intraabdominal vascular trauma. Contemp Surg 1983;23:31. 

Reul GJ, Beall AC Jr, Jordan GLJr, Mattox KL: The early operative manage- 
ment of injuries to the great vessels. Surgery 1973;74:862-873. 



RE.qxd 4/16/04 3:19PM PageS21 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 621 

Reul GJ, Mattox KL, Beall AC Jr, Jordan GLJr: Recent advances in the opera- 
tive management of massive chest trauma. Ann Thorac Surg 1973;16:521- 

662. 
Reul GJ, Rubio P, Beall ACJr: The surgical management of acute injury to the 

thoracic aorta. J Thorac Cardiovasc Surg 1974;67:272-281. 
Reyes L, Rubio P, Korompai F, Guinn G: Successful treatment of transection of 

aortic arch and innominate artery. Ann Thorac Surg 1975;19:468-471. 
Reynolds B, Balsano NA: Venography in pelvic fractures: A clinical evaluation. 

Ann Surg 1971;173:104-106. 
Reynolds R, McDowell H, Diethelm A: The surgical treatment of blunt and 

penetrating injuries of the popliteal artery. Am Surg 1983;49:405-410. 
Reynolds R, McDowell H, Diethelm A: The surgical treatment of arterial injuries 

in the civilian population. Am Surg 1979;189:700-708. 
Rhee PM, Acosta J, Bridgeman A, et al: Survival after emergency department 

thoracotomy: Review of published data from the past 25 years. J Am Coll 

Surg 2000;190:288-298. 
Rhee P, Talon E, Eifert S, et al: Induced hypothermia during emergency depart- 
ment thoracotomy: An animal model. J Trauma 2000;48:439-447. 
Rhodes G, Cox C, Silver D: Arteriovenous fistula and false aneurysm as the 

cause of consumption coagulopathy. Surgery 1973;73:535-540. 
Riccen E, Dickens P: Traumatic aneurysm of the abdominal aorta of 27 years 

duration: Case report. US Nav Med Bull 1942;40:692-694. 
Rice W, Wittstruck K: Acute hypertension and delayed traumatic rupture of the 

aorta. JAMA 1951;147:915-917. 
Rich NM: Vietnam missile wounds evaluated in 750 patients. Milit Med 1968; 1 33:9- 

22. 
Rich NM: Wounding power of various ammunition. Resident Physician 

1968;14:72. 
Rich NM: Missile wound evaluation at the 2nd Surgical Hospital in Vietnam. 

In GeorjiadeN (ed) : Plastic and Maxillofacial Trauma Symposium. St. Louis, 

CVMosby, 1969. 
Rich NM: Vascular trauma in Vietnam. J Cardiovasc Surg 1970;11:368-377. 
Rich NM: Surgery for arterial trauma. In Dale WA (ed): Management of 

Arterial Occlusive Disease. Chicago, Year Book Medical Publishers, 

1971. 
Rich NM: Complications of operations for vascular trauma. In Beebe HG (ed) : 

Complications in Vascular Surgery. Philadelphia, JB Lippincott Co, 

1973. 
Rich NM: Vascular trauma. Surg Clin North Am 1973;53:1367-1392. 
Rich NM: Vascular injuries. In Whelan TJ (ed) : Emergency War Surgery: NATO 

Handbook. Washington, DC, Government Printing Office, 1975. 
Rich NM: Weapons and wounds. J Trauma 1975;15:464-465. 
Rich NM: Bullets and blood vessels. Surg Clin North Am 1978;58:995-1003. 
Rich NM: Principles and indications for primary venous repair. Surgery 

1982;91:492-496. 
Rich NM: Vascular injuries. In Cameron J (ed) : Current Surgical Therapy. 

Philadelphia, BC Decker, 1984. 
Rich NM: The management of trauma to the carotid-vertebral system. In 

Robicsek F (ed): Extracranial Cerebrovascular Disease Diagnosis and 

Management. New York, Macmillan Publishing, 1986. 
Rich NM: Venous injuries. In Sabiston DJ (ed): Textbook of Surgery. 

Philadelphia, WB Saunders, 1986, pp 2009-2018. 



RE.qxd 4/16/04 3:19PM PageS22 



622 VI • THE RICH REFERENCE COLLECTION 



Rich NM: Arterial injuries associated with fracture, dislocation, and extensive 
soft tissue trauma. In Ernst CB, Stanley JC (eds): Current Therapy in Vas- 
cular Surgery. Philadelphia, BC Decker, 1987. 

Rich NM: Management of venous trauma. Surg Clin North Am 1988;68:809- 
821. 

Rich NM: Gunshot wound to the groin. In Brewster DC (ed): Common Prob- 
lems in Vascular Surgery. Chicago, Year Book Medical Publishers, 1989, 
pp 305-308. 

Rich NM: Mechanism and reversibility of proximal arterial dilatation with large 
arteriovenous fistulas. In Veith F (ed): Current Critical Problems in Vas- 
cular Surgery. St. Louis, Quality Medical Publishing, 1989. 

Rich NM: Current status of lower extremity venous repair. In Veith F (ed) : Current 
Critical Problems in Vascular Surgery. St. Louis, Quality Medical Publish- 
ing, 1990. 

Rich NM: Trauma: Responsibility, resources, and responsiveness (Scudder 
Oration). ACS Bull 1990;76:6-13. 

Rich NM: Brief history of vascular trauma. Trauma Q 1991;7:1-5. 

Rich NM: Vascular trauma: Abrief military perspective. WorldJ Surg 1992;16:938- 
939. 

Rich NM: History of vascular trauma. In Flanigan DP, Schuler J, Meyer JP (eds) : 
Civilian Vascular Trauma. Philadelphia, Lea & Febiger, 1992, pp 3-7. 

Rich NM: Military vascular injuries in Croatia. Cardiovasc Surg 1993;1:2. 

Rich NM: Surgeon's response to battlefield vascular trauma (Hume Memorial 
Lecture). Am J Surg 1993;166:91-96. 

Rich NM: Venous injuries. In Sabiston DJ (ed): Textbook of Surgery. 
Philadelphia, WB Saunders, 1995. 

Rich NM: Commentary. In Ivatury RR, Cayten C (eds): The Textbook of 
Penetrating Trauma. Philadelphia, Williams & Wilkins, 1996. 

Rich NM, Amato JJ, Billy LJ: Arterial thrombosis secondary to temporary cavi- 
tation. Surg Dig 197l;6:12. 

Rich NM, Andersen CA, Ricotta J, et al: Arterial trauma: A remaining problem 
of increasing magnitude. Milit Med 1977;141:847-852. 

Rich NM, Baugh JH, Hughes CW: Popliteal artery injuries in Vietnam. Am J 
Surg 1969;118:531-534. 

Rich NM, Baugh JH, Hughes CW: Acute arterial injuries in Vietnam: 1,000 
cases. J Trauma 1970;10:359-369. 

Rich NM, Baugh JH, Hughes CW: Significance of complications associated 
with vascular repairs performed in Vietnam. Arch Surg 1970;100:646- 
651. 

Rich NM, Clagett GP, Salander J, Piscevic S: The Matas/Soubbotitch connec- 
tion. Surgery 1983;93:17-19. 

Rich NM, Clarke J, Baugh JH: Successful repair of a traumatic aneurysm of the 
abdominal aorta. Surgery 1969;66:492-496. 

Rich NM, Collins GJ Jr, Andersen CA, et al: Venous trauma: Successful venous 
reconstruction remains an interesting challenge. Am J Surg 1977;134:226- 
230. 

Rich NM, Collins GJJr, Andersen CA, et al: Missile emboli. J Trauma 1978; 
18:236-239. 

Rich NM, Collins GJ, Andersen CA, McDonald P: Autogenous venous interpo- 
sition grafts in repair of major venous injuries. J Trauma 1977;17:512-520. 

Rich NM, Collins GJJr, Hobson RW II, et al: Carotid-axillary bypass: Clinical 
and experimental evaluation. Am J Surg 1977;134:805-808. 



RE.qxd 4/16/04 3:19PM PageS23 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 623 

Rich NM, Collins GJJr, YoukeyJR: Cervical and Thoracic Outlet Venous Injuries. 

Surgery of the Veins. New York, Grune & Stratton, 1985. 
Rich NM, Gomez ER, Coffey JA, et al: Long-term follow-up of venous recon- 
struction following trauma. In Bergan JJ, Yao JST (eds) : Venous Disorders. 

Philadelphia, WB Saunders, 1991. 
Rich NM, Gomez ER, Jackson MR: Pulsating hematomas and pseudoaneurysms: 

Should they be managed by noninvasive compression under duplex control? 

In Veith F (ed): Current Critical Problems in Vascular Surgery. St. Louis, 

Quality Medical Publishing, 1994, pp 319-323. 
Rich NM, Hobson RW II: Historical background of repair venous injuries. In 

Witkin E (ed) : Venous Diseases, Medical and Surgical Management. Hague, 

the Netherlands, Mouton, 1974. 
Rich NM, Hobson RW II: Trauma to the venous system. In Swan KG, Hobson 

RWII, Reynolds D, etal (eds): Symposium on Venous Surgery in the Lower 

Extremities. St. Louis, Green Publishers, 1975. 
Rich NM, Hobson RW II: Venous trauma: Emphasis for repair is indicated. 

J Cardiovasc Surg 1975; special issue:571. 
Rich NM, Hobson RW II, Collins GJJr: Traumatic arteriovenous fistulas and 

false aneurysms: A review of 558 lesions. Surgery 1975;78:817-828. 
Rich NM, Hobson RW II, Collins GJJr: Elective vascular reconstruction after 

trauma. Am J Surg 1975;130:712-719. 
Rich NM, Hobson RWII, Collins GJJr, Anderson CA: The effect of acute popliteal 

venous interruption. Ann Surg 1976;183:365-368. 
Rich NM, Hobson RW II, Fedde C: Vascular trauma secondary to diagnostic 

and therapeutic procedures. Am J Surg 1974;128:715-721. 
Rich NM, Hobson RW II, Fedde C, Collins GJJr: Acute common femoral arte- 
rial trauma. J Trauma 1975;15:628-637. 
Rich NM, Hobson R II, Hutton JE Jr: Arterial trauma: Lessons of the Vietnam 

war. Paper presented at: Annual International Vascular Meeting, Paris, 

France, 1995. 
Rich NM, Hobson RW II, Hutton JE Jr: Traumatismes arteriels: les lecons de la 

guerre du Vietnam. In Kieffer E (ed) : Traumatismes Arteriels. Paris, AERCV, 

1995. 
Rich NM, Hobson RWIIJarstfer B, Geer TM: Subclavian artery trauma.J Trauma 

1973;13:485-496. 
Rich NM, Hobson RWII, Wright CB: Historical aspects of direct venous recon- 
struction. In Bergan JJ, Yao JST (eds) : Symposium on Venous Problems in 

Honor of Geza de Takats. Chicago: Year Book Medical Publishers, 1978. 
RichNM, Hobson RWII, Wright CB, Fedde C: Repair of lower extremity venous 

trauma: A more aggressive approach required. J Trauma 1974;14:639-652. 
Rich NM, Hobson RW II, Wright CB, Swan KG: Techniques of venous repair. 

In Swan KG, Hobson RWII, Reynolds D, etal (eds): Symposium on Venous 

Surgery in the Lower Extremities. St. Louis, Warren H Green Publishers, 

1975. 
Rich NM, Hughes CW: Vietnam Vascular Registry: A preliminary report. Surgery 

1969;65:218-226. 
Rich NM, Hughes CW: Stistiques sur les plaies vasculaires au Vietnam (4500 

Cas). Social Med Chir 1970;9:805-809. 
Rich NM, Hughes CW: Rapport sur les plais vasculaires au Vietnam (Transla- 
tion, D. Rignault). Revue des corps de Sante 1971;12:673. 
Rich NM, Hughes CW: Fifty years' progress in vascular surgery. Bull Am Coll 

Surg 1972;57:35. 



RE.qxd 4/16/04 3:19PM PageS24 



624 VI • THE RICH REFERENCE COLLECTION 



Rich NM, Hughes CW: The fate of prosthetic material used to repair vascular 
injuries in contaminated wounds. J Trauma 1972;12:459-467. 

Rich NM, Hughes CW, Baugh JH: Management of venous injuries. Ann Surg 
1970;171:724-730. 

Rich NM, Jarstfer B, Geer TM: Popliteal artery repair failure: Causes and pos- 
sible prevention. J Cardiovasc Surg 1974;15:340-351. 

Rich NM, Jarstfer B, Geer TM: Concomitant popliteal arterial and venous trauma. 
In Swan KG, Hobson RWII, Reynolds D, et al (eds): Symposium on venous 
surgery in the lower extremities. St. Louis, Warren H Green Publishers, 
1975, pp 29-40. 

Rich NM, Johnson E, Dimond FJr: Wounding power of missiles used in the 
Republic of Vietnam. JAMA 1967;199:157. 

Rich NM, Levin P, HuttonJEJr: Effect of distal arteriovenous fistulas on venous 
graft patency. In Swan KG, Hobson RW II, Reynolds D, et al (eds) : Sym- 
posium on Venous Surgery in the Lower Extremities. St. Louis, Warren H 
Green Publishers, 1975. 

Rich NM, Manion W, Hughes CW: Surgical and pathological evaluation of vas- 
cular injuries in Vietnam. J Trauma 1969;9:279-291. 

Rich NM, Metz CJ, HuttonJEJr, et al: Internal versus external fixation of frac- 
tures with concomitant vascular injuries in Vietnam. J Trauma 1971 ;1 1:463- 
473. 

Rich NM, Rhee P: An historical tour of vascular injury management: From its 
inception to the new millennium. Surg Clin North Am 2001;81:1199-1215. 

Rich NM, Rob CG: Ischemia-induced myonecrosis, myoglobinuria and secondary 
renal failure. In Ernst CB, Stanley JC (eds): Current Therapy in Vascular 
Surgery. Philadelphia, BC Decker, 1990. 

Rich NM, Salander J, Orecchia P, et al: Venous injuries. In Bergan JJ, Yao JST 
(eds): Vascular Surgical Emergencies. Orlando, Grune & Stratton, 1987, 
pp 275-282. 

Rich NM, Shumacker HB Jr: Cold injury. In Ernst CB, StanleyJC (eds) : Current 
Therapy in Vascular Surgery. Philadelphia, BC Decker, 1990. 

Rich NM, Spencer FC: Vascular Trauma. Philadelphia, WB Saunders, 1978. 

Rich NM, Sullivan W: Clinical recanalization of an autogenous vein graft in the 
popliteal vein. J Trauma 1972;12:919-920. 

Richards AJ, Lamis PJ, Rogers JJ, Bradham GB: Laceration of abdominal aorta 
and study of intact abdominal wall as tamponade: Report of survival and 
literature review. Ann Surg 1966;164:321-324. 

Richardson D, Shina M, Miller F, Bergamini TM: Peripheral vascular compli- 
cations of coronary angioplasty. Am Surg 1989;55:675-680. 

Richardson JD: Indications for thoracotomy in thoracic trauma. Curr Surg 
1985;42:361-364. 

Richardson JD, Fallat M, Nagaraj H, et al: Arterial injuries in children. Arch 
Surg 1981;116:685-690. 

Richardson JD, Simpson C, Miller F: Management of carotid artery trauma. 
Surgery 1988;104:673-680. 

RichardsonJD , Vitale G, Flint LM: Penetrating arterial trauma: Analysis of missed 
vascular injuries. Arch Surg 1987;122:678-683. 

Richardson JJ, Jurkovich GJ, Walker G, et al: A temporary arteriovenous shunt 
in the management of traumatic venous injuries of the lower extremity 
(Scribner). J Trauma 1986;26:503-509. 

Richardson R, Khandeadar A, Moseley P: Traumatic rupture of the thoracic 
aorta. South Med J 1979;72:300-302. 



RE.qxd 4/16/04 3:19PM PageS25 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 625 

Ricketts R, Finck E, Yellin AE: Management of major arteriovenous fistulas by 

arteriography techniques. Arch Surg 1978;113:1153. 
Ricks R, Howell J, Beall AC Jr, DeBakey ME: Gunshot wounds of the heart: A 

review of 31 cases. Surgery 1965;57:787-790. 
Rielly J, Brandt ML, Mattox KL, Pokorny W: Thoracic trauma in children. 

J Trauma 1993;34:329-331. 
Risely T, McClerkin W: Bullet transection of both carotid arteries: Delayed repair 

with recovery. Am J Surg 1971;121:385-386. 
Rittenhouse E, Dillard D, Winterscheid L, Merendino K: Traumatic rupture of 

the thoracic aorta: A review of the literature and a report of five cases with 

attention to special problems in early management. Ann Surg 1969;1 70:87- 

100. 
Ritter D, Chang F: Delayed hemothorax resulting form stab wounds to the inter- 
nal mammary artery. J Trauma 1995;39:586-589. 
Rob CG: Diagnosis of abdominal trauma in warfare. Surg Gynecol Obstet 

1947;85:147-154. 
Rob CG: Reconstruction of a stenosed internal carotid artery. Br Med J 

1956;2:1265. 
Rob CG: Die indikationstellung zur operativen widerherstellung der aorta 

und der groesseren arterien. Arch and Deutschen Zt J Chir 1957;287: 

305. 
Rob CG: Obliterations of the aortic bifurcation (Leriche's syndrome). Car- 

dioangiologica 1957;5:1. 
Rob CG: Arterial wounds, acute ischemia of the limbs. J Cardiovasc Surg 

1963;4:249-252. 
Rob CG: A history of arterial surgery. Arch Surg 1972;105:821-823. 
Rob CG: Military surgery, our debt to our patients and predecessors. J R Army 

Med Corps 1986;132:11-15. 
Rob CG, Battle S: Arteriovenous fistula following the use of the Fogarty balloon 

catheter. Arch Surg 1971;102:144-145. 
Rob CG, Standeven A: Closed traumatic lesions of the axillary and brachial 

arteries. Lancet 1956;1:597-599. 
RobbsJ, Baker LW: Subclavian and axillary artery injury. S Afr MedJ 1977;51 :227- 

231. 
Robbs J, Baker LW: Arterial trauma involving the lower limb. J Trauma 

1978;18:324-328. 
RobbsJ, Baker LW: Major arterial trauma: Review of experience with 267 injuries. 

Br J Surg 1978;65:532-538. 
RobbsJ, Baker LW: Cardiovascular trauma. Curr Prob Surg 1984;21:7-87. 
RobbsJ, Baker LW, Human R, et al: Cervicomediastinal arterial injuries. Arch 

Surg 1981;116:663-668. 
RobbsJ, Naidoo K: Nerve compression injuries due to traumatic false aneurysm. 

Ann Surg 1984;200:80-82. 
RobbsJ, Reddy E: Management options for penetrating injuries to the great 

veins of the neck and superior mediastinum. Surg Gynecol Obstet 

1987;165:323-326. 
Roberson G: Combined stab wounds of the aorta and vena cava of the abdomen. 

Arch Surg 1967;95:12-15. 
Roberts R, String T: Arterial injuries in extremity shotgun wounds: Requisite 

factors for successful management. Surgery 1984;96:902-907. 
Roberts S, Main D, PindertonJ: Surgical therapy of femoral artery pseudoa- 

neurysm after angioplasty. Am J Surg 1987;10:676-680. 



RE.qxd 4/16/04 3:19PM PageS26 



626 VI • THE RICH REFERENCE COLLECTION 



Robinson J, Moyer C: Comparison of late sequelae of common and superficial 

femoral vein ligations. Surgery 1954;35:690-697. 
Robinson N, Flotte C: Traumatic aneurysm of the carotid arteries. Am Surg 

1974;40:121-124. 
Robison R, Brown J, Caldwell R, Stone K: Management of asymptomatic intrac- 
ardiac missiles using echocardiography. J Trauma 1988;28:1402-1403. 
Roche K, Genieser N, Berger DK, Ambrosino MM: Traumatic abdominal pseudoa- 

neurysm secondary to child abuse. Pediatr Radiol 1995;25:S247-S248. 
Roe B: Cardiac trauma including injury of great vessels. Surg Clin North Am 

1972;52:573-583. 
Rohman M, Ivatury RR, Steichen F, et al: Emergency room thoracotomy for 

penetrating cardiac injuries. J Trauma 1983;23:570-576. 
Rohrer M, Cardullo P, Pappas A, et al: Axillary artery compression and throm- 
bosis in throwing athletes. J Vase Surg 1990;11:761-769. 
Rojas R, Levitsky S, Stansel HCJr: Acute traumatic subclavian Steal Syndrome. 

J Thorac Cardiovasc Surg 1966;51:113-115. 
Rollins D, Bernhard VM, TowneJB: Fasciotomy: An appraisal of controversial 

issues. Arch Surg 1981;116:1474-1481. 
Romanoff H, Goldberger S: Major peripheral vein injuries. Vase Surg 

1976;10:157-163. 
Roostar L: Indications for surgery in penetrating chest injuries. Ann Chirur- 

giae Gynaecol 1993;82:177-181. 
Roper B, Provan J: Late thrombosis of the femoral artery complicating fracture 

of the femur. J Bone Joint Surg 1965;47:510-513. 
Roques X, Bourdeaudhui A, Collet D, et al: Traumatic rupture and aneurysm 

of the aortic isthmus: Late results of repair by direct suture. Ann Vase Surg 

1989;3:47-51. 
Rose S, Moore EE: Angiography in patients with arterial trauma: Correlations 

between angiographic abnormalities, operative findings and clinical 

outcome. AJR Am J Roentgenol 1987;149:613-619. 
Rose S, Moore EE: Emergency trauma angiography: Accuracy, safety, and pit- 
falls. AJR Am J Roentgenol 1987;148:1243-1246. 
Rose S, Moore EE: Trauma angiography: The use of clinical findings to improve 

patient selection and case preparation. J Trauma 1988;28:240-245. 
Rosemurgy AS, Norris P, Olson S, et al: Prehospital traumatic cardiac arrest: 

The cost of futility. J Trauma 1993;35:468-474. 
RosenbergJ, Bredenberg CE, Marvasti M, et al: Blunt injuries to the aortic arch 

vessels. Ann Thorac Surg 1989;48:508-513. 
Rosenblatt M, Lemer J, Best LA: Thoracic wounds in Israeli battle casualties 

during the 1982 evacuation of wounded from Lebanon. J Trauma 

1985;4:350. 
Rosenbloom M, Fellows B: Chronic pseudoaneurysm of the popliteal artery 

after blunt trauma. J Vase Surg 1989;10:187-189. 
Rosensweig J, Simon MA: Traumatic aneurysm of the axillary artery: A golf 

hazard. Can Med Assoc J 1965;93:165-167. 
Rosenthal D, Ellison R, Luke J: Traumatic superior mesenteric arteriovenous 

fistula: Report of a case and review of the literature. J Vase Surg 1987;5:486- 

491. 
Rosenthal J, Caspar MR, Gjerdrum T, Newman J: Vascular injuries associated 

with fractures of the femur. Arch Surg 1975;110:494-499. 
Ross S, Ransom K, Shatney C: The management of venous injuries in blunt 

extremity trauma. J Trauma 1985;25:150-153. 



RE.qxd 4/16/04 3:19PM PageS27 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 627 

Roth S, Wheeler J, Gregory R, et al: Blunt injury of the abdominal aorta: A 
review. J Trauma 1997;42:748-755. 

Rothlin M, Naf R, Amgwerd M, et al: Ultrasound in blunt abdominal and tho- 
racic trauma. J Trauma 1993;34:488-495. 

Rotondo M, Schwab CW, McGonigal MD, et al: "Damage control": An approach 
for improved survival in exsanguinating penetrating abdominal injury. 
J Trauma 1993;35:375-383. 

Rovito P: Arterial caval shunting in blunt hepatic vascular injury. Ann Surg 
1987;205:318-321. 

Roy-Shapira A, Levi I, Khoda J: Sternal fractures: A red flag or a red herring? 
J Trauma 1994;37:59-61. 

Rozin L, Perper J: Spontaneous fatal perforation of aorta and vena cava by 
Mobin-Uddin umbrella. Am J Forensic Med Pathol 1989;10:149-151. 

Rozycki G, Ochsner M, Schmidt J, et al: A prospective study of surgeon-per- 
formed ultrasound as the primary adjuvant modality for injured patient 
assessment. J Trauma 1995;39:492-500. 

Rozycki G, Kraut E: Isolated blunt rupture of the infrarenal inferior vena cava: 
The role of ultrasound and computed tomography in an occult injury. 
J Trauma 1995;38:402-405. 

Rpatopoulos V, Sheiman R, Phillips D, et al: Traumatic aortic tear: Screening 
with chest CT. Radiology 1992;182:667-673. 

Ruberti U, Odero A, Arpesani A, et al: Acute ruptures of the thoracic aorta. 
Personal experience. J Cardiovasc Surg 1987;28:81-84. 

Ruberti U, Odero A, Arperani A, et al: Surgical treatment of thoracic aortic 
aneurysms: Personal experience. J Cardiovasc Surg 1988;29:245-256. 

Rubio P, Reul GJ, Beall AC Jr, et al: Acute carotid artery injury: 25 years' expe- 
rience. J Trauma 1974;14:967-973. 

Rubio P, Reul GJ: Penetrating cardiac injury by wire thrown from a lawn mower. 
Int Surg 1979;64:9-10. 

Rudich M, Rowland M, Seibel R, Border J: Survival following a gunshot wound 
of the abdominal aorta and inferior vena cava. J Trauma 1978;18:548-549. 

Rush D, Frame S, Bell RM, Berg EE: Does open fasciotomy contribute to mor- 
bidity and mortality after acute lower extremity ischemia and revascular- 
ization? J Vase Surg 1989;10:343. 

Ruskey J, Lieberman M, Shaikh K, Talucci R: Unusual subclavian artery lacer- 
ations resulting from lap-shoulder seatbelt trauma: Case reports. J Trauma 
1989;29:1598-1600. 

Russell W, Sailors D, Whittle T, et al: Limb salvage versus traumatic amputation: 
A decision based on a seven part predictive index. Ann Surg 1991;213:473. 

Russo P, Orszulak T, Arnold PG, et al: Concomitant repair of a chronic trau- 
matic aortic aneurysm with tracheal erosion. Ann Thorac Surg 1987;43:559- 
560. 

Rutherford R: Diagnostic evaluation of extremity vascular injuries. Surg Clin 
North Am 1988;68:683-691. 

Rutherford R, Baue AE: Extraanatomic bypass. In Rutherford R (ed) : Vascular 
Surgery. Philadelphia, WB Saunders, 1989, pp 705-716. 

Rutsaert R, Van Schil P, Martens C, et al: Occlusion of the left common femoral 
artery after total hip replacement. Report of a case and review of the lit- 
erature. J Cardiovasc Surg 1988;29:216-218. 

Ryan J, Cooper G, Haywood I, Milner S: Field surgery on a future conventional 
battlefield: Strategy and wound management. Ann R Coll Surg Engl 
1991;73:13. 



RE.qxd 4/16/04 3:19PM PageS28 



628 VI • THE RICH REFERENCE COLLECTION 



Ryan W, Snyder WH III, Bell T, Hunt J: Penetrating injuries of the iliac vessels: 
Early recognition and management. Am J Surg 1982;144:642-645. 

Rybeck B: Missile wounding and hemodynamic effects of energy absorption. 
Acta Chir Scand 1974,450:1. 

RybuckJ, Thomford N: Acute occlusion of the infrarenal aorta from blunt trauma. 
Am Surg 1969;36:444-447. 

Saaman H: The hazards of radial artery pressure monitoring. J Cardiovasc Surg 
1971;12:342. 

Sachtello C, Ernst CB, Griffen WJ: The acute ischemic upper extremity: Selected 
management. Surgery 1974;76:1002. 

Sadow S, Murray C, Wilson R, et al: Traumatic rupture of ascending aorta and 
left main bronchus. Ann Thorac Surg 1988;45:682-683. 

Sagalowski A, McConnell J, Peters P: Renal trauma requiring surgery: An analy- 
sis of 185 cases. J Trauma 1983;23:128-131. 

Sahler O, Jaretzki AJ: Thrombosis of the abdominal aorta: Its production by 
ureteral compression during intravenous pyelography. Arch Surg 
1968;96:76-77. 

Sako Y, Varco R: Arteriovenous fistula: Results of management of congenital 
and acquired forms, blood flow measurements and observations on proxi- 
mal arterial degeneration. Surgery 1970;67:40-61. 

Salam A, Stewart M: New approach to wounds of the aortic bifurcation and infe- 
rior vena cava. Surgery 1985;98:105-108. 

Salander J, YoukeyJ, Rich NM, et al: Vascular injury related to lumbar disk 
surgery. J Trauma 1984;24:628-631. 

Salas A, PanesJ, Rosenbloom C, etal: Differential effects of a nitric oxide donor 
on reperfusion-induced microvascular dysfunction in diabetic and non- 
diabetic rats. Diabetologia 1999;42:1350-1358. 

Saletta J, Freeark R: The partially severed artery. Arch Surg 1968;97:198- 
205. 

Saletta J, Freeark R: Vascular injuries associated with fractures. Orthop Clin 
North Am 1970;1:93. 

SalettaJ, Freeark R: Injuries to the profunda femoris artery. J Trauma 1972;12: 
778. 

SalettaJ, Lowe RLL: Penetrating trauma of the neck. J Trauma 1976;16:579- 
587. 

Saletta S, Lederman E, Fein S, et al: Transesophageal echocardiography for the 
initial evaluation of the widened mediastinum in trauma patients. J Trauma 
1995;39:137-141. 

Samaan H: Vascular injuries of the upper thorax and the root of the neck. Br 
J Surg 1971;58:881-886. 

Samson P: Two unusual cases of war wounds of the heart. Surgery 1946;20:373. 

Samson P: Battle wounds and injuries of the heart and pericardium: Experi- 
ences in forward hospitals. Ann Surg 1948;127:1127. 

Samson R, Pasternak B: Traumatic arterial spasm: Rarity or nonentity. J Trauma 
1980;20:607-609. 

Samson R, Sprayregen S, Veith F, et al: Management of angioplasty complica- 
tions, unsuccessful procedures and early and late failures. Ann Surg 
1984;199:234-240. 

Santanello S, Barnes S, Price J, Falcone R: Below the belt: Seat belt related 
injuries. Contemp Surg 1991;39:29-32. 

Sato O, Tada Y, Sudo K, et al: Arteriovenous fistula following central venous 
catheterization. Arch Surg 1986;121:729-731. 



RE.qxd 4/16/04 3:19PM PageS29 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 629 

Saueracker A, McCroskey B, Moore EE: Intraoperative hypogastric artery 
embolization for life-threatening pelvic hemorrhage: Apreliminary report. 
J Trauma 1987;27:1127-1129. 

Sauerbruch: Uber die Verwendbarkeit der pneumatischen Kammer fur die Her- 
schirurgie. Centralbl Chir 1907;34:44. 

Saunders M, Riberi A, Massullo E: Delayed traumatic superior mesenteric arte- 
riovenous fistula after a stab wound: Case report. J Trauma 1992;32:101- 
106. 

Savage R: Popliteal artery injury associated with knee dislocation: Improved 
outlook? Am Surg 1980;46:627-632. 

Saylam A, Melo J, Ahmad A, et al: Early surgical repair in traumatic rupture of 
the thoracic aorta. (Report of 9 cases and review of the current concepts) . 
J Cardiovasc Surg 1980;21:295-302. 

Scalea T, Sclafani S: Angiographically placed balloons for arterial control: A 
description of a technique. J Trauma 1991;31:1671-1677. 

Schaff H, Brawley RK: Operative management of penetrating vascular injuries 
of the thoracic outlet. Surgery 1977;82:182-191. 

Schede M: Zur frage von derjodoformvergiftung. Zentralb Chir Beil 1882;9:33- 
38. 

Scheerlinck T, Van den Brande P: Post-traumatic intima dissection and throm- 
bosis of the external iliac artery in a sportsman. SurgJ Vase Surg 1994;8:645. 

Schenk WJ, Bahn RA, Cordell A, Stephens J: The regional hemodynamics of 
acute experimental arteriovenous fistulas. Surg Gynecol Obstet 
1957;105:733. 

Schenk WJ, MartinJ, Leslie M, Portin B: The regional hemodynamics of chronic 
arteriovenous fistulas. Surg Gynecol Obstet 1960;110:44-50. 

Schlosser W, Spillner G, Breymann T, UrbayniB: Vascular injuries in orthopaedic 
surgery. J Cardiovasc Surg 1982;23:323-327. 

Schmidt C, JacobsonJ: Thoracic aortic injury: A ten-year experience. Arch Surg 
1984;119:1244-1246. 

Schmitt H, Armstrong RC: Wounds causing loss of limb. Surg Gynecol Obstet 
1970;130:682-684. 

Schoenfeld H: Heart Injuries with suture. Ann Surg 2001;87:823-828. 

Schonholtz G, Jahnke EJJr: Occult injury of the thoracic aorta associated with 
orthopaedic trauma. J Bone Joint Surg 1964;46A:1421. 

Schorn B, Reitmeier F, Falk V, et al: True aneurysm of the superior gluteal 
artery: Case report and review of the literature. J Vase Surg 1995;21:851. 

Schott U, Lindbom L, Sjostrand U: Hemodynamic effects of colloid concen- 
tration in experimental hemorrhage: A comparison of Ringer's acetate, 
3% dextran-60 and 6% dextran-70. Crit Care Med 1998;16:346-352. 

Schramek A, Hashmonai M: Distal arteriovenous fistula for the prevention of 
occlusion of venous interposition grafts to veins. J Cardiovasc Surg 
1974;15:392-395. 

Schramek A, Hashmonai M, Farbstein J, Adler O: Reconstructive surgery in 
major vein injuries in the extremities. J Trauma 1975;15:816-822. 

Schramek A, Hashmonai M: Vascular injuries in the extremities in battle casu- 
alties. Br J Surg 1977;64:644-648. 

Schwab C, Lawson R, Lind J, Garland L: Aortic injury: Comparison of supine 
and upright portable chest films to evaluate the widened mediastinum. 
Ann Emerg Med 1984;13:896-899. 

Schwab C, Adcock OT, Max H: Emergency department thoracotomy (EDT) : A 
26-month experience using an "agonal" protocol. Am Surg 1986;52:20-28. 



RE.qxd 4/16/04 3:19 PM Page 630 



630 VI • THE RICH REFERENCE COLLECTION 



Schwab C, McMahan D, Phillips G, Pentecost M: Aortic balloon control of a 

traumatic aortoenteric fistula after damage control laparotomy: A case 

report. J Trauma 1996;40:1021-1023. 
Schwartz A: The historical development of methods of hemostasis. Surgery 

1958;44:604. 
Schwartz D, Haller JAJr: Open anterior hip dislocation with femoral vessel tran- 
section in a child. J Trauma 1974;14:1054. 
Schwartz L,McCannR: Traumatic false aneurysm of the common carotid artery 

presenting as a mediastinal mass: A case report. J Vase Surg 1989;10:281- 

284. 
Schwartz ML, Fisher R, Sako Y, et al: Posttraumatic aneurysms of the thoracic 

aorta. Surgery 1975;78:589-593. 
Sclafani SJ, Panetta T, Goldstein A, et al: The management of arterial injuries 

caused by penetration of Zone III of the neck. J Trauma 1985;25:878- 

881. 
Sclafani S, Cooper R, Shaftan GW, et al: Arterial trauma: Diagnostic and ther- 
apeutic angiography. Radiology 1986;161:165-172. 
Sclafani S, Cavaliere G, Atweh N, et al: The role of angiography in penetrating 

neck trauma. J Trauma 1991;31:557-563. 
Scott C, Pinson W, Inahara T: Common femoral artery injury by blunt trauma: 

A case report. Surgery 1984;96:122-125. 
Scott R. Aspects of military surgery. Injury 1970;2:116. 
Scultetus AH, Villavicencio JL, Rich NM: Historical review: Facts and fiction 

surrounding the discovery of the venous valves. J Vase Surg 2001;33:435- 

441. 
Sechas M, Fotiadis C, Baramily B, Skalkeas G: Plaie des vaisseaux mesenteriques 

superieurs a propos d'un cas. Ann Chir 1988;42:423-425. 
Seeley S: Vascular surgery at Walter Reed Army Hospital. US Armed Forces 

MedJ 1954;8. 
Seeley S, Hughes CW, Cooke F, Elkin DC: Traumatic arteriovenous fistulas and 

aneurysms in war wounded. Am J Surg 1952;83:471-479. 
Seeley S, Hughes CW, Jahnke EJJr: Surgery of the popliteal artery. Ann Surg 

1953;138:712-717. 
Seeley S, Hughes CW, Jahnke EJJr: Major vessel damage in lumbar disc oper- 
ation. Surgery 1954;35:421-429. 
Sefczek D, Sefczek RJ, Deeb Z. Radiographic signs of acute traumatic 

rupture of the thoracic aorta. AJR Am J Roentgenol 1983;141:1259- 

1262. 
Segar R, Palmisano D: Repair of gunshot wounds of the aorta and vena cava. 

Ohio State MedJ 1969;65:501-505. 
Seguin J, Bouillon P, Aubry P, et al: Aorto-right ventricular shunt and associ- 
ated aortic valve injury resulting from a penetrating wound of the heart. 

Thorac Cardiovasc Surg 1984;32:386-388. 
Seidenberg B, Hurwitt E: Retrograde femoral-Seldinger-aortography: Surgical 

complications in twenty-six cases. Ann Surg 1966;163:221-226. 
Seiler JI, Richardson JD: Amputation after extremity injury. Am J Surg 

1986;152:260-264. 
Seltzer S, D'Orsi C, Kirshner R, DeWeese JA: Traumatic aortic rupture: Plain 

radiographic findings. AJR 1981;137:1011-1014. 
Sencert L: Les Blessures des Vaisseaux. Paris, Masson & Cie, 1917. 
Sethi G, Scott S, Takaro T: False aneurysm of the abdominal aorta due to blunt 

trauma. Ann Surg 1975;182:33-36. 



RE.qxd 4/16/04 3:19 PM Page 631 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 631 

Sethi G, Scott S, Bhayana J, Takaro T: Traumatic avulsion of the innominate 

artery: Report of a case and review of literature. J Cardiovasc Surg 

1975;16:171-175. 
Sethi G, Scott S: Subclavian artery laceration due to migration of a Hagie Pin. 

Surgery 1976;80:644. 
Shackford SR, Sise MJ: Renal and mesenteric vascular trauma: In Vascular Injuries 

in Surgical Practice. In Bongard FS, Wilson SE, Perry MO (eds) : Norwalk, 

Conn, San Mateo, Appleton & Lange, 1991, pp 173-184. 
Shadow S, Murray CI, Wilson R, et al: Traumatic rupture of ascending aorta 

and left main bronchus. Ann Thorac Surg 1988;45:682-683. 
Shaftan GW, Chiu C, Dennis C, Harris B: Fundamentals of physiologic control 

of arterial hemorrhage. Surgery 1965;58:851-856. 
Shah D, Leather R, Corson J, Karmody A: Polytetrafluoraethylene grafts in the 

rapid reconstruction of acute contaminated peripheral vascular injuries. 

Am J Surg 1984;148:229-233. 
Shah D, Naraynsingh V, Leather R, et al: Advances in the management of acute 

popliteal vascular blunt injuries. J Trauma 1985;25:793-797. 
Shah D, Corson J, Karmody A, et al: Optimal management of tibial arterial 

trauma. J Trauma 1988;28:228-234. 
Shah PM, Ito K, Clauss RH, et al: Expanded microporous polytetrafluroethyl- 

ene (PTFE) grafts in contaminated wounds: Experimental and clinical study. 

J Trauma 1983;23:1030-1033. 
Shah PM, Mackey R, Babu SC, et al: Pseudoaneurysm of anterior tibial artery 

after occlusion from blunt trauma: Nonoperative management. J Trauma 

1985;25:656-658. 
Shah PM, Agarwal N, Babu SC, et al: Causes of limb loss in civilian arterial 

injuries. J Cardiovasc Surg 1986;27:278-281. 
Shah PM, Ivatury RR, Babu SC, et al: Is limb loss avoidable in civilian vascular 

injuries? Am J Surg 1987;154:202-205. 
Shaker I, White J, Signer R, et al: Special problems of vascular injuries in chil- 
dren. J Trauma 1976;16:863-867. 
Shamoun J, Barraza KR, Jurkovich GJ, Salley R: In extreme use of staples for 

cardiorrhaphy in penetrating cardiac trauma: Case report. J Trauma 

1989;29:1589-1591. 
Shandall A, Leopold P, Shah D, et al: Visceral aortic aneurysm in 4 1/2 year old 

child: An unusual complication of umbilical artery catheterization. Surgery 

1986;100:928-931. 
Shannon J, Vo N, Stanton P: Peripheral arterial missile embolization. J Vase 

Surg 1987;5:773-778. 
ShanthakumarRE,ZwavelingJH, WaterbolkTW, GirbesARJ: Aorto-esophageal 

fistula: An "early" complication following rupture of the thoracic aorta. 

2001. 
Sharar S, Winn R, Murry C, et al: A CD18 monoclonal antibody increases the 

incidence and severity of subcutaneous abscess formation after high-dose 

Staphylococcus aureus injection in rabbits. Surgery 1991;110:213-220. 
Sharma P, Babu SC, Shah PM, Clauss RH: Changing patterns in civilian arter- 
ial injuries. J Cardiovasc Surg 1985;26:7-11. 
Sharma P, Ivatury RR, Simon R, Vinzons A: Central and regional hemodynamics 

determine optimal management of major venous injuries. J Vase Surg 

1992;16:887-894. 
Sharma P, Shah PM, Vinzons A, et al: Meticulously restored lumina of injured 

veins remain patent. Surgery 1992;112:928-932. 



RE.qxd 4/16/04 3:19 PM Page 632 



632 VI • THE RICH REFERENCE COLLECTION 



Shauna R, Main D, PinkertonJ: Surgical therapy of femoral artery pseudoa- 
neurysm after angiography. Am J Surg 1987;154:676-680. 

Sheikh A, Culbertson C: Emergency department thoracotomy in children: Ratio- 
nale for selective application. J Trauma 1993;34:323-328. 

Sheiman R, Brophy D, Perry L, Akbari C: Thrombin injection for the repair of 
brachal artery pseudoaneurysms. AJR Am J Roentgenol 1999;173:1029- 
1030. 

Sheldon G, Lim RC, Yee E, Petersen S: Management of injuries to the porta 
hepatis. Ann Surg 1985;202:539-542. 

Sheldon P, Oglevie S, Kaplan L: Prolonged generalized urticarial reaction after 
percutaneous thrombin injection for treatment of a femoral artery 
pseudoaneurysm.JVIR 2000;11:759-761. 

Shepard G, Rich NM, Dimond F Jr: Punji stick wounds: Experience with 342 
wounds in 324 patients in Vietnam. Ann Surg 1967;166:902-907. 

Sher M: Principles in the management of arterial injuries associated with frac- 
ture/dislocations. Ann Surg 1975;182:630-636. 

Sherman M, Saini V, Yarnox M, et al: Management of penetrating heart wounds. 
Am J Surg 1978;135:553-558. 

Shield CI, Richardson JD, Buckley C, Hagood C: Pseudyaneurysm of the bra- 
chiocephalic arteries: A complication of percutaneous internaljugular vein 
catheterization. Surgery 1975;78:190-194. 

Shih F, Wang S, Dang K, et al: Successful management of traumatic mesenteric 
arteriovenous fistula after failure of steel coil embolization: Case report. 
J Trauma 1994;37:682-686. 

Shin D, Wall MJ Jr, Mattox KL: Combined penetrating injury of the innomi- 
nate artery, left common carotid artery, trachea, and esophagus. J Trauma 
2000;49:780-783. 

Shirkey AL, Quast DC, Jordan GL: Superior mesenteric artery division and intesti- 
nal function. J Trauma 1967;7:7-24. 

Shoemaker WC, Carey J, Yao S: Hemodynamic alterations in acute cardiac tam- 
ponade after penetrating injuries of the heart. Surgery 1970;67:754-764. 

Shoemaker WC, CareyJ, Yao S: Hemodynamic monitoring for physiologic eval- 
uation, diagnosis, and therapy of acute hemopericardial tamponade from 
penetrating wounds. J Trauma 1973;13:36-44. 

Shoenfeld N, Stuchin S, Pearl R, Haveson S: The management of vascular injuries 
associated with total hip arthroplasty. J Vase Surg 1990;11:549-555. 

Shorr R, Crittenden M, Indeck M, et al: Blunt thoracic trauma. Analysis of 515 
patients. Ann Surg 1987;206:200-205. 

Shrock T, Blaisdell FW, Mathewson CJ: Management of blunt trauma to the 
liver and hepatic veins. Arch Surg 1968;96:698-704. 

Shuck J, Trump D: Nonpenetrating abdominal trauma with injuries to blood 
vessels. Am Surg 1961;27:693-697. 

ShuckJ, Omer GJLCJ: Arterial obstruction due to intimal disruption in extrem- 
ity fractures. J Trauma 1972;12:481-489. 

Shumacker HBJr: Incisions in surgery of aneurysm: With special reference to 
explorations in the antecubital and popliteal fossae. Ann Surg 1946;124:586- 
598. 

Shumacker HBJr: Resection of the clavicle with particular reference to the use 
of bone chips in the periosteal bed. Surg Gynecol Obstet 1947;84:245-248. 

Shumacker HBJr: Surgical cure of innominate aneurysm: Report of a case with 
comments on applicability of surgical measures. Surgery 1947;22:729- 
739. 



RE.qxd 4/16/04 3:19 PM Page 633 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 633 

Shumacker HBJr: Operative exposure of the blood vessels in the superior ante- 
rior mediastinum. Am Surg 1948;127:464-475. 

Shumacker HBJr: Problem of maintaining the continuity of artery in surgery 
of aneurysms and arteriovenous fistulae: Notes on the development and 
clinical application of methods of arterial suture. Ann Surg 1948;127:207- 
230. 

Shumacker HBJr: Arterial aneurysms and arterialvenous fistulas. Spontaneous 
problems. Cures in Surgery. In Elkin DC, DeBakey ME (eds): World War 
II, Vascular Surgery. Washington, DC: US Government Printing Office, 
OTSG, Department of the Army, 1955: 361-374. 

Shumacker HBJr: Arterial suture techniques and grafts: Past, present and future. 
Surgery 1969;66:419-433. 

Shumacker HBJr: Book review: Vascular disorders of the upper extremity. Ann 
Surg 1984;199:249. 

Shumacker HBJr: Ramuald Weglowski: Neglected pioneer in vascular surgery. 
J Vase Surg 2001;6:95-97. 

Shumacker HBJr, Carter K: Arteriovenous fistulas and false aneurysms in mil- 
itary personnel. Surgery 1946;20:9-25. 

Shumacker HBJr, Stahl N: A study of the cardiac frontal area in patients with 
arteriovenous fistulas. Surgery 1949;26:928-944. 

Shumacker HBJr, Stokes G: Studies of combined vascular and neurologic injuries. 
Ann Surg 1950;132:386-393. 

Shumacker HB Jr, Wayson E: Spontaneous cure of aneurysms and arteriove- 
nous fistulas, with some notes on intrasaccular thrombosis. Am J Surg 
1950;79:532-544. 

Siavelis H, Marsan R, Marshall W, Maull K: Aortoventricular fistula secondary 
to blunt trauma: A case report and review of the literature. J Trauma 
1997;43:713-715. 

Sibbitt R, Palmaz J, Garcia F, Reuter S: Trauma to the extremities: Prospective 
comparison of digital and conventional angiography. Radiology 
1986;160:179-182. 

SiegelR: Galen on surgery of the pericardium: An early record of therapy based 
on anatomical and experimental studies. Am J Cardiol 1970;26:524-527. 

Siegmeth A, Gaebler C, Sandbach G, Vecsei V: Arare case of a traumatic aneurysm 
of the inferior thyroid artery. J Vase Surg 1995;22:812-813. 

Sigel B, Popky G, Wagner D, et al: Comparison of clinical and Doppler ultra- 
sound evaluation of confirmed lower extremity venous disease. Surgery 
1968;64:332-338. 

Sigler L, Gutierrez-Carreho R, Martinez-Lopez, et al: Aortocaval fistula: Expe- 
rience with five patients. Vase Surg 2001;35:207-212. 

Silen W, Spieker D: Fatal hemorrhage from the innominate artery after tra- 
cheostomy. Ann Surg 1965;162:1005-1012. 

Simeone F, Grillo H, Rundle F: On the question of ligation of the concomitant 
vein when a major artery is interrupted. Surgery 1951;29:932-951. 

Simmons K: Defense against free radicals has therapeutic implications. JAMA 
1984;251:2187-2192. 

Simstein N, Poole GV Jr, Nelson L: Perforation of the intrapericardial aorta 
during subclavian catheterization. South Med J 1984;77:1605. 

Sinclair T, Stephenson HJ: Survival following abdominal aortic rupture from 
blunt trauma: Case report. Mo Med 1972;69:271-272. 

Singh I, Gorman JF: Vascular injuries in closed fractures near junction of middle 
and lower thirds of the tibia. J Trauma 1972;12:592-598. 



RE.qxd 4/16/04 3:19 PM Page 634 



634 VI • THE RICH REFERENCE COLLECTION 



Sinkler W, Spencer A: The importance of early exploration of vascular injuries. 
Surg Gynecol Obstet 1958;107:228-234. 

Sinkler W, Spencer A: The value of peripheral arteriography in assessing acute 
vascular injuries. Arch Surg 1960;80:300-304. 

Sirinek KR, Levine BA: Trauma injury to the proximal superior mesenteric 
vessels. Surgery 1985;98:831-835. 

Sirinek K, Levine B, Gaskill H III, Root H: Reassessment of the role of routine 
operative exploration in vascular trauma. J Trauma 1981;21:339-344. 

Sirinek K, Gaskill H, Root H: Visceral vascular injury: Factors influencing sur- 
vival. J Trauma 1982;22:641. 

Sirinek K, Gaskill H III, Dittman W, Levine B: Exclusion angiography for patients 
with possible vascular injuries of the extremities: A better use of trauma 
center resources. Surgery 1983;94:598-603. 

Sirinek K, Gaskill H III, Root H, Levine B: Truncal vascular injury: Factors influ- 
encing survival. J Trauma 1983;23:372-377. 

SitzmannJ, Ernst CB: Management of arm arterial injuries. Surgery 1984;96:895- 
901. 

SkillmanJ, Ducksoo K, Bain DS: Vascular complications of percutaneous femoral 
cardiac interventions: Incidence and operative repair. Arch Surg 
1988;123:1207-1212. 

Skinner D: Traumatic renal artery thrombosis: A successful thrombectomy and 
revascularization. Ann Surg 1973;177:264-267. 

Skipper R, Debski R: Intramyocardial shotgun pellets diagnosed on initial emer- 
gency room chest x-ray: Case report. J Trauma 1990;30:1609-1610. 

Skyhar M, Hargens A, Strauss M, et al: Hyperbaric oxygen reduces edema and 
necrosis of skeletal muscle in compartment syndromes associated with hem- 
orrhagic hypotension. J Bone Joint Surg 1986;68:1218-1224. 

Slaney G, Ashton F, Abrams LD: Traumatic rupture of the aorta. Br J Surg 
1966;53:361-364. 

Slater R, Edge A, Salman A: Delayed arterial injury after hip replacement. J 
Bone Joint Surg 1989;71:699. 

Sloop R, Robertson K: Non-penetrating traumas of the abdominal aorta due 
to blunt trauma. Ann Surg 1975;182:33-36. 

Sloop R, Robertson K: Nonpenetrating trauma of the abdominal aorta with 
partial vessel occlusion: Report of two cases. Am Surg 1975;41:555-559. 

Smejkal R, Izaznt T, Born C, et al: Pelvic crush injuries with occlusion of the 
iliac artery. J Trauma 1988;28:1479-1482. 

Smith C, Green R: Pediatric vascular injuries. Surgery 1981;90:20-31. 

Smith G, Northrop C: Stab wound causing mesenteric-portal arteriovenous fistula: 
An unusual case with a spontaneous closure. J Trauma 1976;16:408. 

Smith K, Ben-Menachem B, Duke J, Hill G: The superior gluteal: An artery at 
risk in blunt pelvic trauma. J Trauma 1976;16:273-279. 

Smith L, Foran R, Gaspar MR: Acute arterial injuries of the upper extremity. 
Am J Surg 1963;106:144-151. 

Smith M, CassidyJ, Souther S, et al: Transesophageal echocardiography in the 
diagnosis of traumatic rupture of the aorta. N Engl Med J 1995;332:356- 
362. 

Smith P, Lim W, Ferris E, Casali R: Emergency arteriography in extremity trauma: 
Assessment of indications. AJR Am J Roentgenol 1981;137:803-807. 

Smith R, Perdue GJ, Walker LJ, Israel P: Posttraumatic aneurysm of the abdom- 
inal aorta with recurrent emboli of the superior mesenteric artery: A case 
report. Surgery 1968;64:736-742. 



RE.qxd 4/16/04 3:19 PM Page 635 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 635 

Smith RI, Stone H: Traumatic fistulas involving the portal venous system. Am 
JSurgl970;119:570. 

Smith R, Szilagyi D, Pfeifer J: A study of arterial trauma. Arch Surg 1963;86:825- 
835. 

Smith R, Szilagyi D, Elliott J Jr: Fracture of long bones with arterial injury due 
to blunt trauma. Arch Surg 1969;99:315-324. 

Smith R, Elliot J, Hageman J: Acute penetrating arterial injuries of the neck 
and limbs. Arch Surg 1974;109:198-205. 

Smith R, Chang F: Traumatic rupture of the aorta: Still a lethal injury. Am J 
Surg 1986;152:660-663. 

Smith V, Hughes CW, Sapp O, et al: High-output circulatory failure due to arte- 
riovenous fistula: complication of intervertebral-disk surgery. AMA Arch 
IntMed 1957;100:833-841. 

Smith W: Cardiorraphy in acute injuries. Ann Surg 1923;78:696-710. 

Smyth E: Major arterial injury in closed fracture of the neck of the humerus: 
Report of a case. J Bone Joint Surg 1969;51:B:508. 

Snyder WH III: Vascular injuries near the knee: An updated series and overview 
of the problem. Surgery 1982;91:502-506. 

Snyder III WH: Popliteal and shank arterial injury. Surg Clin North Am 
1988;68:787-807. 

Snyder WH III, Thai E, Bridges RA, et al: The validity of normal arteriography 
in penetrating trauma: A review of 114 cases. Arch Surg 1978;113:424-428. 

Snyder III WH, Thai E, Perry MO. Vascular injuries of the extremities. In Vas- 
cular Surgery. In Rutherford R (ed) : Philadelphia, PA: WB Saunders, 1989: 
613-637. 

Snyder WH III, Watkins W, Whiddon L: Civilian popliteal artery trauma: An 1 1- 
year experience with 83 injuries. Surgery 1979;85:101-108. 

Soldano S, Rich NM, Collins GJ, et al: Long-term follow-up of penetrating abdom- 
inal aortic injuries after 15 years. J Trauma 1988;28:1358-1362. 

Solovei G, Alame A, Bardoux JL: Paraplegia and dissection of the abdominal 
aorta after closed trauma: A proposal of a case. Current review of the lit- 
erature (1982-1993) [inFrench] JChirurgie 1994;131:236-244. 

Sondenaa K, Tveit B, Kordt K, et al: Traumatic rupture of the thoracic aorta: 
A clinicopathological study. Acta Chir Scand 1990;156:137-143. 

Soots G, Warembourg HJ, Pratt A, Roux J: Acute traumatic rupture of the tho- 
racic aorta: Place of delayed surgical repair.J Cardiovasc Surg 1989;30:173- 
177. 

Sotta R: Vascular problems in the proximal upper extremity. Clin Sports Med 
1990;9:379. 

Soubbotitch V: Military experiences of traumatic aneurysms. Lancet 1913;2:720- 
721. 

Soyer R, Brunet A, Piwnica A, et al: Traumatic rupture of the thoracic aorta 
with reference to 34 operated cases. J Cardiovasc Surg 1981;22:103-108. 

Soyer R, Bessou JP, Bouchart F, et al: Long-term results of surgical treatment 
of traumatic rupture of the thoracic aorta: 47 cases. J Cardiovasc Surg 
1990;31(suppl):102. 

Speechly-Dick M, Grover G, Yellon D: Does ischemic preconditioning in the 
human involve protein kinase C and the ATP dependent K+ channel? Circ 
Res 1995;77:1030-1035. 

Spellman M, Mandal A, Freeman HP: Successful repair of an arteriovenous 
fistula between the superior mesenteric vessels secondary to a gunshot 
wound. Ann Surg 1967;165:458. 



RE.qxd 4/16/04 3:19 PM Page 636 



636 VI • THE RICH REFERENCE COLLECTION 



Spence R, DelRossi AJ, CilleyJ, Civil I: Exsanguinating upper extremity vascu- 
lar injury: Is an initial approach by clavicular resection adequate? J Car- 
diovasc Surg 1989;30:450. 

Spencer A: The reliability of signs of peripheral vascular injury. Surg Gynec 
Obstet 1962;114:490-494. 

Spencer FC: The use of optical magnification in coronary bypass grafting. 
J Thorac Cardiovasc Surg 1970. 

Spencer FC, Grewe R: The management of acute arterial injuries in battle casu- 
alties. Ann Surg 1955;141:304-313. 

Spencer FC, Tompkins R: Management of acute arterial injuries. Postgrad Med 
1960;28:476-481. 

Spencer FC, Guerin P, Blake HA, Bahnson HT: A report of fifteen patients with 
traumatic rupture of the thoracic aorta. J Thorac Cardiovasc Surg 
1961;41:1-22. 

Spirnak J, Resnick M: Revascularization of traumatic thrombosis of the renal 
artery. Surg Gynec Obstet 1987;164:22-26. 

Sponge A, Borrows PE, Armstrong D, Daneman A: Traumatic aortic rupture 
in the pediatric population: Role of plain film, CT, and angiography in the 
diagnosis. Pediatr Radiat 1991;21:324-328. 

Stables D, Fouche R, de Villiers van Niekerek J, et al: Traumatic renal artery 
occlusion: 21 cases. J Urol 1976;115:229. 

Stafford G, O'Brien M: Traumatic rupture of the thoracic aorta. AustNZJ Surg 
1977;47:175-179. 

Stain SC, Yellin AE, Weaver FA, Pentecost M: Selective management of nonoc- 
clusive arterial injuries. Arch Surg 1989;124:1136-1141. 

Stain SC, Weaver FA, Yellin AE: Extra-anatomic bypass of failed traumatic arte- 
rial repairs. J Trauma 1991;31:575-578. 

Starnes BW, O'Donnell SD, Gillespie DL, et al: Endovascular management of 
renal ischemia in a patient with acute aortic dissection and renovascular 
hypertension. Ann Vase Surg 2002. 

Starzl T, Kaupp HJ, Beheler EM: The treatment of penetrating wounds to the 
inferior vena cava. Surgery 1962;51:195-204. 

StavensB, Hasim S, Hammond G, et al: Optimal methods of repair of descend- 
ing thoracic aortic transections and aneurysms. Am J Surg 1983;145:508- 
513. 

Steenburg R, Ravitch MM: Cervico-thoracic approach for subclavian vessel injury 
from compound fracture of the clavicle: Consideration of subclavian- 
axillary exposure. Ann Surg 1963;15j7:839-846. 

Steichen F, Dargan E, Efron G, et al: A graded approach to the management 
of penetrating wounds of the heart. Arch Surg 1971;103:574-580. 

Stein AJ: Arterial injury in orthopaedic surgery. J Bone Joint Surg 1956;38:669. 

Stein L, Shubin H, Weil M: Recognition in management of pericardial 
tamponade. JAMA 1973;225:503. 

Stein M, Mirvis S, Wiles CI: Delayed embolization of a shotgun pellet from the 
chest to the middle cerebral artery. J Trauma 1995;39:1006-1009. 

Stein R, Bono J, Korn J, Wolff W: Axillary artery injury in closed fracture of the 
neck of the scapular: A case report. J Trauma 1971;11:528-531. 

Steinbauer M, Harris A, Leiderer R, et al: Impact of dextran on microvascular 
disturbances and tissue injury following ischemia/reperfusion in striated 
muscle. Shock 1998;9:345-351. 

Stelzner V, Horatz K: Correction of gunshot wounds of extrapericardial ascend- 
ing aorta. Thoraxchirurgie 1963;10:632. 



RE.qxd 4/16/04 3:19 PM Page 637 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 637 

Stemmer E, Oliver C, CareyJ, Connolly JE: Fatal complications of tracheotomy. 

Am J Surg 1976;131:288-290. 
Stewart F: Arteriovenous aneurysm treated by angiography (angiorrhaphy). 

Abb Surg 1913;57:247-254. 
Stich R: Ueber gefaess und organ transplantationen mittelst gefaessnaht. Ergeon 

ChirOrth 1910;1:1. 
Stiles Q, Cohlmia G, Smith J, et al: Management of injuries of the thoracic and 

abdominal aorta. Am J Surg 1985;150:132-140. 
Stinnett D, Graham JM, Edwards W: Fibromuscular dysplasia and thrombosed 

aneurysm of the popliteal artery in a child. J Vase Surg 1987;5:769-772. 
Stipa S, Vollmar J: Arterial Trauma. Rome, Italy, Ares-Serono Symposia, 1986. 
Stokes J, McAfee C: Increasing limb survival in vascular injury with fracture. 

J Trauma 1965;5:162. 
Stollone R, Ecker R, Samson P: Management of major acute thoracic vascular 

injuries. Am J Surg 1974;128:249-254. 
Stone H, Oxford W, Austin JT: Penetrating wounds of the abdominal aorta. 

South Med J 1973;66:1351-1355. 
Stone H, Fabian TC, Turkleson M: Wounds of the portal venous system. World 

J Surg 1982;6:335-341. 
Stone K, Walshaw R, Sugiyama G, et al: Polytetrafluoroethylene versus autoge- 
nous vein grafts for vascular reconstruction in contaminated wounds. Am 

J Surg 1984;147:692-695. 
Stone W: Observations on the treatment of wounded arteries, with cases. New 

Orleans Med SurgJ 1857;2:168. 
Stoney R, Roe B, RedingtonJ: Rupture of the thoracic aorta due to closed chest 

trauma. Arch Surg 1964;89:840-847. 
Storey C, Nardi G, Sewell W: Traumatic aneurysms of the thoracic aorta: Report 

of two cases, one successfully treated by resection and graft replacement 

with the aid of a shunt. Ann Surg 1956;144:69-78. 
Stothert JJ, McBride L, Tidik S, et al: Multiple aortic tears treated by primary 

suture repair. J Trauma 1987;27:955-956. 
Strandness DE Jr: Collateral Circulation in Clinical Surgery. Philadelphia, WB 

Saunders, 1969. 
Strandness DE Jr, Bell JW: Peripheral vascular disease: Diagnosis and objective 

evaluation using a mercury strain gauge. Ann Surg 1965;161 (suppl 1):3- 

35. 
Strassmann G: Traumatic rupture of the aorta. Am Heart J 1947;33:508-515. 
Straus R: Pulmonary embolism caused by a lead bullet following a gunshot 

wound of the abdomen. Arch Pathol 1942;33:63-68. 
Stringer W, Kelly DJ: Traumatic dissection of the extracranial internal carotid 

artery. Neurosurgery 1980;6:123-130. 
Strodel W, Eckhauser F, Lemmer J: Presentation and perioperative manage- 
ment of arterioportal fistulas. Arch Surg 1987;122:563. 
Stromberg B: Symptomatic lead toxicity secondary to retained shotgun pellets: 

Case report. J Trauma 1990;30:356-357. 
Stubbs D, Dorner D, Johnson R: Thrombosis of the iliofemoral artery during 

revision of a total hip replacement: A case report. J Bone Joint Surg [Am] 

1986;68:454-455. 
Sturm J, Strate R, Mowlem A, et al: Blunt trauma to the subclavian artery. Surg 

Gynec Obstet 1974;138:915-918. 
Sturm J, Perry JJ, Cass AS: Renal artery and vein injury following blunt trauma. 

Ann Surg 1975;182:696. 



RE.qxd 4/16/04 3:19 PM Page 638 



638 VI • THE RICH REFERENCE COLLECTION 



Sturm J, Marsh D, Bodily KC: Ruptured thoracic aorta: Evolving radiological 
concepts. Surgery 1979;85:363-367. 

Sturm J, Bodily KC, Rothenberger D, Perry JF: Arterial injuries of the extrem- 
ities following blunt trauma. J Trauma 1980;20:933-936. 

Sturm J, Cicero J: The clinical diagnosis of ruptured subclavian artery follow- 
ing blunt thoracic trauma. Ann Emerg Med 1983;12:17-19. 

Sturm J, Perry JFJr, Olson F, Cicero J: Significance of symptoms and signs in 
patients with traumatic aortic rupture. Ann Emerg Med 1984;13:876- 
878. 

Sturm J, Billiar TR, DorseyJ, et al: Risk factors for survival following treatment 
of traumatic aortic rupture. Ann Thorac Surg 1985;39:418-421. 

Sturm J, Perry JJ: Brachial plexus injuries form blunt thoracic trauma: A 
harbinger of vascular and thoracic injury. Ann Emerg Med 1987; 16:404- 
406. 

Sturm J, Billiar TR, Luxenberg M, Perry JF: Risk factors for the development 
of renal failure following the surgical treatment of traumatic aortic rupture. 
Ann Thorac Surg 1987;43:425-427. 

Sturm J, McGee M, Luxenberg M: An analysis of risk factors for death at the 
scene following traumatic aortic rupture. J Trauma 1988;28:1578-1580. 

Suarez C, Bernstein DM, Goldberger J, et al: The role of nuclear scanning in 
the evaluation of arterial injuries. Am Surg 1983;49:209-210. 

Sugg S, Gerndt S, Hamilton B, et al: Pseudoaneurysms of the intraparenchy- 
mal splenic artery after blunt abdominal trauma: A complication of non- 
operative therapy and its management. J Trauma 1995;39:593-595. 

Sugg W, Rea W, Ecker R: Penetrating wounds of the heart: An analysis of 459 
cases. J Thorac Cardiovasc Surg 1986;56:531-545. 

Sullivan M: Rupture of the brachial artery from posterior discoloration of the 
elbow treated by vein graft. Br J Surg 1971;58:470-471. 

Sullivan M, Smalley R, Banowsky LH: Renal artery occlusion secondary to blunt 
abdominal trauma. J Trauma 1972;12:509-515. 

Sullivan W, Thorton F, Baker LH, et al: Early influence of popliteal vein repair 
in the treatment of popliteal vessel injuries. Am J Surg 1971;122:528-531. 

Summerall C, Lee WJ, Boone JA: Intracardiac shunts after penetrating wounds 
of the heart. N Engl J Med 1965;272:240-242. 

Sumpio G, Gusberg R: Aortic thrombosis with paraplegia: An unusual conse- 
quence of blunt abdominal trauma. J Vase Surg 1987;6:412-414. 

Sutorius D, Schreiber J, HelmsworthJ: Traumatic disruption of the thoracic 
aorta. J Trauma 1973;13:583-590. 

Svane H, Ottosen T: Traumatic vascular lesions. J Cardiovasc Surg 1963;4:303- 
305. 

Svensson L, Antunes MDJ, Kinsley R: Traumatic rupture of the thoracic aorta: 
A report of 14 cases and a review of the literature. S Afr Med J 1985;67:853- 
857. 

Swan KG, HobsonRWII, Reynolds D, etal: Venous surgery in the lower extrem- 
ities. In Swan KG, Hobson R, Reynolds D, et al (eds): St Louis, Warren H 
Green Publishers, 1975. 

Sweetman W: Subclavian steal syndrome following trauma: Case report. Am 
Surg 1965;31:463-466. 

Swetnam J, Hardin WD, Kerstein MD: Successful management of trifurcation 
injuries. Am Surg 1986;52:585-587. 

Symbas P, Vlasis-Hale S, Picone A: Missiles in the heart. Ann Thorac Surg 
1989;48:192-194. 



RE.qxd 4/16/04 3:19 PM Page 639 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 639 

Symbas P, Pourhamidi A, Levin J: Traumatic rupture of the aortic arch between 

the left common carotid and left subclavian arteries and avulsion of the 

left subclavian artery. Ann Surg 1969;170:152-156. 
Symbas P, Sehdeva J, Abbott OA, et al: Penetrating wounds of the thoracic aorta 

and great arteries. South Med J 1970;63:853-857. 
Symbas P, Sehdeva J: Penetrating wounds of the thoracic aorta. Ann Surg 

1970;171:441-450. 
Symbas P, Tyras D, Ware R, Hatcher CJ: Rupture of the aorta. Ann Thorac Surg 

1973;15:405-410. 
Symbas P, Tyras D, Ware R, Diroio D: Traumatic rupture of the aorta. Ann Surg 

1973;178:6-12. 
Symbas P, Kourias E, Tyras D, Hatcher CJ: Penetrating wounds of great vessels. 

Ann Surg 1974;179:757-762. 
Symbas P, Harlaftis N, Waldo W: Penetrating cardiac wounds: A comparison of 

different therapeutic methods. Ann Surg 1976;183:377-381. 
Symbas P, Harlaftis N: Bullet emboli in the pulmonary and systemic arteries. 

Am Surg 1977;185:318-320. 
SymesJ, Eadie D, Maclean A: Traumatic aorto-caval fistula associated with horse- 
shoe kidney. J Trauma 1974;14:402-408. 
Symonds F, Games A, Porter V, Crikelair G: Pitfalls in the management of 

penetrating injuries of the forearm. J Trauma 1971;11:47-52. 
Synder C, Eyer S: Blunt chest trauma with transection of the azygos vein: Case 

report. J Trauma 1989;29:889-890. 
Synder S, Wheeler J, Gregory R, etal: Freshly harvested cadaveric venous homo- 
grafts as arterial conduits in infected fields. Surgery 1987;101:283-291. 
Syracuse D, Seaver PRJr, Amato JJ: Aortic gunshot injury and paraplegia: Pre- 
operative definition with arteriography and computerized axial tomogra- 
phy. J Trauma 1985;25:271-273. 
Szentpetery S, Lower R: Changing concepts in the treatment of penetrating 

cardiac injuries. J Trauma 1977;17:457-461. 
Szentpetery S: Management of aortic cannulation site blowouts. Ann Thorac 

Surg 1992;54:582-583. 
Szilagyi D: Discussion on "Choice of vascular graft material for patients." In 

Wesolowski S, Dennis C (eds): Fundamentals of Vascular Grafting. New 

York, McGraw-Hill, 1963, pp 397-419. 
Szilagyi D: Spinal cord damage in surgery of the abdominal aorta. Surgery 

1978;83:38-56. 
Szuchmacher P, Freed J: Immediate revascularization of the popliteal artery 

and vein: Report of a case. J Trauma 1978;18:142-144. 
Tailor A, Granger D: Role of adhesion molecules in vascular regulation and 

damage. Curr Hyperten Rep 2000;2:78-83. 
Takeuchi M, Maruyama K, Nakamura M, et al: Posttraumatic inferior vena caval 

thrombosis: Case report and review of the literature.J Trauma 1995;39:605- 

608. 
Tamim W, Arbid EJ, Andrews LS, Arous EJ: Percutaneous induced thrombosis 

of iatrogenic femoral pseudoaneurysms following catheterization. Ann Vase 

Surg 2000;14:254-259. 
Tarlow S, Achterman CA, Hayhurst J, Ovadia D: Acute compartment syndrome 

in the thigh complicating fracture of the femur: A report of three cases. 

J Bone Joint Surg 1986;68:1439-1443. 
Tassi A, DaviesA: Pericardial tamponade due to penetrating fragment wounds 

of the heart. Am J Surg 1969;118:535-538. 



RE.qxd 4/16/04 3:19 PM Page 640 



640 VI • THE RICH REFERENCE COLLECTION 



Tawes RJ, Etheredge S, Webb R, et al: Popliteal artery injury complicating arthro- 
scopic meniscectomy. Am J Surg 1988;156:136-138. 

Taylor B, Rhee R, Muluk S: Thrombin injection versus compression of femoral 
artery pseudoaneurysms. J Vase Surg 1999;30:1052-1059. 

Taylor LJ, Troutman R, Feliciano P, et al: Late complications after femoral artery 
catheterization in children less than five years of age. J Vase Surg 
1990;11:297-306. 

Taylor TKF, Wardill JC: Successful primary repair of rupture of the popliteal 
artery in association with compound dislocation of the knee-joint. Br Surg 
1964;51:163-166. 

Tector A, Reuben C, Hoffman J, et al: Coronary artery wounds treated with 
saphenous vein bypass grafts. JAMA 1973;225:282-284. 

Tector A, Worman L, Romer J, et al: Unusual injury to the aortic arch: A case 
report. J Thorac Cardiovasc Surg 1974;67:547-552. 

Tegner Y, Bergdahl L, Ekestrom S: Traumatic disruption of the thoracic aorta. 
Acta Chir Scand 1984;150:635-638. 

TepasJ, Alexander RH, Snyder J: Real time ultrasonography as a diagnostic 
screen in experimental arterial injury. J Trauma 1985;25:720. 

Thai E, Perry MO, Crighton J: Traumatic abdominal aortic occlusion. South 
MedJ 1971;64:653-656. 

ThalE, Snyder WH III, Hays R, Perry MO: Management of carotid artery injuries. 
Surgery 1974;76:955-962. 

Theodorides T, de Keizer G: Injuries of the axillary artery caused by fractures 
of the neck of the humerus. Injury 1976;8:120-123. 

Theunis P, Coenen L, BrouwersJ: Traumatic injuries to the porta hepatis. Injury 
1989;20:152. 

Thevenet A, DuCailiar C: Chronic traumatic aneurysms of the thoracic aorta. 
World J Surg 1989;13:112-116. 

Thio R: False Aneurysm of the ulnar artery after surgery employing a tourni- 
quet. Am J Surg 1972;123:604-605. 

Thio R, Stanton P, Logan W: Simultaneous avulsion of the innominate and left 
intrathoracic subclavian arteries. J Thorac Cardiovasc Surg 1973;66:96- 
98. 

Thomas A, Goodman P, Roon A: Role of angiography in cervicothoracic trauma. 
J Thorac Cardiovasc Surg 1978;76:633-638. 

Thomas CJ, Carter J, Lowder S: Pericardial tamponade from central venous 
catheters. Arch Surg 1969;98:217-218. 

Thomas D, Wilson R, Wieneck R: Vascular Injury about the knee: Improved 
outcome. Ann Surg 1989;55:370-377. 

Thomas T: Management of cardiac and intrathoracic great vessel injuries. Surg 
Gynecol Obstet 1967;125:997-1002. 

Thomford N, Curtiss P, Marable S: Injuries of the iliac and femoral arteries asso- 
ciated with blunt skeletal trauma. J Trauma 1969;9:126-134. 

Thomford N, Pace W, Meckstroth C: Traumatic rupture of the thoracic artery. 
Am Surg 1969;35:244-249. 

Thorlacius H, Vollmar B, Westermann S, et al: Effects of local cooling on microvas- 
cular hemodynamics and leukocyte adhesion in the striated muscle of ham- 
sters. J Trauma 1998;45:715-719. 

Tilney N, McLamb J: Leg trauma with posterior tibial artery tear. J Trauma 
1967;7:807. 

Timberlake G, O'Connell RC, Kerstein MD: Venous Injuries, to repair or ligate, 
the dilemma. J Vase Surg 1986;4:533-538. 



RE.qxd 4/16/04 3:19 PM Page 641 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 641 

Timberlake G, Adinolfi MF, McSwain N Jr: Problem: Rupture of the thoracic 

aorta. Emerg Med 1987;19:45-49. 
Timberlake G, Kerstein MD, McSwain N Jr: Penetrating thoracic aortic injuries: 

Rare but potentially salvageable sequelae of urban warfare. South Med J 

1989;82:970-972. 
Timberlake G, Kerstein MD: Venous injury: To repair or litigate, the dilemma 

revisited. Am Surgeon 1995;61:139-145. 
Tipton W, D 'Ambrosia R: Vascular Impairment as a result of fracture- 
dislocation of the ankle. J Trauma 1975;15:524-527. 
Tisnado J, Tsai F, Als A, Roach J: A new radiographic sign of acute traumatic 

rupture of the thoracic aorta: Displacement of the nasogastric tube to the 

right. Radiology 1977;125:603-608. 
Todd G, Bregman D, Voorhees A, Reemtsma K: Vascular Complications asso- 
ciated with percutaneous intra-aortic balloon pumping. ArchSurg 

1983;118:963-964. 
Toguri A, Liu T, Bayliss C: Traumatic bilateral renal artery thrombosis. J Urol 

1974;112:430. 
Toivio I, Karlsson B: Treatment of ischemia of extremity caused by a shotgun 

blast. Ann Med Milit Fenn 1975;4:159. 
Tomatis L, Doornobs F, Beard JA: Circumferential intimal tear of the aorta with 

complete occlusion due to blunt trauma. J Trauma 1968;8:1096-1101. 
Tomiak M, Rosenblum J, Messersmith R, Zarins C: Use of CT for diagnosis of 

traumatic rupture of the thoracic aorta. Ann Vase Surg 1993;7:130- 

139. 
Toursarkissian B, Allen BT, Petrinec D: Spontaneous closure of selected iatro- 
genic pseudoaneurysms and arteriovenous fistulae. J Vase Surg 1997;25:803- 

809. 
Towne JB, Delbert D, Smith J: Thrombosis of the internal carotid artery fol- 
lowing blunt cervical trauma. Arch Surg 1972;104:565-568. 
Townsend R, Colella J, Diamond D: Traumatic rupture of the aorta: Critical 

decisions for trauma surgeons. J Trauma 1990;30:1169-1174. 
Traber D: Nitric oxide synthase and tissue injury. Shock 2000;14:243-244. 
Trask A, Richards F, Schwartzbach C, Kurtzke R: Massive orthopedic, vascular, 

and soft tissue wounds from military type assault weapons: A case report. 

J Trauma 1995;30:1169-1174. 
Travers B, Cooper A: On wounds and ligature of veins. Surgical Essays (Lond) 

1818;1:243. 
Treiman G, Yellin AE, Weaver FA, et al: Examination of a patient with a knee 

dislocation: The case for selective angiography. Arch Surg 1992;127:1056- 

1063. 
Treiman R, Doty D, Caspar MR: Acute vascular trauma: A fifteen year study. 

Am Surg 1966;111:469-473. 
Trent M, Parsonnet V, Shoenfeld R: A balloon expandable-intravascular stent 

for obliterating aortic dissection. J Vase Surg 1990;11:707-717. 
Tribble C, Crosby I: Traumatic rupture of the thoracic aorta. South Med J 

1988;81:963-968. 
Trimble C: Arterial bullet embolism following thoracic gunshot wounds. Ann 

Surg 1968;168:911-916. 
Trinkle JK, Marcos J, Grover FL, Cuello L: Management of the wounded heart. 

Ann Thorac Surg 1974;38:181-182. 
Trinkle JK, Toon R, Franz J, et al: Affairs of the wounded heart: Penetrating 

cardiac wounds. J Trauma 1979;19:467-472. 



RE.qxd 4/16/04 3:19 PM Page 642 



642 VI • THE RICH REFERENCE COLLECTION 

Trinkle JK: Penetrating heart wounds: Difficulty in evaluating clinical series 
(editorial) . Ann Thorac Surg 1984;38:181-182. 

Trueblood H, Wuerflein R, Angell WW: Blunt trauma rupture of the heart. 
Ann Surg 1973;177:66-69. 

Trunkey DD,BlaisdellFW: Abdominal vascular injuries. WestJ Med 1975;123:321- 
324. 

Trunkey DD: Trauma: Accidental and intentional injuries account for more 
years of life loss in the U.S. than cancer and heart disease. Among the pre- 
scribed remedies are improved prevention efforts, speedier surgery and 
further research. Sci Am 1983;249:28-35. 

Tse D, Slabaugh P, Carlson P: Injury to the axillary artery by a closed fracture 
of the clavicle. J Bone Joint Surg 1980;62:1372-1373. 

Tuffier M: Contemporary French surgery. Br J Surg 1915;3:100-112. 

Turner G: Gunshotwounds of the heart. War Medicine, a Symposium. In Pingh 
WS (ed): Podolsky Edward, 1942. 

Turney SZ, Attar S, Ayella R, et al: Traumatic rupture of the aorta: A five-year 
experience. J Thorac Cardiovasc Surg 1976;72:727-734. 

Turney SZ, Attar S, Ayella R, et al: Is arteriography necessary in the manage- 
ment of vascular trauma of the extremities? Surgery 1978;84:557-562. 

Turney SZ, Rodriguez A, Attar S: Repair of traumatic thoracic aortic rupture: 
Risk factors of paraplegia. J Cardiovasc Surg 1990;31:107. 

Turpin I, State D, Schwartz A: Injuries to the inferior vena cava and their man- 
agement. Am J Surg 1977;134:25-34. 

Tuzeo S, Saad S, Hastings O, Swan KG: Management of brachial artery injuries. 
Surg Gynec Obstet 1978;146:21-24. 

TyburskiJG, Wilson RF, Dente C, et al: Factors affecting mortality rates in patients 
with abdominal vascular injuries. J Trauma 2001;50:1020-1026. 

Uhlmann D, Uhlmann S, Spiegel H: Endothelin/nitric oxide balance influ- 
ences hepatic ischemia-reperfusion injury. J Cardiovasc Pharmacol 
2000;36:S212-S214. 

Ulflacker R, SaadiJ: Transcatheter embolization of superior mesenteric arte- 
riovenous fistula. AJR Am J Roentgenol 1982;139:1212. 

Ulvestad L: Repair of laceration of superior mesenteric artery acquired by non- 
penetrating injury to the abdomen. Ann Surg 1954;140:752-754. 

UtleyJ, Singer M, Roe B, et al: Definitive management of innominate artery 
hemorrhage complicating tracheostomy. JAMA 1972;220:577-579. 

Valle A: War injuries of heart and mediastinum. Arch Surg 1955;70:398- 
404. 

Van De Wal H, Draaisma J, Vincent J, Goris R: Rupture of the supradiaphrag- 
matic inferior vena cava by blunt decelerating trauma: Case report.J Trauma 
1990;30:111-113. 

Van Gilder J, Coxe W: Shotgun Pellet embolus of the middle cerebral artery. 
Neurosurgery 1970;32:711-714. 

van Leeuwen J, Broos P, Rommens P, Nevelsteen A: Combined blunt rupture 
of the diaphragm and the thoracic artery. Injury 1990;21:117-119. 

Van NiekerkJ, Heijstraten F, Goris R, et al: Spinal cord injury following surgery 
for acute traumatic rupture of the thoracic aorta. Thorac Cardiovasc Surg 
1986;34:30-34. 

Van Way C: Intrathoracic and intravascular migratory foreign bodies. Surg Clin 
North Am 1989;69:125-133. 

Varner J, Oliver R: Superior mesenteric arteriovenous fistula. Ann Surg 
1969;170:862. 



RE.qxd 4/16/04 3:19PM PageS43 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 643 

VaskoJ, Raess D, Williams TEJr: Non-penetrating trauma to the thoracic aorta. 

Surgery 1977;82:400-406. 
Vaughan G, Mattox KL, Feliciano DV, et al: Surgical experience with expanded 

polytetrafluorethylene (PTFE) as a replacement graft for traumatized vessels. 

J Trauma 1979;19:403-408. 
VautheyJ, Maddern G, Balsiger D, et al: Superselective embolization of supe- 
rior gluteal artery pseudoaneurysms following intramuscular injection: Case 

report. J Trauma 1991;31:1174. 
Veith F, Gupta S, Daly V: Technique for occluding the supraceliac aorta through 

the abdomen. Surg Gynec Obstet 1980;151:426-428. 
Veith F, Ascer E, Gupta S, Wengerter K: Lateral approach to the popliteal artery. 

JVascSurgl987;6:119. 
Verdant A, Mercier C, Page A, et al: Major mediastinal vascular injuries. Can J 

Surg 1983;26:38-42. 
Verdant A, CossetteR, DontignyL, etal: Acute and chronic traumatic aneurysms 

of the descending thoracic aorta: A 10-year experience with a single method 

of aortic shunting. J Trauma 1985;25:601-607. 
Verdant A: Major mediastinal vessel injury: An underestimated lesion. Can J 

Surg 1987;30:402-404. 
Verdant A: Traumatic rupture of the thoracic aorta. Ann Thorac Surg 

1990;49:686-687. 
Veterans Administration Cooperative Study Group 141: Comparative 

evaluation of prosthetic, reversed, and in situ vein bypass grafts in 

distal popliteal and tibioperoneal revascularization. Arch Surg 1988;123:434- 

438. 
Vignon P, Gueret P, Vedrinne J, et al: Role to transesophageal echocardiogra- 
phy in the diagnosis and management of traumatic aortic disruption. Cir- 
culation 1995;92:2959-2968. 
Vrj D, Simoni E, Smith R: Resuscitative thoracotomy for patients with traumatic 

injury. Surgery 1983;94:554-561. 
Vincent K, Blair WF: A false aneurysm of the ulnar artery after a forearm com- 
partment syndrome. Orthopedics 1984;7:1834-1836. 
Visser P, Hermreck A, Pierce G, et al: Prognosis of nerve injuries incurred during 

acute trauma to peripheral arteries. Am J Surg 1980;140:596-599. 
Vitale G, Richardson JD, George SMJr, Miller F: Fasciotomy for severe, blunt, 

and penetrating trauma of the extremity. Surg Gynecol Obstet 1988; 166:397- 

401. 
Vitelli C, Scalea T, Phillips T, et al: A technique for controlling injuries of the 

iliac vein in the patient with trauma. Surg Gynecol Obstet 1988;166:551- 

552. 
Vollmar J, Krumhaar D: Surgical experience with 200 traumatic arteriovenous 

fistulae. In Hiertonn T, Rybeck B (eds) : Traumatic Arterial Lesions. 

Stockholm, Forsvarets Forskningsanstalt, 1968. 
Vollmar J: Surgical experience with 197 traumatic arterial lesions (1953-66). 

In Hiertonn T, Rybeck B (eds): Traumatic Arterial Lesions. Stockholm, 

Forsvarets Forskningsanstalt, 1968. 
Vollmar J: Knochenbruch und Gefassverletzung. Langenbecks Arch Chir 

2001;339:473-477. 
von Horoch C: Die Gefassnaht. Allg Wien Med Ztg 1888;33:263-279. 
von Oppell U, Beningfield SJ, Odel J, Reichart B: Cardiac rupture caused by 

blunt trauma as well as false angiographic aortic rupture: A case report. S 

Afr Med J 1988;74:519-520. 



RE.qxd 4/16/04 3:19 PM Page 644 



644 VI • THE RICH REFERENCE COLLECTION 

von Oppell U, Dunne T, DeGroot M, Zilla P: Traumatic aortic rupture: Twenty 

year metaanalysis of mortality and risk of paraplegia. Ann Thorac Surg 

1994;58:585-593. 
Von Zoege-Manteuffel: Demonstration eines Praparates von aneurysma arte- 

riosovenosum ossificans der art femoralis profunda. Verhandl Deutsch 

Gesellsch Chir 1895;24:167-170. 
Vorwerk D, Gtinther R, Wendt G: Chronic aortoiliac dissection treated by 

self-expanding stent placement. Cardiovasc Intervent Radiol 1995;18: 

43. 
Vosloo S, Reichart B: Inflow occlusion in the surgical management of a pene- 
trating aortic arch injury: Case report. J Trauma 1990;30:514-515. 
Waddell J, Lenczer E: Arterial injury associated with skeletal trauma. Injury 

1974;6:28-32. 
Waddell W, Vogelfanger I, Prudhomme P, et al: Venous valve transplantation. 

Arch Surg 1964;88:5-15. 
Wagner W, Calkins E, Weaver FA: Blunt popliteal artery trauma: 100 consecu- 
tive cases injuries. J Vase Surg 1988;7:736-748. 
Wagner W, Calkins E, Weaver FA, et al: Blunt popliteal artery trauma: One 

hundred consecutive injures. J Vase Surg 1988;5:736-743. 
Wagner W, Yellin AE, Weaver FA, et al: Acute treatment of penetrating popliteal 

artery trauma: The importance of soft tissue injury. Ann Vase Surg 

1994;8:557-565. 
Waibel P, Ludin H: Unusual sources for peripheral arterial embolization: A case 

report. Arch Surg 1966;92:105-106. 
WaigandJ, Uhlich F, Gross C: Percutaneous treatment of pseudoaneurysms and 

arteriovenous fistulas after invasive vascular procedures. Catheter Cardio- 
vasc Interv 1999;47:157-164. 
Walden R, Lynn M, Golan M, Garniek A: Plastic bullet arterial embolization 

following gunshot injury to the heart: Case report and review of the liter- 

ature.J Cardiovasc Surg 1990;31:482-485. 
Walker A, Walker R: Traumatic thrombosis of the aorta. Br Med J 1961;5238: 

1514. 
Walker M, PoindexterJ, Stovall I: Principles of management of shotgun wounds. 

Surg Gynecol Obstet 1990;170:97-106. 
Walker P, Dake M, Mitchell R, Miller D: The use of endovascular techniques 

for the treatment of complications of aortic dissection. J Vase Surg 

1993;18:1042-1051. 
Walker T, Geller S, Brewster DC: Transcatheter occlusion of a profunda femoral 

artery pseudoaneurysm using thrombin. AJRAmJ Roentgenol 1987;149:185- 

186. 
Walker W, Pate J: Medical management of acute traumatic rupture of the aorta. 

Ann Thorac Surg 1990;50:965-967. 
Walker W, Cooley DA, Duncan J, et al: The management of aortoduodenal 

fistula by in situ replacement of the infected abdominal aortic graft. Ann 

Surg 1987;205:727-732. 
Wall MJ Jr, Pape PE, Martin RR, et al: Immediate versus delayed fluid resusci- 
tation for hypotensive patients with penetrating torso injuries. N Engl J 

Med 1994;331:1105-1109. 
Wall MJJr, Granchi T, Liscum K, Mattox KL: Penetrating thoracic vascular injuries. 

Surg Clin North Am 1996;76:749-761. 
Wallenhaupt S, Huspeth A, Mills S, et al: Current treatment of traumatic aortic 

disruptions. Am Surg 1989;55:316-320. 



RE.qxd 4/16/04 3:19PM PageS45 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 645 

Waller B, Girod D, DillionJ: Transverse aortic wall in infants after balloon angio- 
plasty for aortic valve stenosis: Relation of aortic wall damage to diameter 
of inflated angioplasty balloon and aortic lumen in seven necropsy cases. 
J Am Coll Cardio 1984;4:1235-1241. 

Walls J, Curtis J, Boley T: Sarns centrifugal pump for repair of thoracic aortic 
injury: Case report. J Trauma 1989;29:1283-1285. 

Walls J, Boley TM, Curtis J, Schmaltz R: Experience with four surgical tech- 
niques to repair traumatic aortic pseudoaneurysm. J Thorac Cardiovasc 
Surg 1993;106:283-287. 

Walsh J, Meyer JR Rosenbloom M: Production and treatment of mechanically 
induced arterial vasospasm in a rabbit model. Surg Fo 1987;38:313-314. 

Wand J, Zuckerman J: Delayed rupture of false aneurysm following a femoral 
fracture. J Bone Joint Surg 1989;7lB:700. 

Wang N, Sparks S, Bailey LL: Staged repair using omentum for posttraumatic 
aortoesophageal fistula. Ann Thorac Surg 1994;58:557-559. 

Wang P, Singh G, Rana M, et al: Preheparinization improves organ function 
after hemorrhage and resuscitation. Am J Physiol 1990;259:R645-R650. 

Wang P, Ba ZF, Chaudry IH: Endothelial cell dysfunction occurs after hemor- 
rhage in nonheparinized but not in preheparinized models. J Surg Res 
1993;54:499-506. 

Wang P, Tait S, Ba ZF, Chaudry IH: ATP-MgC12 administration normalizes 
macrophage cAMP and beta-adrenergic receptors after Hhemorrhage and 
resuscitation. J Physiol 1994;267:G52-G58. 

Wang P, Ba ZF, Chaudry IH: Chemically modified heparin improves hepato- 
cellular function, cardiac output, and microcirculation after trauma: Hem- 
orrhage and resuscitation. Surgery 1994;116:169-175. 

Wang P, Ba ZF, Chaudry IH: Nitric oxide. To block or enhance its production 
during sepsis? Arch Surg 1994;129:1137-1142. 

Wang P, Ba ZF, Chaudry IH: ATP-MgC12 restores depressed endothelial cell 
function after hemorrhagic shock and resuscitation. Am J Physiol 
1995;268:H1390-H1396. 

Wang P, Ba ZF, Chaudry IH: Endothelium-dependent relaxation is depressed 
at the macro- and microcirculatory levels during sepsis. Am J Physiol 
1995;269:R988-R994. 

Wang P, Ba ZF, Stepp K, Chaudry IH: Pentoxifylline attenuates the depressed 
endothelial cell function and vascular muscle contractility following trauma 
and hemorrhagic shock. J Trauma 1995;39:121-126. 

Wang P, Ba ZF, Reich S, et al: Effects of nonanticoagulant heparin on cardio- 
vascular and hepatocellular function after hemorrhagic shock. Am J Physiol 
1996;270:H1294-H1302. 

Wang P, Ba ZF, Cioffi W, et al: Salutary effects of ATP-MgCl 2 on the depressed 
endothelium-dependent relaxation during hyperdynamic sepsis. Crit Care 
Med 1999;27:959-964. 

Ward P, Suzuki A: Gunshot wound of the heart with peripheral embolization. 
J Thorac Cardiovasc Surg 1974;68:440. 

Warren R: Report to the Surgeon General. Washington, DC, Department of 
the Army, 1952. 

Warren R, Akins CW, Conn A, et al: Acute traumatic disruption of the thoracic 
aorta: Emergency department management. Ann Emerg Med 1992;21 :39 1- 
396. 

Warrian R, Shoenut J, Iannicello C, et al: Seatbelt injury to the abdominal aorta. 
J Trauma 1988;28:1505-1507. 



RE.qxd 4/16/04 3:19PM PageS46 



646 VI • THE RICH REFERENCE COLLECTION 

Wascher R, Gwinn B: Air rifle pellet injury to the heart with retrograde caval 
migration. J Trauma 1995;38:379-381. 

Watson W, Silverstone S: Ligature of the common carotid artery in cancer of 
head and neck. Ann Surg 1939;109:1-27. 

Waxman K, Soliman M, Braunstein P, et al: Diagnosis of traumatic cardiac con- 
tusion. Arch Surg 1986;121:689-692. 

Waxman K: Shock: Ischemia, reperfusion, and inflammation. New Horiz 
1996;4:153-160. 

Weaver FA, Rosenthal R, Waterhouse G, Adkins RB: Combined skeletal and vas- 
cular injuries of the lower extremities. Am Surg 1984;50:189-197. 

Weaver FA, Yellin AE, Wagner W, et al: The role of arterial reconstruction in 
penetrating carotid injuries. Arch Surg 1988;123:1106-1111. 

Weaver FA, Suda R, Stiles G, Yellin AE: Injuries to the ascending aorta, aortic 
arch and great vessels. Surg Gynecol Obstet 1989;169:27-31. 

Weaver FA, Yellin AE, Bauer MAP, et al: Is arterial proximity a valid indication 
for arteriography in penetrating extremity trauma? A perspective analysis. 
Arch Surg 1990;125:1256-1260. 

Weber T, Dent T, Lindenaur S: Viable vein graft preservation. J Surg Res 
1975;18:247-255. 

Wei J, Chang C, Chang Y: Traumatic sinus of Valsalva fistula. Ann Thorac Surg 
1991;52:852-854. 

Weiland A, Robinson H, FutrellJ: External stabilization of a replanted upper 
extremity: Case report. J Trauma 1976;16:239-241. 

Weimann S, San Nicolo M, Sandbichler P, et al: Civilian popliteal artery trauma. 
J Cardiovasc Surg 1987;28:145-151. 

Weinstein M, Golding A: Temporary external shunt bypass in the traumatically 
amputated upper extremity. J Trauma 1975;15:912-915. 

Weisberg D, Goetz R: Necrosis of arterial wall following application of methyl- 
2-cyanoacrylate. Surg Gynecol Obstet 1964;119:1248. 

Weiser M, Gibbs S, Valeri C, et al: Anti-selectin therapy modifies skeletal muscle 
ischemia and reperfusion injury. Shock 1996;5:402-407. 

Weiss J, Bernadine HB, Hutchins G, Mason S: Two-dimensional echocardio- 
graphy recognition of myocardial injury in man: Comparison with post- 
mortem studies. Circulation 1981;63:401-408. 

Welborn MJ, Sas J: Acute abdominal aortic occlusion due to nonpenetrating 
trauma. Am J Surg 1969;118:112-116. 

Welling R, Kakkasseril J, Cranley J: Complete dislocations of the knee with 
popliteal vascular injury. J Trauma 1981;21:450-453. 

Welling R, Rremchek T, Rath R, et al: Blunt arterial injuries associated with mul- 
tiple trauma. Ann Vase Surg 1989;3:345-350. 

Werne C, Sagraves S, Costa C: Mitral and tricuspid valve rupture from blunt 
trauma sustained during a motor vehicle collision. J Trauma 1989;29:113- 
115. 

Wernly J, Campbell C, Replogle R: Traumatic avulsion of the innominate and 
left carotid arteries. J Thorac Cardiovasc Surg 1982;84:392-397. 

Wexler L, Silverman J: Traumatic rupture of the innominate artery: A seat belt 
injury. N Engl J Med 1970;282:1186-1187. 

Whelan TJ, Baugh JH: Non-Atherosclerotic Arterial Lesions and Their Man- 
agement: Current Problems in Surgery. Chicago, Year Book Medical Pub- 
lishers, 1967. 

White GH, White RA, Kopchok G, Wilson SE: Angioscopic thromboembolec- 
tomy: Preliminary observations with a recent technique. J Vase Surg 
1988;7:318-325. 



RE.qxd 4/16/04 3:19 PM Page 647 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 647 

White GH, White RA, Kopchok G: Endoscopic intravascular surgery removes 

intraluminal flaps, dissections and thrombus. J Vase Surg 1990;11:280-299. 
White J: Shark attack in Natal. Injury 1975;6:187-197. 

White J, Talbert J, Haller JAJr: Peripheral arterial injury in infants and chil- 
dren. Ann Surg 1968;167:757-766. 
Whitehouse W, Coran A, Stanley JC: Pediatric vascular trauma. Arch Surg 

1976;111:1269-1275. 
Whitesides T, Haney T, Morimoto K: Tissue pressure measurement as a deter- 
minant for the need for fasciotomy. Clin Orthop 1975;113:43-51. 
Whitman G, McCroskey B, Moore EE, et al: Traumatic popliteal and trifurca- 

tion vascular injuries: Determinants of functional limb salvage. Am J Surg 

1987;154:681-684. 
Wholey M, Bocher J: Angiography in musculoskeletal trauma. Surg Gynecol 

Obstet 1967;125:730-736. 
Wholey M, Bocher J: Angiographic features of aortic and peripheral arterial 

trauma. Arch Surg 1968;97:68-74. 
Whye D, Barish R, Alquist T, et al: Echocardiographic diagnosis of acute peri- 
cardial affusion in penetrating chest trauma. Am J Emerg Med 1988;6:2T 

23. 
Wiechert R, Hewitt RL: Injuries to the vena cava: Report of 35 cases. J Trauma 

1970;10:649-657. 
Wiedeman J, Mills J, Robison JG: Special problems after iatrogenic vascular 

injuries. Surg Gynecol Obstet 1988;166:323-326. 
Wiencek R, Wilson R: Abdominal venous injuries. J Trauma 1986;26:771-778. 
Wiencek R, Wilson R: Injuries to the abdominal vascular system: How much 

does aggressive resuscitation and prelaparotomy really help. Surgery 

1987;102:731-736. 
Wiener I, Flye M: Traumatic false aneurysm of the vertebral artery. J Trauma 

1984;24:346-349. 
Wigle R, Morgan J: Spontaneous healing of a traumatic thoracic aortic tear: 

Case report. J Trauma 1991;31:280-283. 
Wildegans H: Verletzungen der aorta. Deutsch Med Wchnschr 1926;52:1810- 

1812. 
Williams C, Brenowitz JB: Sequential aortic and inferior caval clamping for 

control of suprarenal vena caval injuries: Case report.J Trauma 1977; 17:1 64- 

167. 
Williams C, Robinson D: Traumatic abdominal aorta-inferior vena cava fistula 

with immediate repair. Ann Surg 1961;154:998-1000. 
Williams D: Stab wound of the heart and pericardium-recovery-patient alive 

three years afterward. Medical Record 1897;51:437-439. 
Williams D: Embolization of a bullet to the posterior tibial artery following a 

gunshot wound of the thorax. J Trauma 1964;4:258-265. 
Williams D, Brothers T, Messina L: Relief of mesenteric ischemia in type III 

aortic dissection with percutaneous fenestration of aortic septum. Radiol- 
ogy 1990;171:450-452. 
Williams G: Peripheral vascular trauma: Report of 90 cases. Am J Surg 

1968;116:725-730. 
Williams G, CrumplerJ, Campbell G: Effect of sympathectomy on the severely 

traumatized artery. Arch Surg 1970;101:704-707. 
Williams J, Silver D, Laws H: Successful management of heart rupture from 

blunt trauma. J Trauma 1981;21:534-537. 
Williams J, Graff J, Uku JM, SteinigJ: Aortic injury in vehicular trauma. Ann 

Thorac Surg 1994;57:726-730. 



RE.qxd 4/16/04 3:19PM PageS48 



648 VI • THE RICH REFERENCE COLLECTION 

Williams J, Sherman R: Penetrating wounds of the neck: Surgical management. 
J Trauma 1973;13:435-442. 

Williams T, Knopp R, EllysonJ: Compartment syndrome after anti-shock trouser 
use without lower extremity trauma. J Trauma 1982;22:595-597. 

Wilson C: Aortic bypass graft in a child. Am J Surg 1974;128:797-798. 

Wilson J: Fracture of the pelvis complicated by ischemia of the lower limb. 
J Bone Joint Surg 1952;34-B:68-69. 

Wilson L: Dispersion of bullet energy in relation to wound effects. Milit Surg 
1921;49:241-251. 

Wilson R, Bassett JS: Penetrating wounds of the pericardium or its contents. 
JAMA 1966;195:513-518. 

Wilson R, Arbulu A, BassetJS: Acute mediastinal widening following blunt chest 
trauma: Critical decisions. Arch Surg 1972;104:551-558. 

Wilson R, Wiencek R, Balog M: Predicting and preventing infection after abdom- 
inal vascular injuries. J Trauma 1989;29:1371-1375. 

Wilson R, Wiencek R, Balog M: Factors affecting mortality rate with iliac vein 
injuries. J Trauma 1990;30:320-323. 

Wilson SE, Veith F, Hobson RW II, Williams R: Noninvasive Diagnosis of Venous 
Obstruction and Insuficency. New York, McGraw-Hill, 1987. 

Wilson S, Au FC: In extreme use of a Foley catheter in a cardiac stab wound. 
J Trauma 1986;26:400-402. 

Wind G, Finley R, Rich NM: Three-dimensional computer graphic modeling 
of ballistic injuries. J Trauma 1988;28:S16-S20. 

Wiser E, Cheek R, Britt L: Gunshot wounds of the suprarenal abdominal aorta. 
Am Surg 1976;42:430-435. 

Witte C, Smith C: Single anastomosis vein bypass for subclavian vein obstruc- 
tion. Arch Surg 1966;93:664-666. 

Wixon C, PhilpottJ, Bogey WM, Powell C: Duplex-directed thrombin injection 
as a method to treat femoral artery pseudoaneurysms. J Am Coll Surg 
1998;187:464-466. 

Wolf R, Berry RE: Transaxillary intra-aortic balloon tamponade in trauma. 
J Vase Surg 1986;4:95-97. 

Wolf Y, Reyna T, Schropp K, Harmel R: Arterial trauma of the upper extrem- 
ity in children. J Trauma 1990;30:903-905. 

Wolfel G, Moore EE, Cogbill TH: Severe thoracic and abdominal injuries asso- 
ciated with lap-harness seatbelts. J Trauma 1984;24:166-167. 

Wood J, Fabian TC, Mangiante E: Penetrating neck injuries: recommendations 
for selective management. J Trauma 1989;29:602-605. 

Wood M, Nykamp P: Traumatic arteriovenous fistula of the superior mesen- 
teric vessels. J Trauma 1980;20:378. 

Woodhall B, Nulsen F: Peripheral nerve wounds. In Bowers WF (ed): Surgery 
of Trauma. Philadelphia, JB Lippincott Co, 1953. 

Woodhurst W, Robertson W, Thompson G: Carotid injury due to intraoral 
trauma: Case report and review of the literature. Neurosurgery 1980;6:559- 
563. 

WoodringJ, Fried A, Hatfield D, et al: Fractures of first and second ribs: Pre- 
dictive value for arterial and bronchial injury. AJR Am J Roentgenol 
1982;138:211-215. 

Woodruff C: The cases of the explosive effect of modern small caliber bullets. 
NYJMed 1898;67:593. 

Woodson G, Kendrick B: Laryngeal paralysis as the presenting sign of aortic 
trauma. Arch Otol Head Neck Surg 1989;115:1100-1102. 



RE.qxd 4/16/04 3:19 PM Page 649 



26 • RICH'S HISTORIC COLLECTION OF VASCULAR REFERENCES 649 

Woolsey R: Aortic laceration after anterior spinal fusion. Surg Neurol 
1986;25:267-268. 

Wright CB, Swan KG: Hemodynamics of venous occlusion in the canine hindlimb. 
Surgery 1973;73:141-146. 

Wright CB, Swan KG: Hemodynamics of venous repair in the canine hind limb. 
J Thorac Cardiovasc Surg 1973;65:195-199. 

Wright CB, Hobson R: Hemodynamic effects of femoral occlusion in the sub- 
human primate. Surgery 1974;75:453-460. 

Wright CB, Hobson RW II, Swan KG, Rich NM: Extremity venous ligation: 
Clinical and hemodynamic correlation. Am Surg 1975;41:203-208. 

Wright CB, Hobson RW II: Prediction of intestinal viability using Doppler ultra- 
sound techniques. Am J Surg 1975;129:642-645. 

Wright CB, Hobson RW II, Giordano J, et al: Acute femoral venous occlusion: 
Management by segmental venous replacement. J Cardiovasc Surg 
1977;18:523-529. 

Wright CB, Hiratzka L, Hobson RWII, et al: Management of vena caval injuries: 
The Vietnam Vascular Registry review. J Cardiovasc Surg 1981;22:203-212. 

Wright R, Edwards W: Manifestations and management of acute arterial injuries. 
J Trauma 1973;463-468. 

Wyatt D, Kellum CD, Joob A, Daniel T: Isolated injury of the proximal verte- 
bral artery associated with blunt chest trauma. J Vase Surg 1986;4:196-198. 

Yajko R, Trimble C: Arterial bullet embolism following abdominal gunshot 
wounds. J Trauma 1974;14:200-211. 

Yamada S, Kindt G, Youmans J: Carotid artery occlusion due to nonpenetrat- 
ing trauma. J Trauma 1967;7:333-342. 

Yamada T, Sato H, Seki M, et al: Successful salvage of aortoesophageal fistula 
caused by a fish bone. Ann Thorac Surg 1996;61:1843-1845. 

Yamaguchi Y, Matsumura F, Liang J, et al: Neutrophil elastase and oxygen 
radicals enhance monocyte chemoattractant protein-expression after 
ischemia/reperfusion in rat liver. Transplantation 1999;68:1459-1468. 

Yao JST, Vanecko RM, Printen K, Shoemaker WC: Penetrating wounds of the 
heart: A review of 80 cases. Ann Surg 1968;168:67-78. 

Yao J, Suri R, Patt N, et al: An unusual subclavian artery injury. J Trauma 
1970;10:176-180. 

Yarbrough DR III, Grooms G, Flint L: Repair of two traumatic aneurysms of 
the supraceliac abdominal aorta in a single patient. Surgery 1972;72:371- 
377. 

Yaw P, Van Beek A, Glover JL: Successful repair of a gunshot wound to the first 
part of the superior mesenteric artery. J Trauma 1974;14:885. 

Yeager RA, Hobson RW II, Lynch TG, et al: Popliteal and infrapopliteal arter- 
ial injuries: Differential management and amputation rates. Am Surg 
1984;50:155-158. 

Yee L, Olcott E, Knudson M, Lim RC Jr: Extraluminal, transluminal, and obser- 
vational treatment for vertebral artery injuries. J Trauma 1995;39:480-486. 

Yellin A, Golan M, Klein E: Penetrating thoracic wounds caused by plastic bullets. 
Thorac Cardiovasc Surg 1992;2:381. 

Yellin AE, Shore E: Surgical management of arterial occlusion following per- 
cutaneous femoral angiography. Surgery 1973;73:772-777. 

Yellin AE, Lundell C, Finck E: Diagnosis and control of post-traumatic pelvic 
hemorrhage. Arch Surg 1983;118:1378-1388. 

Yelon J, Scalea T: Venous injuries of the lower extremities and pelvis: Repair 
versus ligation. J Trauma 1992;33:532-538. 



RE.qxd 4/16/04 3:19PM PageS50 



650 VI • THE RICH REFERENCE COLLECTION 



Yelon J, Barrett L, Evans J: Distal innominate artery transection and cervical 
spine injury. J Trauma 1995;39:590-592. 

Yeo M, DomanskisE, BartlettRH, Gazzaniga A: Penetrating injuries of the abdom- 
inal aorta. Arch Surg 1974;108:839-844. 

Yoder R, Merck D: Innocuous appearing stab wounds to the neck: Is explo- 
ration always indicated? South MedJ 1969;62:113-114. 

Yoshioka H, Seibel R, Pillai K, Luchette F: Shotgun wounds and pellet emboli: 
Case reports and review of the literature. J Trauma 1995;39:596-601. 

Yosowitz P, Hobson RWII, Rich NM: Iliac vein laceration caused by blunt trauma 
to the pelvis. Am J Surg 1972;124:91-93. 

Youkey J, Clagett GP, Rich NM, et al: Vascular trauma secondary to diagnostic 
and therapeutic procedures: 1974 through 1982. Am J Surg 1983;146:788- 
791. 

Young J, Stallone R, Iverson L, et al: Surgical management of traumatic dis- 
ruption of the descending aorta. West J Med 1989;150:662-664. 

Young P: Following microdiskectomy: Laceration of the abdominal aorta and 
vena cava. IMJ 1988;174:93-94. 

Zabin A: Cholecystectomy complicated by hemorrhage from the inferior vena 
cava. N YJ Med 1950;50:1500. 

Zakharia AT: Cardiovascular and thoracic battle injuries in the Lebanon war. 
J Thorac Cardiovasc Surg 1985;89:723. 

Zakharia A: Thoracic battle injuries in the Lebanon war: Review of the early 
operative approach in 1,992 patients. Ann Thorac Surg 1985;3:209-213. 

Zehnder M: Delayed posttraumatic rupture of the aorta in a young healthy 
individual after closed injury: Mechanical-etiological considerations. Angi- 
ology 1956;7:252. 

Zehntner MK, Petropoulos P, Burch HB: Arterial injuries associated with frac- 
tures of the extremities. Proc of the First Mediterranean Congress of Angi- 
ology 1988;223-226. 

Zeiger M, Clark DE, Morton JR: Reappraisal of surgical treatment of traumatic 
transection of the thoracic aorta. J Cardiovasc Surg 1990;31:607-610. 

Zelenock G, Kazmers A, Whitehouse W: Extracranial internal carotid artery 
dissections. Arch Surg 1982;117:425-432. 

Zelenock G, Kazmers A, Graham L, et al: Nonpenetrating subclavian artery 
injuries. Arch Surg 1985;120:685-692. 

Zellweger R, Ayala A, Zhu X, et al: A novel nonanticoagulant heparin improves 
splenocyte and peritoneal macrophage immune function after trauma: 
Hemorrhage and resuscitation. J Surg Res 1995;59:211-218. 

Zhou M, Wang P, Chaudry IH: Endothelial nitric oxide synthase is downregu- 
lated during hyperdynamic sepsis. Biochim Biophys Acta 1997;1335:182- 
190. 

Zilkha A: Traumatic occlusion of the internal carotid artery. Radiology 1970; 
97:543-548. 

Ziperman H: Acute arterial injuries in the Korean War: A statistical study. Ann 
Surg 1954;139:1-8. 

Zuckerman J, Flugstad D, Teitz C, King H: Axillary artery injury as a compli- 
cation of proximal humeral fractures. Clin Orthop 1984;189:234-237. 

Zweifach S, Hargens A, Evans K, et al: Skeletal muscle necrosis in pressurized 
compartments associated with hemorrhagic hypotension. J Trauma 1980; 
20:941-947. 



INDEX. qxd 4/16/04 3:18PM Page 651 




INDEX 



Abdominal aorta, 340 

Abdominal aortic aneurysm (AAA), 207. See also 

Endovascular grafts. 
Abdominal aortic injury, 200 
Abdominal compartment syndrome, 111, 444-445, 

445-446, 448, 453 
Abdominal injuries, 299-313 

celiac trunk, 304-305 

clinical presentation, 300-301 

complications, 312 

Huey Long rule, 310 

incidence, 300 

inframesocolic area (zone 1), 307-309 

infrarenal abdominal aorta, 307-309 

kidney compartment syndrome, 310 

pathophysiology, 300 

renal artery, 307, 309-312 

renal revascularization, 310-311 

superior mesenteric artery, 305-307 

supramesocolic area (zone 1), 301-307 

suprarenal abdominal aorta, 301-304 

upper lateral retroperitoneum (zone 2), 302-312 

zones, 371 
Abdominal vascular injuries, 149-153, 200-201 
Abductor pollicis longus muscle, 373f 
Access, control and repair techniques, 137-164. See also 

Surgical techniques. 
Ace pressure monitor, 449 
Acidosis, 105 

Activated partial thromboplastin time (APTT), 108 
Active core rewarming, 106 
Active external rewarming, 106 
Acute arterial insufficiency, 126 
Acute arteriovenous fistula, 229 
Acute lung injury (ALI), 108, 109 
Acute respiratory distress syndrome (ARDS), 79, 

109 
Adenosine triphosphate (ATP), 75 
Advanced trauma life support (ATLS) guidelines, 

117 
Aeneid (Virgil), 252 
Afghanistan War, 27t 
Air hammer, 54 



Algorithms 

DVT, 534f 

extremity vascular injury, 129f 

penetrating extremity injuries, 93f 
ALI, 108, 109 
Allopurinol, 78, 453 
Amputation 

axillary artery injury, 367 

brachial artery injury, 371 

combat extremity arterial injuries, 405t 

combined extremity injuries, 406, 412-414 

common femoral artery injury, 379 

extremity vascular injury, 358 

ligation of femoral arteries, 1 70 

mangled lower extremity, 387 

profunda femoral artery injury, 379 

subclavian artery injury, 363 

superficial femoral artery injury, 380 
Anastomosis, 159-161 
Anatomy. See Surgical anatomy. 

Ancillary tests, 119-121. See also Imaging techniques. 
Anderson, John T, 113 
Anesthesia, 104 
AneuRx grafts, 209 
Aneurysmal abscess, 423 
Angio-Seal device, 432, 432f 
Angiogram /angiography 

AV fistula, 465f, 470f, 474f, 483f, 484f, 498f 

complications, 51t 

contrast media-related risks, 181 

CTA. SeeCT/CTK. 

false aneurysm, 61f, 510f, 512f, 513f 

findings, 184-192 

follow-up angiogram, 60f 

follow-up femoral angiogram, 62f 

gold standard, as, 102, 182 

gunshot wound (chest), 68f, 214f 

iatrogenic AVF, 218f 

iliac vessel injury, 342f, 344 

indications/contraindications, 180-182 

intra-arterial metallic fragment, 65f 

MRA. S^MRA. 

occlusion (right common iliac artery), 342f 



651 



INDEX. qxd 4/16/04 3:18PM Page 652 



652 INDEX 



Angiogram /angiography (Continued) 

PE, 531 

pelvis, 202 

penetrating cervical injuries, 231-232 

technical complications, 181-182 
Angiographic catheter complications, 51t 
Angiographically placed stents, 348-349 
Anterior circumflex humeral artery, 364f 
Anterior spinal artery, 448 
Anterior spinal artery syndrome, 448 
Anterior tibial artery, 381f, 382 
Anterior tibial recurrent artery, 381f 
Anterior ulnar recurrent artery, 372f 
Anterolateral thoracotomy, 146, 147, 292 
Anteroposterior chest radiograph, 255 
Antibiotics, 110 
Anticoagulation, 198 
Antiplatelet therapy, 198 
Antyllus, 499 

Aortic compression device, 303f 
Aortic occlusion, 292 
Aortography 

arch, 196f 

blunt thoracic injury, 271 

deficiencies, 271 

pelvic, 202 

"pullout" abdominal, 312 

subtraction, 257f 
API, 118-119 
APTT, 108 

Arch aortography, 196f 
Archigenes, 5 
ARDS, 79, 109 
Arm, 447 

Arm injury, 202-203 

Armed Forces. See Military vascular trauma experience. 
Arterial aneurysms, 502. See also Traumatic false 

aneurysms. 
Arterial closure devices, 431-435 
Arterial injury 

blunt, 103 

hard/soft signs, lOOt 

types, lOlf 
Arterial pressure index (API), 118-119 
Arterialization, 477 
Arteriogram/arteriography 

AV fistula, 472f, 473f, 474f, 494f, 498f 

axillary artery injury, 365 

bleeding (right hepatic artery), 194f 

calf, 205 

coil occlusion, 193f 

combined extremity injuries, 407, 408f 

common femoral artery injury, 377 

completion, 162, 398, 411f, 416 

contrast, 127, 132 

elbow, 194f 

external carotid artery, 198 

extremity vascular injuries, 356 

ICA injury, 198f 

indications, 120t 

intraoperative, 408f, 417 

nonsuture anastomsis, 59f 



Arteriogram/arteriography (Continued) 

pelvis, 202 

penetrating thoracic injury, 255-256 

popliteal/ tibial artery injury, 383 

posterior knee dislocation, 396 

profunda femoral artery injury, 377 

pseudoaneurysm (subclavian artery), 21 7f 

vascular injury repair, 41 If 
Arteriovenous (AV) fistula, 190f, 458-499 

Branham-Nicoladoni sign, 478, 480 

cardiac enlargement, 481, 482f, 484 

clinical features, 480-481 

clinical pathology, 478-480 

diagnostic considerations, 481-484 

edema, 479f 

etiology, 470-475 

follow-up, 497-498 

historical overview, 458-461 

incidence, 461-470 

Korean conflict, 468t, 495t 

Location, 465t, 466f, 467t, 468t, 469t 

oscillometric studies, 493t 

pathophysiology, 475-478 

pressure flow, 477-478 

pulsating venous lakes, 479f 

results, 488-497 

spontaneous cure, 486-488 

surgical treatment, 484-486 

types, 464f 

vein transplantation, 492t 

Vietnam Vascular Registry, 464t, 465t, 469t, 47lt, 
495t, 496t, 497 

World War II, 490t 
Artery injury, 371-374 
Artery of Adamkiewicz, 200-201 
Ascending aorta injury, 262-263 
Ascending lumbar vein, 3l7f 
Ascending pharyngeal artery, 226f 
Asensio, Juan A., 339 
Asthma, 181 
Athletic injuries, 53 
ATLS guidelines, 117 
ATP, 75 
ATP-MgCl 2 , 80 
Autogenous conduits, 162 
Auto transfusion, 105 

AV fistula. See Arteriovenous (AV) fistula. 
Axillary artery, 365f 
Axillary artery injuries, 148, 364-367 
Axillary artery laceration with pseudoaneurysm, 357f 
Axillary subclavian artery, 210f 
Axillary vein, 364, 365f 
Azygos vein injury, 147-148, 264 



Babcock clamps, 322, 322f 

Bailout procedure, 168, 169 

Balkan Wars, 13-14, 488 

Ballistics, 55-56 

Balloon catheter tamponade, 140, 170, I7lf 

Balloon occlusion, 200 

Baseball players, 53 



INDEX. qxd 4/16/04 3:18PM Page 653 



INDEX 



653 



Basilar artery, 226f 

Basilic vein, 368f 

BCVL See Blunt cervical vascular trauma. 

BCVI grading scale, 246, 247t 

Bee de corbin, 6f 

Beck's triad, 444 

Below the knee medial incision, 156f 

Bibliography, 537-649 

Bicipital aponeurosis, 373f 

Biffl, Walter L., 241 

Bifurcation, 317, 320 

Bilateral hypogastric angiography, 202 

Blaisdell, R William, 113 

Bleeding/bleeding control. See also 

Hemorrhage/hemorrhage control. 

BCVIs, 248 

blood transfusion, 108 

iliac vessel injury, 347-348 

postoperative phase, 111 

primary survey, 99 

quantification of blood loss, 99t 
Blood transfusion 

autotransfusion, 105 

complications, 108, 109b 

perioperative care, 105 

postoperative phase, 107 
Blue toes, 437 

Blunt abdominal vascular injuries, 102 
Blunt arterial injuries, 103 
Blunt cervical vascular trauma, 241-250 

anatomic considerations, 242 

clinical presentation, 244 

diagnostic evaluation, 246 

incidence, 245 

injury grading, 246 

mechanism of injury, 242-243 

pathophysiology, 243 

patient at risk, 245-246 

screening, 245 

summary and guidelines, 249-250 

treatment/ outcome, 2246-249 
Blunt thoracic aortic injury (BTAI). See Blunt thoracic 

vascular trauma. 
Blunt thoracic vascular trauma, 269-283 

aortogram, 271-272 

chest x-ray, 272-273 

clamp-and-sew technique, 277-278 

CT, 273-275 

demographics/pattern of injury, 270-271 

diagnosis, 271-276 

DPL, 276 

exposure, 279t 

innominate artery, 278-279 

preoperative management, 276-277 

presentation, 271 

primary repair vs. prosthetic graft, 278 

pulmonary artery/vein, 280 

shunt techniques, 278 

subclavian artery, 279-280 

surgical technique, 277-278 

TRR, 275-276 

vena cava, 280 



Blunt trauma 

cervical injuries, 241-250 

concomitant injuries, 103-104 

mechanism of injury, 53 

thoracic injuries, 269-283 
Bogota bag, 453 
Boyden, Allen M., 17 
Brachial artery, 367-368 
Brachial artery injuries, 148-149, 182f, 202-203, 

367-371 
Brachialis muscle, 368f, 373f 
Brachiocephalic artery, 359f 
Brachiocephalic vessel injury, 130-132, 199-200 
Brachioradialis muscle, 373f 
Bradley, Kevin M., 315 
Branham-Nicoladoni sign, 478, 480 
BTAI, 269-283. See also Blunt thoracic vascular trauma. 
Buckman, Robert R, Jr., 315 
Bulldog clamps, 323 
Bullet embolism, 119 
Burch,JonM., 241 
Butterfly fracture, 343 



CABG, 294f 

Calcium replacement, 108 

Calf compartment syndrome, 385, 446 

Calf injury, 205 

Cannulation of large veins, 99 

Cannulation of right subclavian vein, 435 

Carbon dioxide, 181 

Cardiac compressions, 292 

Cardiac enlargement, 481, 482f, 484 

Cardiac manipulation, 292 

Cardiac septal injuries, 294-295 

Cardiac trauma. See Wounds of the heart. 

Cardiac valvular injury, 295 

Cardiopulmonary bypass, 296 

Carotid bulb, 225 

Carotid/internal jugular vein fistula, 436f 

Carotid sheath, 227f 

Catheter-based angiography, 182. See also 

Angiogram/angiography. 
Cattell-Braasch maneuver, 152, 153f 
Causalgia, 375-376 
Caval bifurcation zone, 317, 320 
Caval wounds. See Inferior vena cava injuries. 
Cayne, NealS., 207 
CD11/CD18, 76 
Celiac artery, 340f 
Celiac trunk injury, 304-305 
Celsus, 5 

Central venous access, 435 
Cephalic vein, 368f 
Cephalosporin, 104 
Cerebral vasospasm, 229 
Cervical injuries 

blunt trauma, 241-250. See also Blunt cervical 
vascular trauma. 

neck injuries. See Neck injuries. 

penetrating injuries, 223-240. See also Penetrating 
cervical vascular injuries. 



INDEX. qxd 4/16/04 3:18PM Page 654 



654 INDEX 



CFA, 204, 376 

Chaudry, Irshad H., 73 

Chest injuries, 146-148 

Chest radiography, 255t, 257-258f 

Chest tube sizes, 254t 

Chest x-ray, 272-273 

Chronic venous insufficiency, 47f 

Circle of Willis, 225, 228f 

Circumflex nerve, 365f 

Citrate intoxication, 108 

Civilian penetrating trauma, 115 

Clamp-and-sew technique, 277-278 

Clamping 

cross, 296 

innominate artery/vein, 261 

IVC injuries, 322-323 

surgical technique, 151, 154 

walking the clamps, 154 
Classification of injury, 114 
cNOS, 78 

Coagulation monitoring, 107-108 
Cocaine, 425-426 
Coil blockade, 202 
Coil embolization, 193, 208 
Coil occlusion, 193, 202 
Coimbra, Raul, 97 
Collagen plug, 432 
Color flow duplex imaging. See Duplex 

ultrasonography. 
Colorectal operations, 437 

Combined Kocher and Cattell-Brassch maneuver, 153f 
Combined Palmaz stent/ ePTFE graft, 209, 211f, 214f, 

21 7f, 218f 
Combined vascular/ skeletal extremity trauma, 
404-414 

amputation, 405t, 412-414 

diagnosis, 406-408 

epidemiology/prognostic factors, 404-406 

fracture management, 412 

mechanism of injury, 406 

treatment, 408-412 
Common bile duct, 328f 
Common carotid artery, 226f, 227f, 435f 
Common femoral artery (CFA), 204, 376 
Common femoral artery injury, 376-379 
Common iliac veins, 340 
Common interosseous artery, 373f 
Compartment hypertension, 417 
Compartment syndrome, 443-455 

abdomen, 448 

abdominal, 111, 444-445, 445-446, 448, 453 

arm, 447 

calf, 446 

diagnosis, 448 

diabetes, 446 

extremity, 375, 385, 450-454 

extremity compartment measurement, 449 

hand, 447 

laboratory evaluation, 450 

lower extremity, 385 

noninvasive assessment, 449-450 

pathophysiology, 116, 117, 450 



Compartment syndrome (Continued) 

pericardial, 444 

pericardium, 448 

postoperative phase, 111-112 

presentation, 445-446 

prophylactic fasciotomy, 133 

spinal, 444 

spinal cord, 448 

systemic diseases, 446 

thigh, 446 

treatment, 450-454 

upper extremity, 375 
Completion arteriography, 162, 398, 411f, 416. See also 

Arteriogram/arteriography. 
Complex repairs, 170 
Complications 

abdominal injuries, 312 

angiographic procedures, 51t 

angiography, 181-182 

autogenous venous grafts, 45 

blood transfusion, 108, 109b 

elective operative procedures, 50t 

extremity revascularization, 129-130 

iliac vessel injury, 349-350 

incidence, 51t 

passive rewarming, 106 

thromboembolic, 525-535. See also Thromboembolic 
complications. 
Computed tomography/computed tomographic 

angiography. See CT/CTA. 
Concomitant blunt thoracic/abdominal trauma, 103 
Concomitant injuries, 103-104 
Conduit occlusion, 193 
Conklin, Lori D., 251 
Constitutive NOS (cNOS), 78 
Continuous anastomosis, 160 
Contrast arteriography, 127, 132. See also 

Arteriography. 
Contrast media-related risks, 181 
Contusion, 54f 

Coracobrachialis muscle, 368f 
Coronary artery bypass grafting (CABG) , 294f 
Coronary artery injuries, 293-294 
Coronary vein, 328f 

Corvita endovascular graft, 209f, 211-212 
Costocervical trunk, 200, 359f, 360f 
Coverage, 163 
Covered stents, 195 
Crafoord clamps, 323 
Cragg Endopro, 209f 
Crawford, E. Stanley, 439 
Croatia, 27t, 405t 
Cross clamping, 296 
Cross-matched blood, 105 
Crutches, 53 
Cryoprecipitate, 106 
Crystalloids, 105 
CT angiography. See CT/CTA. 
CT/CTA, 182-184 

arterial injury, 183f 

BCVIs, 246 

BTAI, 273-275 



INDEX. qxd 4/16/04 3:18PM Page 655 



INDEX 



655 



CT/CTA (Continued) 
effectiveness, 131-132 
extremity vascular trauma, 356 
hepatic hilar arterial injury, 197f 
iliac vessel injury, 343-344 
laceration of liver, 194f 
PE, 531, 532f 

penetrating cervical injury, 232-233 
penetrating thoracic injury, 256 
radial and ulnar artery injury, 373 
renal artery, 310 
retroperitoneal hematoma, 432f 

Cyclic hyper-resuscitation, 142 



Dacron graft. SsePTFE (Dacron) graft. 
Dacron substitute conduits, 162 
Damage control, 165-176 

acidosis, 168 

coagulopathy, 168 

historical overview, 166, 167t 

hypothermia, 167-168 

initial operation, 168-169 

penetrating thoracic injury, 259-260 

perioperative care, 104-105 

phases, 168-170 

physiologic envelope, 167-168 

planned reoperation, 169, 173-174 

postoperative limb ischemia, 172-173 

repair, 170-171 

resuscitation, 169 

surgery, 157 

temporary shunts, 171-172 
De Takats, Geza, 14, 15f 
Dead space, 184 

DeBakey, Michael E., I7f, 18, 59, 443 
Decompression, 451-453 
Deep femoral artery, 377f 
Deep mattress sutures, 293 
Deep radial nerve, 373f 
Deep venous thrombosis (DVT) . See also 
Thromboembolic complications. 

algorithm, 534f 

diagnosis, 530 

heparin, 531 

iliac vessel injury, 350 
Deferoxamine, 453 
Demetriades, Demetrios, 339 
Descending genicular artery, 377f, 381f 
Descending hypoglossal nerve, 227f 
Descending thoracic aortic injury, 263 
Dextran, 388 

Diabetes, compartment syndrome, 446 
Diagnosis of vascular trauma, 113-14. See also Imaging 
techniques. 

ancillary tests, 119-122 

classification, 114 

compartment syndrome, 116-117 

diagnosis, 117-122 

hard/ soft signs, 118-119 

history, 117-118 

ischemia, 116 



Diagnosis of vascular trauma (Continued) 

mechanism, 115-116 

pathophysiology, 114-117 

physical examination, 118 

reperfusion injury, 116 
Diagnostic imaging. See Imaging techniques. 
Diagnostic peritoneal lavage (DPL), 276 
Diaphragmatic aorta, 301, 302f 
DIC, 108 

Die Chirgurie der Blutgefasse und des Herzens (Jeger), 16 
Digital subtraction angiography, 271-272 
Digital thoracotomy, 290 
Direct balloon occlusion, 199 
Dislocation of knee, 52-53 

Disseminated intravascular coagulation (DIC), 108 
Distal brachial artery injury, 149 
Distal renal artery, 309 
Distal superficial femoral artery, 155 
Distal superficial femoral artery injury, 115f 
Djanelidze, Yustin, 252 
DopplerAPI, 127-128 

Dorsal cutaneous branch ulnar nerve, 373f 
Dorsal interosseous artery, 372f 
Dorsal scapular artery, 359f, 360f 
Double Pringle maneuver, 153 
DPL, 276 

Duett device, 433, 433f 
Dum-dum bullet, 56 
Duplex ultrasonography, 120, 121 

axillary vein valve transfer, 64f 

BCVIs, 246 

brachial artery injury, 182f 

cardiac trauma, 290 

combined extremity injuries, 408 

DVT, 530, 531f, 533t 

effectiveness, 128-129, 131, 133 

penetrating cervical injury, 232 

popliteal/ tibial artery injury 382 

venous injuries (lower extremity), 388 

venous injury, 401, 404 
DVT. See Deep venous thrombosis (DVT). 



Early ALI/ARDS, 109 

Early direct vascular reconstruction, 7-13 

Eber's papyrus, 5 

EC thoracotomy, 100, 254, 290 

Echocardiography, 290, 295 

Edema 

AV fistula, 479f 

incidence, 45t 
Elbow dislocation, 410f 
Elective operative procedures, vascular injury, 

50t 
Elliott, David C, 269 
Embolization, 193-195 

categories, 193 

circumflex femoral arteries, 205 

coil, 193, 208 

empirical, 201 

gelfoam, 194f 

ICA injury, 196 



INDEX. qxd 4/16/04 3:18PM Page 656 



656 INDEX 



Embolization (Continued) 

iliac vessel injury, 349f 

lumbar artery injury, 200, 201 

pelvic injury, 201, 202 

upper extremity injury, 203 

vertebral artery injury, 198-199 
Emergency center (EC) thoracotomy, 100, 254, 

294 
Empirical embolization, 201 
End-to-end repair, 160-161 

Endoaneurysmorrhaphy, 500, 501f, 515f, 516f, 518t 
Endothelial and neuronal NOS, 78 
Endovascular aortic aneurysm repair, 439-440 
Endovascular grafts, 207-220 

background, 208-211 

lesions, location, characteristics, 210t 

Montefiore experience, 211-218 

types of devices, 209 
Endovascular interventions, 192-196, 207-220 

embolization, 193-195. See also Embolization. 

grafts, 207-220. See also Traumatic vascular lesions. 

stent/ stent grafts, 195-196 

temporary hemostasis, 192 

traumatic vascular lesions, 207-220 

vascular bed occlusion, 195 
Endovascular stent grafts, 199 
Endovascular stenting, 256-259 
ePTFE graft, 209, 211f, 214f, 2l7f, 218f 
Esmarch, Freidrich van, 6 
Etomidate, 104 
Expanded PTFE (ePTFE) graft, 209, 211f, 214f, 2l7f, 

218f 
Exploratory laparotomy, 103 
Exposure. See Incisions. 
Extended Kocher maneuver, 152 
Extensive Kocher maneuver, 302, 307 
External carotid artery, 225, 226f 
External carotid artery injury, 198 
External compression, 54f 
External iliac artery, 340, 377f 
External iliac vein, 31 7f 
Extra-anatomic bypass, 398, 424 
Extravasation, 184, 185f 

Extremity compartment measurements, 449 
Extremity compartment syndrome, 111-112, 

450-453 
Extremity injuries, 78-79, 101-106 

algorithm, 129f 

diagnosis, 126-130 

evaluation (flowchart), 122f 

fractures, 103b 

initial care, 102-103 

lower extremity. See Lower extremity. 

mangled extremities, 404. See also Combined 
vascular/skeletal extremity trauma. 

revascularization, 129-130 

upper extremity. See Upper extremity. 

vascular trauma. See Extremity vascular trauma. 
Extremity revascularization, 129-130 
Extremity vascular trauma, 353-389 

axillary artery injuries, 364-367 

brachial artery injuries, 367-371 



Extremity vascular trauma (Continued) 
clinical presentation, 355 

compartment syndrome (upper extremity), 375 
diagnosis, 355-356 
hard/ soft signs, 356t 
lower extremity vascular injuries, 376-389. See also 

Lower extremity vascular injuries, 
management, 359t 
nonoperative management, 356-357 
operative management, 357-359 
post-traumatic causalgia, 375-376 
radial/ulnar artery injury, 371-374 
subclavian artery injuries, 359-364 
venous injuries (upper extremity), 374-375 



Facial artery, 226f 

Failed repair of arterial injury, 416-417 

False aneurysms. See Traumatic false aneurysms. 

Farrier's (veterinarian's) stitch, 8f 

Fasciotomy 

combined extremity injuries, 412 

compartment injury of upper extremity, 375 

compartment syndrome, 444, 450-451 

extremity vascular injury, 358 

iliac vessel injury, 348 

popliteal artery injury, 399 

prophylactic, 130, 133 

when needed, 163, 417 
Feliciano, David V., 299 
Femoral access site complications, 182 
Femoral arteries, 204-205 
Femoral artery, 21 Of 
Femoral nerve, 376 
Femoral-to-fe moral artery bypass, 424 
Fentanyl, 104 
FFP, 106 

Flexor carpi radialis muscle, 373f 
Flexor carpi radialis tendon, 373f 
Flexor carpi ulnaris muscle, 373f, 44 7f 
Flexor digitorum profundus muscle, 373f, 447f 
Flexor digitorum sublimis muscle, 373f, 44 7f 
Flexor digitorum superficialis tendons, 373f 
Flexor pollicis longus muscle, 373f 
Fogarty balloon catheter, 141, 157, 158b, 440 
Fogarty catheterization, 425 
Foley catheter balloon tamponade, 347f 
Follow-up, 58-59. .SVe Vietnam Vascular Registry. 
Follow-up angiogram, 60f 
Follow-up femoral angiogram, 62f 
Forearm, 447 

Foreign body embolism, 265 
Four-compartment decompression, 451f 
Four-compartment fasciotomy, 41 2 
Fracture, 103b 
Fractures, 52 
French sheaths, 428 
Fresh frozen plasma (FFP), 106 
Fresh fungus vegetations, 423 
Frykberg, Eric R., 393 
Fullen zone I injuries, 301, 306 
Fullen zone II injuries, 301, 306 



INDEX. qxd 4/16/04 3:18PM Page 657 



INDEX 



657 



Gadolinium, 181 
Galen, 5, 286 
Garcia, Priscilla, 443 
Gargiulo, Nicholas J., Ill, 207 
Gelfoam 

pelvic injury, 202 

upper extremity injury, 202 

vascular bed occlusion, 195 
Gelfoam pledget embolization, 1943f 
Gerota's fascia, 310, 448 
Gluteal compartment syndrome, 446 
Gott shunt, 278 
Graft/grafting 

Corvita stent graft, 209f, 211-212 

endovascular grafts, 207-220 

ePTFE graft, 209, 211f, 214f, 2l7f, 218f 

interposition grafting, 310 

IVC injury, 325, 326f 

new devices, 209,212 

patch grafting, 414 

portal vein, 335f 

prosthetic grafts, 414-415 

PTFE graft. SeePTFE (Dacron) graft. 

stent grafts. See Stent/ stent grafts. 

substitute conduit, 161-162 

vascular grafts, 414-416 

vein graft, 209 

wall graft, 196f, 211, 212 
Granchi, Thomas S., 443 
Grenada, 57 

Groin injury, 154-155, 203-204 
Gulf War, 57 
Gunshot wound 

abdominal aorta injury, 470f 

ballistics, 55-56 

chest, 68f, 214f, 215f 

etiology, 38t, 39t 

iliac vessel injury, 341 1 

infrarenal abdominal aorta, 308 

IVC injury, 318 

location, 38t 

minimal injury, 89t 

neck, 233f, 238f 

right media knee, 356f 

thrombosis of superficial femoral artery, 191f 
Gustilo III-C complex extremity crush injury, 413f 



Haiti, 57 
Hand, 447 
Hard signs 

arterial injury, lOOt 

combined extremity injuries, 407 

knee dislocation, 397t 

vascular injury, 118-119, 356t 
Heart. See Wounds of the heart. 
Heliodorus, 5 
Hemodilution, 168 

Hemorrhage/hemorrhage control. See also 
Bleeding/bleeding control. 

cardiac trauma, 292-293 

common femoral artery injury, 377 



Hemorrhage /hemorrhage control (Continued) 

high-risk injuries, 143b 

infrahepatic IVC, 321-323 

pelvic fracture, 201 

penetrating thoracic injury, 266 

percutaneous vascular access, 430-431 

popliteal artery injury, 398 

profunda femoral artery injury, 377 

retrohepatic IVC, 320, 323-324 

superficial femoral artery injury, 380 

suprarenal abdominal aorta, 303 

surgery, 139-142 
Henry principle of extensile exposure, 149, 155 
Heparin 

BCVIs, 249t 

cocaine, vascular injury from, 426 

DVT/PE, 528-529, 531, 533t 

perioperative care, 105 

surgery, 157-159 

thromboembolic complications, 528-529, 531, 
533t 
Heparin-bonded Gott shunt, 278 
Hepatic hilar arterial injury, 197f 
Heroin, 422 
Herophilus, 5 

High-risk orthopedic injuries, 94-95 
Hip arthroplasty, 440 
Hip injury, 203-204 
Hirshberg, Asher, 137, 165 
Historic classification of vascular injury, 54-55 
Historic observations 

iatrogenic injury, 48-52 

mechanism of injury, 52-54 

site of injury, 47-48 
Historical overview, 3-72. See also Military vascular 
trauma experience. 

AVF, 458-461 

Balkan Wars, 13-14 

ballistics, 55-56 

bee de corbin, 6f 

civilian vascular injuries, 28-37 

damage control, 166, 167t 

diagnosis of extremity injury, 126-127 

early direct vascular reconstruction, 7-13 

Egyptians (1600 B.C.), 5 

false aneurysms, 499-502 

first end-to-end anastomosis, 9f, 1 If 

heart, wounds of, 285-286 

Korean Conflict. See Korean Conflict. 

Military vascular trauma experience, 13-28 

minimal injuries, 87-88 

Pare, 6 

penetrating cervical injuries, 224 

penetrating thoracic injuries, 253 

popliteal artery injuries, 394-395 

post-Vietnam military armed conflict, 27-28, 57 

reflections and projections, 56-68 

site of injury, 47-48 

tourniquet, 6 

thromboembolic complications, 525-526 

20th century, 37-46 

venous injuries, 399-400 



INDEX. qxd 4/16/04 3:18PM Page 65 8 



658 INDEX 



Historical overview (Continued) 

Vietnam War, 23-27. See also Vietnam War. 

World War I. See World War I. 

World War II. See World War II. 
Historical reflections and projections, 56-58 
History, 117-118 
HMS Tonnant, 6, 224 
Hoyt, David B., 97 
Huey's Long rule, 310 
Hughes, Carl, 21-23 
Hunter, John, 6 

Hunter, William, 6, 458, 459t, 478, 499 
Hyperkalemia, 108 
Hypocalcemia, 108 
Hypogastric arteriography, 202 
Hypogastric vein, 3l7f 
Hypothermia, 106, 167-168 
Hypothermic circulatory arrest, 324 
Hypoxia, 74 



IABP, 435-437 

Iatrogenic vascular trauma, 203, 427-442 

arterial closure devices, 431-435 

central venous access, 435 

colorectal procedures, 437 

hemorrhage, 430-431 

historical overview, 48-52 

IABP, 435-437 

intraoperative vascular injuries, 437-441 

laparoscopic procedures, 437-438 

neurosurgery, 440-441 

orthopedic surgery, 440 

pancreatobiliary procedures, 437 

percutaneous vascular access, 428-437 

pseudoaneurysms, 429-430 

vascular/ endovascular surgery, 438 
ICA, 225, 226f 
ICA injuries, 196-198 
ICAM-1, 75, 76 
Ideal conduit, 57 
Iliac arterial injuries, 345-346 
Iliac artery, 21 Of 
Iliac vascular injury, 154 
Iliac venous injury, 346-347 
Iliac vessel injury, 339-351 

anatomy, 340-341 

angiography, 344 

arterial injuries, 345-346 

bleeding control, 347 

clinical presentation, 342 

complications, 349-350 

CT, 343-344 

diagnostic investigations, 342-344 

fasciotomy, 348 

incidence/epidemiology, 341 

interventional radiology, 348-349 

mortality, 351 

operative management, 344-348 

perioperative management, 348-351 

radiographic studies, 342-343 

venous injuries, 346-347 



/^(Horner), 252 

Illicit street drugs, 421-426 

cocaine, 425-426 

direct arterial injection, 422-423 

mycotic aneurysms, 423-424, 425 

perivascular hematoma/abscess, 421-422 

vascular reconstruction, 424 

venous aneurysms, 425 
Imaging techniques 

alternate, 182-184 

ancillary tests, 119-121 

angiography. .See Angiography 

Aortography. See Aortography. 

arteriography. See Arteriography. 

best practices, 125-134 

chest radiography, 255t, 257-258f 

CTA. SeeCT/CTA. 

MRA. SeeMRA. 

most important role, 181 

TCD studies, 132 

TEE, 275-276 

ultrasound. See Duplex ultrasonography. 

venogram/venography, 43f, 44f, 400, 404 

x-rays, 119 
Incisions, 140b 

antecubital fosca, 51 7f 

anterior tibial injury, 386f 

AV fistula, 485 

below the knee medial, 156f 

brachial artery injury, 370f 

cardiac trauma, 291-292 

common feinoral artery, 378f 

compartment syndrome, 447f, 452f 

false aneurysms, 5l7f 

midline laparotomy, 149 

neck, 143-144 

out-of-favor techniques, 143b 

popliteal exposure, 384f 

profunda femoral artery, 378, 378f 

radial and ulnar artery injury, 374f 

radial/ulnar arteries, 447f 

subclavian artery injury, 361 

thoracic vascular injuries, 256t 

venous injuries, 401 
Incomplete transection, 54f 
Inducible NOS (iNOS), 78 
Inferior gluteal artery, 377f 
Inferior lateral genicular artery, 381f 
Inferior medial genicular artery, 381f 
Inferior mesenteric vein, 328f 
Inferior ulnar collateral artery, 372f 
Inferior vena cava injuries, 264, 316-327 

exposure and control, 319-324 

hemorrhage control, 321-324 

initial assessment and inanagement, 319 

patterns of injury, 318-319 

postoperative inanagement, 325-326 

repair, 324-325 

surgical anatomy, 317-318 
Inferior vena cava repair, 324-325 
Infrarenal abdominal aorta, 307-309 
Initial anesthesia, 104 



INDEX. qxd 4/16/04 3:18PM Page 659 



INDEX 



659 



Initial care 

abdominal vascular injuries, 102 

concomitant injuries, 103-104 

extremity injuries, 102-103 

general guidelines, 98-100 

neck vascular injuries, 101 

primary survey, 98-100 

secondary survey, 100 

thoracic vascular injuries, 101-102 
Innominate vein/artery 

blunt thoracic injuries, 278-279 

penetrating cervical injuries, 233 

penetrating thoracic injuries, 261-262 

thoracic outlet injury, 146 
iNOS, 78 

Intercostal artery injury, 264 
Interior carotid artery, 226f 
Interior iliac artery, 377f 
Interior jugular vein, 227f 
Interior thoracic artery, 359f, 360f 
Internal carotid artery (ICA), 225, 226f 
Internal carotid artery (ICA) injuries, 196-198 
Internal iliac artery, 340 
Internal jugular vein, 435, 435f 
Internal thoracic injury, 264 
Interosseous artery, 372f 
Interosseous recurrent artery, 372f 
Interposition grafting, 310, 414 
Interventional radiology, 180, 348. See also 

Endovascular interventions; Surgical techniques; 
Surgical treatment. 
Intimal flaps, 190 

Intra-aortic balloon pump (IABP), 435-437 
Intra-arterial metallic fragment, 65f 
Intraluminal balloon control of arteries, 208 
Intraluminal filling defects, 187, 187f, 189 
Intraluminal shunting, 399 
Intraluminal shunts, 171 
Intraluminal thrombus, 189-190 
Intraoperative arteriogram, 417 
Intraoperative monitoring, 104 
Intraoperative vascular injuries, 437-441 

colorectal procedures, 437 

laparoscopic procedures, 437-438 

neurosurgery, 441-442 

orthopedic surgery, 440 

pancreatobiliary procedures, 437 

vascular/ endovascular surgery, 438-440 
Intrapericardial inferior vena cava injury, 

295-296 
Intrathoracic inferior vena cava injury, 264 
Intravascular stents, 208 
Introducer sheath, 434f 
I /R injury. .S^Ischemia-reperfusion injury. 
Ischemia 

defined, 74 

extremity vascular injury, 360 

five Ps, 74 

iliac arterial injuries, 346 

pathophysiology, 116 

postoperative limb, 172-173 

signs, 74 



Ischemic preconditioning, 80 
Ischemia-reperfusion injury, 73-84 

comorbidities, 79 

endothelial cells, 76 

inflammatory mediators, 79 

ischemic preconditioning, 80 

leukocyte-endothelial cell interaction, 76-77 

leukocytes, 75 

modifying factors, 80 

NO, 75, 78-79 

pathophysiologic changes, 78f 

pharmacologic adjuncts, 80 

platelets, 77-78 
Israel, 27t, 405t 
Ivatury, Rao R., 223 
IVC injuries. See Inferior vena cava injuries. 



Jarrar, Doraid, 73 
Jeger, Ernst, 16 
Jerome of Brunswick, 5 
Johansen, Kaj, 125 
Johnston, Albert Sidney, 394 



Ketamine, 104 

Kidney compartment syndrome, 310 

Knee arthroplasty, 440 

Knee dislocation, 397-399 

Knee injury, 205 

Knudson, M. Margaret, 525 

Kocher maneuver 

combined Kocher and Cattell-Brassch maneuver, 
153f 

extended, 152 

extensive, 302, 307 

wide, 331 
Korean Conflict 

acute vascular trauma, 41 1 

amputation, 405t 

AV fistula, 461,462, 468t 

carotid artery injury, 229t 

importance of venous repair, 46 

location of injuries, 36t 

major vessel lesions, 495t 

MASH, 23f 

popliteal artery injury, 394, 395t 

renewal of interest in repair, 40 

surgical heritage, 18-23 

venous injury, 399 



Laceration, 54f 

Lactic acidosis, 168 

Laparoscopic surgery, 437-438 

Laparotomy, 168, 444 

LateARDS, 109 

Lateral aortorrhaphy, 303 

Lateral arteriorrhaphy, 159, 398 

Lateral circumflex femoral artery, 377f 

Lateral circumflex iliac vein, 155 

Lateral repair, 170 



INDEX. qxd 4/16/04 3:18PM Page 66 



660 



INDEX 



Lateral superior genicular artery, 377f 

Lateral thoracic artery, 364f 

Lateral venorrhaphy/arteriorrhaphy, 159 

Lateral venorrhaphy, 401 

LDUH, 528-529, 533t 

Lebanon, 27t, 395t 

Ledgerwood, Anna M., 421 

Left anterolateral thoracotomy, 292 

Left common carotid artery, 262, 359f 

Left common iliac dissection, 429f 

Left common iliac vein, 340 

Left gastric vein, 328f 

Left posterolateral thoracotomy, 139 

Left renal vein, 340f 

Left-sided medial mobilization maneuver, 301-302, 

302f, 303f 
Left subclavian artery, 359f 
Lemaire, Scott A., 251 
Lepore, Michael R., Jr., 427 

Lesions, 210. See also Traumatic vascular lesions. 
Ligation, 170 
Limb ischemia, 172-173 
Lingual artery, 226f 
LMWH, 529, 533t 

Local vascular occluding devices, 159 
Low-molecular-weight heparin (LMWH), 529, 533t 
Lower extremity. See also Extremity injuries. 

compartment syndrome, 385 

fracture, 103b 

mangled, 385-387 

treatment/repair, 154-155, 203-206 

vascular injuries, 376-389. See also Lower extremity 
vascular injuries. 

venous injuries, 387-390 
Lower extremity compartment syndrome, 385 
Lower extremity vascular injuries, 376-389 

common femoral/profunda femoral arteries, 
376-379 

lower extremity compartment syndrome, 385 

mangled lower extremity, 385-387 

popliteal and tibial arteries, 381-385 

superficial femoral artery, 379-381 

venous injuries, 387-389 
Lucas, Charles E., 421 
Lumbar artery injuries, 200-201 
Lumbar disk surgery, 441 
Lumbar veins, 31 7f 
Luminal dilatation, 187, 188f 
Luminal filling defects, 187, 189, 189f 
Luminal narrowing, 184, 186f 



Magnetic resonance. SeeMR/MRA. 

Mainlining, 422 

"Management of Venous Injuries: Clinical and 

Experimental Evaluation," 46f 
Mangled extremities, 404. See also Combined 

vascular/skeletal extremity trauma. 
Mangled lower extremity, 385-387 
MASH, 23f 

Massive hemothorax, 254 
MAST suits, 252, 253 



Mathewson, Carleton, Jr., 66, 67f 
Mattox, Kenneth L., 137, 443 
Mattox maneuver, 151, 152f 
Mechanical prophylactic devices, 528 
Mechanical ventilation, 108-110 
Mechanism of injury, 115-116 

athletic injuries, 53 

BCVIs, 242-243 

blunt trauma, 53 

children, 53 

civilian trauma, 36t 

combined extremity injuries, 406 

crutches, 53 

extremity vascular injury, 360 

fractures, 52 

historic observations, 52-54 

popliteal artery injury, 115f 

posterior dislocation of knee, 52-53 

radiation, 53-54 

vibratory tools, 54 
Medial distal thigh femoral incision, 155 
Median nerve, 365f, 368f, 373f 
Median sternotomy, 145, 264, 292 
Mediastinal traverse injuries, 264-265 
Mesenteric vein, 329 
Mesocaval shunting, 334 
Metallic fragment, 65 
Michael DeBakey International Military Surgeons 

Award, l7f 
Microvascular bleeding, 108 
Middle colic vein, 328f 
Midline laparotomy incision, 149 
Midline looping, 152 
Midshaft femur fracture, 41 If 

Military antishock trousers (MAST suits), 252, 253 
Military vascular trauma experience, 13-28 

Afghanistan War, 27t 

Balkan Wars, 13-14,488 

Bosnia, 57 

Croatia, 27t, 405t 

Grenada, 57 

Gulf War, 57 

Haiti, 57 

Ireland, 395t 

Israel, 27t, 405t 

Korean Conflict, 18-23. See also Korean Conflict. 

Lebanon, 27t, 395t 

Panama, 57 

Serbo-Bulgarian Wars, 488 

Somalia, 57 

Vietnam, 23-27. See also Vietnam Vascular Registry; 
Vietnam War. 

WWI, 14-16. See also World War I. 

WWII, 16-18. See also World War II. 
Minimal vascular injuries, 85-96 

defined, 85-87 

high-risk orthopedic injuries, 94-95 

historical overview, 87-88 

natural history, 88-91 

penetrating neck injuries, 92-94 

penetrating proximity extremity trauma, 91-92 
Misregistration artifacts, 189 



INDEX. qxd 4/16/04 3:18PM Page 661 



INDEX 661 



Mobile army surgical hospital (MASH), 23f 

Mobile wad, 447 

Money, Samuel R., 427 

Monoclonal antibodies, 80 

Monson's zones, 101, 131, 230, 231f 

Montefiore Medical Center (endovascular grafts), 

211-218 
Moore, Ernest E., 241 
MR/MRA, 184 

BCVIs, 246 

penetrating cervical injuries, 233 
Mural thrombus, 187 
Murphy,JohnB.,9-12 
Murray, James A., 339 
Musculocutaneous nerve, 365f 
Mycotic aneurysms, 423-424 
Mycotic aneurysms of carotid vessels, 425 
Myoglobinuria, 450 



Narrowing, 184 

ND, 75, 78-79, 104 

Near-infrared spectroscopy (NIRS), 449 

Neck injuries. See also Cervical injuries. 

external carotid artery, 198 

gunshot wounds, 233f, 238f 

ICA injury, 196-198 

initial care, 101 

minimal injuries, 92-94 

penetrating injuries, 92-94, 101 

treatment/repair, 143-144, 196-200 

vertebral artery, 198-199 

zones, 230, 231f 
Nephrectomy, 311 
Nephrotoxicity, 181 
Neurosurgery, 440-441 
NF-kB, 76 
Nifedipine, 184 
99m Tc-MIBI, 450 
NIRS, 449 

Nitric oxide (ND), 75, 78-79, 104 
Noninvasive physiologic vascular tests, 127-129 
Nonsuture method of bridging arterial defects, 19 



Obliterative endoaneurysmorrhaphy, 501f, 515f, 518t 
Occipital artery, 226f 
Occlusion, 191f 

angiogram, 342f 

balloon, 199, 200, 214f 

coil, 193f, 202 

conduit, 193 

defined, 191 

parenchymal, 193 

temporary balloon, 192f, 197 

tissue bed, 193 

vascular bed, 195 
Occlusion balloon, 214f 
Ohki,Takao, 207 
Omohyoid muscle, 227f 

Operating procedures. See Surgical techniques. 
Orthopedic injuries, 103b 



Orthopedic surgery, 440 
Oxyhemoglobin saturation, 449 



Ps 

acute arterial insufficiency, 126b 

ischemia, 74 
Packing, 170-171 
Palmar carpal ligament, 373f 
Palmaris longus tendon, 373f 
Palmaz stent, 209, 21 If, 214f, 21 7f, 218f 
Panama, 57 

Pancreaticoduodenal veins, 328f 
Pancreatobiliary operations, 437 
Paraplegia, 263 

Pare, Ambroise, 6, 56, 224, 286 
Parenchymal disruptions, 195 
Parenchymal occlusion, 193 
Parietal pleura, 141 

Partial thromboplastic time (PTT), 106 
Particulate embolization, 199 
PASGs, 277 

Passive external rewarming, 106 
Patch angioplasty, 162 
Patch grafting, 414 
Pathak, Abhijit S., 315 
Pathophysiology, 114-117 

abdominal injuries, 300 

AV fistula, 475-478 

BCVIs, 243 

classification, 114 

compartment syndrome, 116-117, 450 

heart, wounds of, 288-289 

ischemia, 116 

mechanism, 115-116 

reperfusion injury, 116 

thromboembolic complications, 526 
Patient history, 117-118 
Patient positioning, 138-139 
PE. See Pulmonary embolism. 
PECAM-1, 75, 76 
Pectoralis minor muscle, 368f 
Pedicle vascular injury, 201 
PEEP, 109 

Pelvic aortography, 202 
Pelvic binder, 343 
Pelvic injuries, 149-153, 201-202 
Penetrating aortic arch injuries, 263 
Penetrating cervical vascular injuries, 223-240. See also 
Neck injuries. 

angiography, 231-232 

carotid artery injuries, 233-236 

color flow Doppler, 232 

CTA, 232-233 

historical overview, 224 

incidence/etiology, 228 

initial evaluation/management, 229-300 

innominate/subclavian vessel injuries, 233, 235f 

MRA, 233 

pathology, 228-229 

physical examination, 230-231 

surgical anatomy, 224-228 



INDEX. qxd 4/16/04 3:18PM Page 662 



662 INDEX 



Penetrating cervical vascular injuries (Continued) 

treatment, 233-237 

venous injuries, 237 

vertebral artery injuries, 236-237 

zones of neck, 230, 231f 
Penetrating injuries 

cervical injuries, 223-240 

concomitant injuries, 104 

thoracic injuries, 251-267 
Penetrating mechanisms, 115 

Penetrating proximity extremity trauma, 91-92, 93f 
Penetrating thoracic vascular trauma, 251-267 

ascending aorta, 262-263 

azygos vein, 264 

catheter arteriography, 255-256 

chest radiography, 255t, 257-258f 

CTA, 256 

damage control, 259-260 

descending thoracic aorta, 263 

diagnostic studies, 255-256 

EC thoracotomy, 254 

emergency center management, 253-255 

endovascular stenting, 256-259 

foreign air embolism, 265 

history, 253 

incisions, 256t 

initial examination, 253 

innominate artery/vein, 261-262 

internal thoracic/intercostal arteries, 264 

intravenous access/fluid administration, 253-254 

left common carotid artery, 262 

mediastinal traverse injuries, 264-265 

pericardiocentesis, 254 

physical examination, 255t 

postoperative management, 265-267 

prehospital management, 252-253 

preoperative considerations, 259 

primary survey, 253-254 

pulmonary artery/vein, 263-264 

secondary survey, 254-255 

subclavian artery/vein, 260-261, 262f 

surgical repair, 259-260 

systemic air embolism, 265, 266f 

thoracic aorta, 262-263 

thoracic duct injury, 265 

thoracic inlet, 260-262 

thoracic vena cava, 264 

transverse aortic arch, 263 

treatment options, 256-260 

tube thoracostomy, 254 
Perclose device, 434, 434f 
Percutaneous vascular access, 428-437 

arterial closure devices, 431-435 

central venous access, 435 

hemorrhage, 430-431 

IABP, 435-437 

pseudoaneurysms, 429-430 
Pericardial compartment syndrome, 444, 445 
Pericardiocentesis, 254, 290 
Pericardiotomy, 254 
Pericardium, 141, 448 
Periconduit infections, 162 



Perigraft infections, 162 
Perioperative blood transfusion, 107 
Perioperative care, 104-107 

damage control, 106-107 

hypothermia, 106 

initial anesthesia, 104 

intraoperative monitoring, 80-81 

volume therapy, 105-106 
Peripheral vascular trauma. See Extremity vascular 

trauma. 
Peripheral venous injuries, 400 
Perivascular hematoma/access, 421-423 
Permissive hypercapnia, 109 
Peroneal artery, 382 
PFA injury, 204-205 
Phrenic artery, 340f 
Phrenic nerve, 227f 
Physical examination, 118 

brachial artery injury, 369 

brachiocephalic vessels, 132 

penetrating cervical injuries, 230-231 

penetrating thoracic injury, 255t 

perivascular hematoma, 422 

scapulothoracic dissociation, 363 
Physiologic envelope, 167-168 
Planned reoperation, 169, 173-174 
"Pinky", 421, 422 
Platelet function, 108 
Platelets, 106 
Platysma, 224 

Pneumatic antishock garments (PASGs), 277 
Pneumatic tourniquet, 155 
Pneumonectomy, 260 
Popliteal artery, 381f, 387 

Popliteal artery and branch injuries, 381-385, 
394-399 

diagnostic issues, 395-397 

history/ epidemiology, 394-395 

incision/access, 155, 156f 

mechanism of injury, 115f 

military experience, 395t 

nonoperative observation, 399 

posterior knee dislocation, 397-398 

surgical adjuncts, 398 

treatment, 205-206, 398-399 
Porcine xenograft/homograft, 163 
Portal vein, 327-328 
Portal vein injuries, 327-335 

associated injuries, 329-330 

exposure/initial vascular control, 330-332 

initial assessment and management, 330 

multiple vascular injuries, 332 

pancreatic division, 332 

patterns of injury, 329-330 

portal vein, 327-328 

portal vein ligation, 333-334 

postoperative management, 334-335 

retropancreatic exposure, 332 

retropancreatic wounds, 333 

splenic vein, 328-329 

stable hematoma, 330-331 

superior mesenteric vein, 328-329 



INDEX. qxd 4/16/04 3:18PM Page 663 



INDEX 663 



Portal vein injuries (Continued) 

suprapancreatic exposure, 331-332 

suprapancreatic wounds, 333 

surgical anatomy, 327-329 

treatment, 333-334 
Portal vein ligation, 333-334, 335f 
Portocaval shunting, 334 

Post-Vietnam military armed conflict, 27-28, 57 
Positioning, 138-139 

Positive end-expiratory pressure (PEEP), 109 
Posterior cerebral artery, 226f 
Posterior circumflex humeral artery, 364f 
Posterior communicating artery, 226f 
Posterior dislocation of knee, 52-53 
Posterior intrapericardial inferior vena cava, 296f 
Posterior knee dislocation, 397-399 
Posterior tibial artery, 381f, 382 
Posterior ulnar recurrent artery, 372f 
Postoperative limb ischemia, 172-173 
Postoperative priorities, 107-112 

antibiotics, 110 

assessment/determination of take back, 
110-112 

blood transfusion, 107, 108, 109b 

coagulation monitoring, 107-108 

hemodynamic monitoring, 107 

mechanical ventilation, 108-110 
Preconditioning, 80 
Prednisone, 181 
Primary survey, 98-100 
Principle of extensile exposure, 149, 155 
Pringle maneuver, 153, 324 
Procoagulant activity, 108 
Profunda femoral artery, 204-205, 376 
Profunda femoral artery injury, 376-379 
Pronator teres muscle, 373f 
Prone ventilation, 110 
Prophylactic fasciotomy, 130, 133 
Prophylactic inferior vena cava filters, 350 
Prophylactic vena cava filters, 529-530, 533t 
Prosthetic grafts, 414-415 
Prosthetic vascular graft, 266 
Prothrombin time (PT), 108 
Proximal control, 140 
Proximal embolization, 199 
Proximal renal arteries, 307 
Pseudoaneurysms, 429-430 
PT, 108 
PTFE (Dacron) graft 

BTAI, 278, 279 

common femoral artery injury, 378 

effectiveness, 415 

iliac arterial injuries, 345 

infrarenal abdominal aorta, 308 

profunda femoral artery injury, 378 

substitute conduit, 161, 162 

superficial femoral artery injury, 380 

suprarenal abdominal aorta, 304 

vascular reconstruction, 424 
PTT, 106 

Pullout abdominal aortogram, 312 
Pulmonary artery/vein injury, 263-264 



Pulmonary embolism. See also Thromboembolic 
complications. 

diagnosis, 531 

heparin, 531 

IVC injuries, 326 
Pyloric vein, 328f 



Quadrangulation method, 13f 



Radial and ulnar artery injury, 371-374 
Radial artery, 371, 372f, 373f 
Radial collateral artery, 372f 
Radial nerve, 365f, 368f, 373f 
Radial recurrent artery, 372f 

Repair principles/techniques, 156-163. See also Surgical 
treatment. 

ligation, 170 

packing, 170-171 

step 1 (control), 156 

step 2 (explore the injury), 156 

step 3 (enter the hematoma), 156-157 

step 4 (vascular damage control), 157 

step 5 (debridement), 157 

step 6 (type of repair required), 157 

step 7 (stay sutures), 157 

step 8 (Fogarty catheters), 157, 158b 

step 9 (heparin), 157-159 

step 10 (local vascular occluding devices), 159 

step 11a (simple repair techniques), 159-160 

step lib (end-to-end repair), 160-161 

step lie (substitute conduit), 161-162 

step lid (patch angioplasty), 162 

step 12 (flush), 162 

step 13 (complete suture line/clamp removal), 162 

step 14 (distal circulation), 163-164 

step 15 (fasciotomy), 163 

step 16 (coverage), 163 
Radiation, 53-54 

Radical hepatic mobilization, 321 
Radiographic studies. See Imaging techniques. 
Rapid infusion systems, 99 
References, 537-649 
Renal artery, 201 
Renal artery injury, 307, 309-312 
Renal revascularization, 310-311 
Reperfusion injury, 116. See also Ischemia-reper fusion 

injury. 
Resuscitation, 99, 169 
Resuscitative thoracotomy, 147 

Retrohepatic vena caval injuries, 321, 323-324, 325 
Retropancreatic confluence zone, 327 
Retroperitoneal hematoma, 432f 
Retroperitoneum, 431 
Rewarming, 106 
Rice, David, 285 
Rich, Norman M., 3, 457 
Right common carotid artery, 359f 
Right common iliac artery, 340, 377f 
Right common iliac vein, 340 
Right decubitus position, 138f 



INDEX. qxd 4/16/04 3:18PM Page 664 



664 INDEX 



Right gastroepiploic vein, 328f 
Rob, Charles G., 2 If, 60f 
Rodriguez, Aurelio, 269 
Rufus of Ephesus, 5, 6 



Sandbags, 182 

Scalenus anticus muscle, 359f, 360f 

Scalenus medius muscle, 359f, 360f 

Scapulothoracic dissociation, 191f, 363-364 

Scintigraphy, 450 

Sclafani, Salvatore J. A., 179 

Scoliosis, 441 

Scott, Bradford G., 165 

Screw tourniquet, 6 

Secondary survey, 100 

Seeley, Sam, 23 

Segmental veins, 3l7f 

Selective reconstructive endoaneurysmorrhaphy, 516f 

Self-tamp on ade, 318 

Septic phlebitis, 425 

Serial venography, 42 

Seven Ps of acute arterial insufficiency, 126 

SFA, 204, 205, 377f, 379 

SFA injury, 204, 205 

Shackford, Steven R., 353 

Sheaths, 428, 434f 

Sherman, H. M., 286 

Shotgun wound. See Gunshot wound. 

Shumacker, Harris, 19f 

Shunt/shunting 

BTA1, 278 

combined extremity injuries, 409 

Gott shunt, 278 

popliteal artery injury, 399 

portal vein, 334 

temporary shunts, 171-172 
Silastic intraluminal shunts, 409 
Simple vascular repair techniques, 159-160, 170 
Sise, Michael J., 353 
Site of injury, 47-48 
Small branch injuries, 200 
Soft signs 

arterial injury, lOOt 

vascular injury, 118-119, 229, 356t 
Soltero, Ernesto, 285 
Somalia, 57 

Soubbotitch, V., 13, 14f 
Spinal compartment syndrome, 444 
Spinal cord, 448 

Spiral computed tomographic angiography, 356 
Splenic arteriography, 193f 
Splenic artery, 201 
Splenic vein, 328f, 329 
Sponge stick, 154 
Spontaneous cure 

AVF, 486-488 

false aneurysms, 520 
Spontaneous tamponade, 318 
Stab wound 

cardiac trauma, 293, 294, 294f 

etiology, 38t, 39t 



Stab wound (Continued) 

iliac vessel injury, 341 1 

IVC injury, 318 

location, 38t 

minimal injury, 89t, 235 
Stable hematoma, 330 
Stay sutures, 157 
Stent/ stent grafts, 195-196 

brachiocephalic vessel injury, 200 

ICA injury, 198 

iliac vessel injury, 348-349 

Palmaz stent, 209, 211f, 214f, 2l7f, 218f 

penetrating thoracic injuries, 256-259 

thoracic vascular injuries, 199 

uncovered/ covered, 195 
Steri-Strips, 453 

Sternocleidomastoid muscle, 224, 227f 
Sternohyoid muscle, 227f 
Sternothyroid muscle, 227f 
Stocking-glove distribution sensory deficit, 116 
Stoner, Michael C., 223 
Stretch injuries, 201 
Stryker pressure monitor, 449 
Subclavian artery, 436f 
Subclavian vein, 436f 
Subclavian vein/artery 

arterial injuries, 359-364 

balloon tamponade, 260f 

blunt thoracic injuries, 279-280 

mortality, 363 

penetrating cervical injuries, 233, 235f 

penetrating thoracic injuries, 260-261, 262f 

results, 362-363 

scapulothoracic dissociation, 363-364 

surgical anatomy, 359-360 

surgical treatment, 361-362 
Subclavian vein/artery fistula, 436f 
Subclavian venous access, 435 
Subscapular artery, 364f 
Substitute interposition conduit, 161-162 
Subtraction angiography, 271-272 
Subtraction aortography, 257f 
Superficial epigastric artery, 377f 
Superficial femoral artery (SFA), 204, 205, 377f, 379 
Superficial femoral artery injury, 379-381 
Superficial femoral vein, 379 
Superficial iliac circumflex artery, 377f 
Superficial radial nerve, 373f 
Superficial temporal artery, 226f 
Superficial volar branch radial artery, 372f 
Superior collateral artery, 368f 
Superior gluteal artery, 377f 
Superior lateral genicular artery, 381f 
Superior medial genicular artery, 381f 
Superior mesenteric artery, 305-307 
Superior mesenteric vein, 328f, 329 
Superior thoracic artery, 364f 
Superior thyroid artery, 226f, 227f 
Superior ulnar collateral artery, 368, 368f, 372f 
Superior vena cava injuries, 264 
Superoxide, 78, 453 
Supine decubitus position, 138f 



INDEX. qxd 4/16/04 3:18PM Page 665 



INDEX 



665 



Supraclavicular incision, 361 
Suprahepatic vena cava, 280 
Supramesocolic hematoma, 302 
Suprapancreatic portal vein, 331-333 
Suprarenal abdominal aorta, 301-304 
Surgical anatomy 

axillary artery injuries, 364 

BCVIs, 242 

brachial artery injuries, 367-368 

common fern oral /profunda femoral arteries, 376, 
377f 

iliac vessel injuries, 340-341 

IVC injuries, 317-318 

penetrating cervical injuries, 224-228 

popliteal/ tibial arteries, 381-382 

portal vein injuries, 327-329 

radial/ulnar artery injury, 371, 372f 

subclavian artery injuries, 359-360 

superficial femoral artery, 379 

venous injuries (lower extremity), 387 
Surgical techniques, 137-164. See also Surgical 
treatment. 

abdomen/pelvis, 149-153 

axillary artery, 148 

brachial artery, 148-149 

Cattell-Braasch maneuver, 152, 153f 

chest, 146-148 

colorectal procedures, 437 

decompression, 451-453 

distal superficial femoral artery, 155 

groin, 154-155 

hemorrhage control, 139-142 

high-risk injuries, 143b 

incisions, 140b, 143b. See also Incisions. 

laparoscopic procedures, 437-438 

lower extremity, 154-155 

Mattox maneuver, 151, 152f 

neck, 143-144 

neurosurgery, 441-442 

orthopedic surgery, 440 

pancreatobiliary procedures, 437 

percutaneous vascular access, 428-437 

popliteal artery, 155, 156f 

positioning, 138-139 

Pringle maneuver, 153 

repair, 156-163. See also Repair 
principles/techniques. 

thoracic outlet, 144-146 

upper extremity, 148-149 

vascular/ endovascular surgery, 438-440 
Surgical treatment. See also Repair 

principles/techniques; Surgical techniques. 

abdominal vascular injury, 200-201 

arm injury, 202-203 

AVF, 484-486 

axillary artery injuries, 362t, 366-367 

balloon catheter tamponade, 170, l7lf 

BCVIs, 246-249 

blunt thoracic injuries, 277-278 

brachial artery injuries, 362t, 369-371 

brachiocephalic vessels, 199-200 

calf injury, 205 



Surgical treatment (Continued) 

clamp-and-sew technique, 277-278 

combined extremity injuries, 408-412 

common femoral/profunda femoral arteries, 
378-379 

extremity compartment syndrome, 450-453 

external carotid artery injury, 198 

extremity vascular trauma, 357-359 

false aneurysms, 513-520 

femoral arteries, 204-205 

groin injury, 203-204 

hip injury, 203-204 

ICA injuries, 196-198 

iliac vessel injuries, 344-348 

knee injury, 205 

lateral repair, 170 

ligation, 170 

lumbar artery injury, 200-201 

lower extremity, 203-206 

neck, 196-200 

packing, 170-171 

pedicle vascular injury, 201 

pelvic injury, 201-202 

penetrating cervical injuries, 233-237 

penetrating thoracic injuries, 259-260 

planned reoperation, 169, 173-174 

popliteal artery & branches, 205-206 

popliteal/tibial arteries, 383-385 

portal vein ligation, 333-334 

radial/ulnar artery injuries, 362t, 372-373 

simple/complex repairs, 170 

small branch injuries, 200 

stretch injuries, 201 

subclavian artery injuries, 361-362, 362t 

superficial femoral artery, 380 

thigh injury, 205 

thoracic vascular injuries, 199-200 

upper extremity, 202-203 

vascular repair techniques, 170-171 

venous injuries, 401-404 

venous injuries (lower extremity), 388 

venous ligation, 403-404 

vertebral artery injury, 198-199 
Swan-Ganz catheter, 107 
Swan-Ganz monitoring, 265 
Synthetic conduits, 162 
Systemic air embolism, 265, 266f 

""Tc-MIBI, 450 

TCD studies, 132 

Technetium-99 methoxyisobutyl isonitrile 

( 99m Tc-MIBI),450 
TEE, 275-276, 295 

Temporary balloon occlusion, 192f, 197 
Temporary hemostasis, 192 
Temporary intraluminal arterial shunt, 66f 
Temporary shunts, 171-172 
10/30 rule, 107 

Testicular (ovarian) artery, 340f 
Testicular (ovarian) vein, 340f 
Thigh compartment syndrome, 385, 446 



INDEX. qxd 4/16/04 3:18PM Page 66 6 



666 INDEX 



Thigh injury, 205 
Thoracic aorta injury, 262-263 
Thoracic duct injury, 265 
Thoracic injuries, 101-102 

blunt injuries, 269-283. See also Blunt thoracic 
vascular trauma. 

operative principles, 146-148 

penetrating injuries, 2521-267. See also Penetrating 
thoracic vascular trauma. 
Thoracic inlet, 2630-2632 
Thoracic outlet injury, 144-146 
Thoracic vascular injuries, 199-200 
Thoracic vena cava, 280 
Thoracic vena cava injury, 264 
Thoracoabdominal aortic operations, 439 
Thoracoabdominal exposure of aorta, 439f 
Thoracoacromial artery, 364f 
Thoracotomy 

anterolateral, 146, 147, 292 

digital, 290 

EC, 100, 254, 290 

left anterolateral, 292 

left posterolateral, 139 

resuscitative, 147 
Thromboelastography, 108 
Thromboembolic complications, 525-535 

algorithm, 534f 

clinical epidemiology/risk factor analysis, 526-527 

diagnosis/ treatment, 530-533 

historical overview, 525-526 

LDUH, 528-529 

LMWH, 529 

mechanical prophylactic devices, 528 

outcomes research, 532-533 

pathophysiology, 526 

prophylactic measures, 527-528 

prophylactic vena cava filters, 529-530 

recommendations/future directions, 533-535 
Thyrocervical trunk, 200, 359f, 360f 
Thyrohyoid muscle, 227f 
Tibial arterial injuries, 381-385 
Tibial artery, 382 
Tissue bed occlusion, 193 
Tissue manometry, 130 
Topical hemostasis, 142 
Total hip arthroplasty, 440 
Total knee arthroplasty, 440 
Tourniquet, 6 
Trachea, 227f 

Transcatheter embolization. See Embolization. 
Transcranial Doppler (TCD) studies, 132 
Transection, 54f 

Transfusion. See Blood transfusion. 
Transesophageal echocardiography (TEE), 275-276, 

295 
Transversalis fascia, 448 
Transverse aortic arch, 263 
Trauma surgery. See Surgical techniques; Surgical 

treatment. 
Traumatic false aneurysms, 54f, 484f, 499-521 

Angiography, 61f, 510, 512f, 513f 

clinical features, 510 



Traumatic false aneurysms (Continued) 

clinical pathology, 506-510 

closed fractures, 504t 

diagnostic considerations, 510-513 

etiology, 503-506 

follow-up, 520-521 

historical overview, 499-502 

incidence, 502-503 

incisions, 51 7f 

e n do aneurysm or rhaphy, 500, 501f, 515f, 516f, 
518t 

peripheral nerve lesions, 508t 

spontaneous cure, 520 

surgical treatment, 513-520 

Vietnam Vascular Registry, 464t, 465t, 469t, 47lt, 
495t, 496t, 508f 

World War II, 51 7f, 518t, 519t 
Traumatic hemothorax, 254 
Traumatic vascular lesions, 207-220 

background, 208-211 

characteristics of lesions/outcome, 210f 

coil embolization, 208 

intraluminal balloon control of arteries, 208 

intravascular stents, 208 

Montefiore experience, 211-218 
Treatment. See Surgical techniques; Surgical treatment. 
Triangulation method of suturing, 12f 
Tube thoracostomy, 254, 290 



Ulnar artery, 371, 372f, 373f, 447f 
Ulnar nerve, 365f, 368f, 44 7f 
Ultrasound. See Duplex ultrasonography. 
Ultrasound-guided pseudoaneurysm compression, 430 
Uncovered stents, 195 
Uncross-matched type O blood, 105 
Unfractioned heparin (LDUH), 528-529, 533t 
Upper extremity. See also Extremity injuries, 
compartment syndrome, 375 

fracture, 103b 

treatment/repair, 148-149, 202-203 

venous injury, 374-375 
Ureter, 340f 



Vagus nerve, 22 7f 

Vascular anastomosis, 159-161 

Vascular bed occlusion, 195 

Vascular/ en dovascular surgery, 438-440 

Vascular grafts, 414-416 

Vascular patency, 111 

VasoSeal device, 432, 433f 

Vasospasm, 181, 184, 186f 

Vein graft, 209 

Vein transplantation, 492t 

Veith, Frank J., 207 

Vena cava filters, 326, 350, 529-530, 533t 

Vena comitans, 368f 

Venization, 477 

Venogram/venography, 43f, 44f, 400, 404 

Venous access, 435 

Venous aneurysms, 425 



INDEX. qxd 4/16/04 3:18PM Page 667 



INDEX 



667 



Venous injuries, 399-404 

diagnosis, 400-401 

history/ epidemiology, 399-400 

iliac vessel injuries, 346-347 

ligation, 403-404 

lower extremity, 387-389 

penetrating cervical injuries, 237 

treatment, 401-404 

upper extremity, 374-375 
Venous ligation, 403-404 
Venous patency, 388 
Venous repair, 401-404 
Venovenous bypass, 324 
Ventricular septal defects, 294, 295f 
Veress needle, 438, 439f 
Vertebral artery, 225, 226f, 359f, 360f 
Vertebral artery injury, 198-199, 236-237 
Veterinarian's stitch, 8f 
Vibratory tools, 54 

Vietnam Vascular Registry, 24, 228, 458. See also 
Vietnam War. 

AV fistula, 464t, 465t, 469t, 47lt, 495t, 496t, 497 

exhibit, 26f 

extremity venous injuries, 27t 

false aneurysm, 464t, 465t, 469t, 471 1, 495t, 496t, 508f 

importance, 224 

preliminary report, 26f, 41, 42t 

pulmonary emboli, 46 

studies, 43 

venous ligation, 399 

wrist pressure, 64f 
Vietnam War 

amputation, 405t 

AV fistula, 461,462 

cardiac trauma, 287, 287t 

carotid artery injury, 229t 

combined extremity injuries, 405, 412 

follow-up. See Vietnam Vascular Registry. 

interposition grafting, 414 

location of injuries, 36t 

popliteal artery injury, 394, 395t 

prosthetic grafts, 414 

surgical heritage, 25-27 

venous injuries, 399, 400, 402 
Virchow triad, 526 
Visceral aorta, 301, 302f 
Volar interosseous artery, 372f 
Volkmann's ischemic contracture, 445 
Volume therapy, 105-106 



Walking the clamps, 154 

Wall, Matthew J., Jr., 251, 285 

Wallgraft, 196f, 211, 212 

Wang, Ping, 73 

War. See Military vascular trauma experience. 

Wide Kocher maneuver, 331 

World War I, 14-16 

carotid artery injury, 229t 

location of injuries, 36t 

popliteal artery injury, 394, 395t 

venous injury, 399 
World War II, 16-18 

AF fistula, 463t, 488-489, 490t 

amputation, 405t 

carotid artery injury, 229t 

false aneurysm, 5l7f, 518t, 519t 

location of injuries, 36t 

Matas endoaneurysmorrhaphy, 518t 

popliteal artery injury, 394, 395t 

venous injury, 399 
Wounds of the heart, 285-297 

aortic occlusion, 292 

cardiac manipulation, 292 

cardiac septal injuries, 294-295 

cardiac valvular injury, 295 

clinical pathology, 287-288 

complex injuries, 293-294 

EC thoracotomy, 290-291 

emergency center, 289-291 

hemorrhage control, 292-293 

historical overview, 285-286 

incidence, 286-287 

incisions, 291-292 

i n trap eri cardial inferior vena cava injury, 
295-296 

operative management, 291-296 

pathophysiology, 288-289 

prehospital management, 289 



X-rays, 119 

Xanthine oxidase-derived oxidants, 78 

Xanthine oxidase inhibitors, 453 



Zone I neck wounds, 131, 230, 231f 
Zone II neck wounds, 101, 131, 230, 231f 
Zone III neck wounds, 101, 131, 230, 231f