Skip to main content

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

See other formats


Mo tries Metta 



Colleen M.Brophy Editors 



\ 


I 1 


,) 


tmm 







M 



Lumley Series Editor 








w 




Springer 
Specialist 
Surgery 
Series 



Other titles in this series include: 

Upper Gastrointestinal Surgery, edited by Fielding & Hallissey, 2005 
Neurosurgery: Principles and Practice, edited by Moore & Newell, 2004 
Transplantation Surgery, Edited by Hakim & Danovitch, 2001 



Alun H. Davies and Colleen M. Brophy (Eds) 



Vascular Surgery 



With 46 Illustrations 



^Spri 



ringer 



Alun H. Davies, MA, DM, FRCS Colleen M. Brophy, MD, FACS 

Department of Vascular Surgery Chief of Vascular Surgery 

Reader and Honorary Consultant in Surgery Carl T. Hayden VAMC 

Imperial College London Research Professor Bioengineering 

Charing Cross Hospital Arizona State University 

London, UK Clinical Professor of Surgery 

University of Arizona 

Phoenix, AZ 

USA 



A catalogue record for this book is available from the British Libuary 

Library of Congress Control Number: 2005923614 

ISBN-10: 1-85233-288-3 Printed on acid-free paper. 

ISBN-13: 978-1-85233-288-4 

© Springer- Verlag London Limited 2006 

Apart from any fair dealing for the purposes of research or private study, or criticism, or review, as permitted 
under the Copyright, Designs and Patents Act 1988, this publication may only be reproduced, stored or trans- 
mitted, in any form or by any means, with the prior permission in writing of the publishers, or in the case of 
reprographic reproduction in accordance with the terms of licenses issued by the Copyright Licensing Agency. 
Inquiries concerning reproduction outside those terms should be sent to the publishers. 
The use of registered names, trademarks, etc, in this publication does not imply, even in the absence of a specific 
statement, that such names are exempt from the relevant laws and regulations and therefore free for general use. 
Product liability: The publisher can give no guarantee for information about drug dosage and application 
thereof contained in this book. In every individual case the respective user must check its accuracy by con- 
sulting other pharmaceutical literature. 

Printed in the United States of America. (BS/MVY) 

987654321 

Springer Science+Business Media 
springeronline.com 



Preface 



This book provides coverage of a broad range of topics in the field of vascular surgery to 
residents, registrars in training, and to recent graduates of training programs. The book 
is meant to be a practical rendition of the basic knowledge and clinical management 
required for optimal care of vascular surgical patients. Each chapter contains input from 
specialists in vascular surgery from the United States and Great Britain. There are up- 
to-date perspectives on common clinical conditions and emerging techniques encoun- 
tered by vascular surgeons from both an American and British perspective. The chapters 
are organized under broad topics including medical management, noninvasive and inva- 
sive diagnostic approaches, perioperative care, indications and approaches for vascular 
procedures, and a discussion of newer endovascular techniques. The information con- 
tained in this text is not meant to be exhaustive, but rather a practical overview that will 
be useful in directing the management of patients with vascular diseases. The informa- 
tion in this text is also meant to be useful for certification examinations and recent grad- 
uates of vascular surgical training programs can utilize this text as an update of the most 
important vascular topics. 

Alun H. Davies 
Colleen M. Brophy 



Contents 



Preface v 

Contributors ix 

1. The Epidemiology and Etiology of Atherosclerosis 

Paul B. Kreienberg, R. Clement Darling III, and F.G.R. Fowkes 1 

2. Clinical Evaluation of Patients with Vascular Disease 

William G. Tennant 9 

3. Noninvasive Vascular Examination 

Colleen M. Brophy 19 

4. Radiological Investigations 

Steven M. Thomas, Kong T. Tan, and Mark F. Fillinger 25 

5. Bleeding and Clotting Disorders 

Vivienne J. Halpern and Frank C.T Smith 39 

6. Medical Management of Peripheral Arterial Disease 

Jill J.F. Belch and Andrew H. Muir 53 

7. Anesthesia for Vascular Surgery 

Jamal J. Hoballah and Farid Moulla 65 

8. Nonatherosclerotic Vascular Disease 

Jonathan R.B. Hutt and Alun H. Davies 73 

9. Lower Limb Ischemia 

Rajabrata Sarkar and Alun H. Davies 91 

vii 




> 



CONTENTS 



10. Chronic Venous Insufficiency, Varicose Veins, Lymphedema, 
and Arteriovenous Fistulas 

Andrew W. Bradbury and Peter J. Pappas 105 

1 1 . Vascular Trauma 

Kathleen J. Ozsvath, R. Clement Darling III, Laila Tabatabai, 

Sacha Hamdani, Alun H. Davies, and Meryl Davis 125 

12. Complications in Vascular Surgery 
Jeremy S. Crane, Nicholas J.W. Cheshire, and 

Gilbert R. Upchurch, Jr. 133 

13. Vascular Access 

David C. Mitchell and C. Keith Ozaki 141 

14. Outcome Measures in Vascular Surgery 

Christopher J. Kwolek and Alun H. Davies 149 

15. Carotid Artery Disease 

A. Ross Naylor, Peter H. Lin, and Elliot L. Chaikof 155 

16. Arch Vessel, Vertebrobasilar, and Upper Extremity 

Eva M. Rzucidlo and A. Ross Naylor 181 

17. Aneurysmal Disease 

Philip Davey and Michael G. Wyatt 191 

18. Renovascular Hypertension and Ischemic Nephropathy 

Sherry D. Scovell 221 

19. Visceral Ischemic Syndromes 

George Geroulakos, Peter A. Robless, and William L. Smead 231 

20. Endovascular Approaches and Techniques 

Steven M. Thomas, Kong T Tan, and Mark F. Fillinger 237 

Index 253 



Contributors 



Jill J.F. Belch, MB ChB, MD, FRCP 
Peripheral Vascular Diseases Research Unit, 
Department of Medicine, Ninewells Hospital 
and Medical School, Dundee, UK 

Andrew W. Bradbury, BSc, MB ChB, MD, FRCS 
University Department of Vascular Surgery, 
Research Institute, Birmingham Heartlands 
Hospital, Birmingham, UK 

Colleen M. Brophy, MD, FACS 

Chief of Vascular Surgery, Carl T. Hayden 

VAMC, Research Professor Bioengineering 

Arizona State University, Clinical Professor of 

Surgery, University of Arizona, Phoenix, AZ, 

USA 



Elliot L. Chaikof, MD, PhD, FACS 
Division of Vascular Surgery, Brown 
Whitehead Department of Surgery, Emory 
University School of Medicine, Atlanta, GA, 
USA 



Nicholas J.W. Cheshire, MB ChB, MD, FRCS 
Regional Vascular Unit, St Mary's Hospital, 
London, UK 



Jeremy S. Crane, MB ChB, MRCS 
Regional Vascular Unit, St Mary's Hospital, 
London, UK 



R. Clement Darling III, MD 

Institute for Vascular Health and Disease, 

Albany Medical Center, Albany, NY, USA 



Philip Davey, MB ChB 
Newcastle upon Tyne, UK 

Alun H. Davies, MA, DM, FRCS 
Department of Vascular Surgery, Reader and 
Honorary Consultant in Surgery, Imperial 
College, Charing Cross Hospital, London, UK 
London 

Meryl Davis 

Charing Cross Hospital, London, UK 

Mark F. Fillinger, MD, FACS 
Department of Vascular Surgery, Dartmouth 
Medical School, Dartmouth-Hitchcock 
Medical Center, Lebanon, NH, USA 

F.G.R. Fowkes, MB ChB, PhD, FRCPE, FFPHM 
Wolfson Unit for Prevention of Peripheral 
Vascular Diseases, Public Health Sciences, The 
University of Edinburgh, Edinburgh, UK 

George Geroulakos, MD, FRCS, DIC, PhD 
Vascular Unit, Ealing Hospital, London, UK 

Vivienne J. Halpern, MD, FACS 
Department of Surgery, Division of Vascular 
Surgery, Long Island Jewish Medical Center, 
New Hyde Park, NY, USA 

Sacha Hamdani 

Institute for Vascular Health and Disease, 

Albany Medical Center, Albany, NY, USA 

Jamal J. Hoballah.MD 

Department of Surgery, Division of Vascular 
Surgery, University of Iowa Hospitals and 
Clinics, Iowa City, IA, USA 




> 



CONTRIBUTORS 



Jonathan R.B. Hutt, BA, MBBS 
Department of Accident and Emergency, 
Imperial College, Charing Cross Hospital, 
London, UK 

Paul B. Kreienberg, MD 

Institute for Vascular Health and Disease, 

Albany Medical Center, Albany, NY, USA 

Christopher J. Kwolek, MD, FACS 

Division of Vascular Surgery, Massachusetts 

General Hospital, Boston, MA, USA 

Peter H. Lin, MD, FACS 
Division of Vascular Surgery and 
Endovascular Therapy, Michael E. DeBakey 
Department of Surgery, Baylor College of 
Medicine, Houston, TX, USA 

David C. Mitchell, MA, MS, FRCS 
Department of Surgery, Southmead Hospital, 
North Bristol NHS Trust, Bristol, UK 

Farid Moulla, MD, MBA 

Department of Anaesthetics, Charing Cross 

Hospital, London, UK 

Andrew H. Muir, MB ChB 
Peripheral Vascular Diseases Research Unit, 
Department of Medicine, Ninewells Hospital 
and Medical School, Dundee, UK 

A. Ross Naylor, MB ChB, MD, FRCS 
Department of Vascular Surgery, Leicester 
Royal Infirmary, Leicester, UK 

C. Keith Ozaki, MD, FACS 
Division of Vascular Surgery and 
Endovascular Therapy, University of Florida 
College of Medicine, Gainesville, FL, USA 

Kathleen J. Ozsvath, MD 

Institute for Vascular Health and Disease, 

Albany Medical Center, Albany, NY, USA 

Peter J. Pappas, MD 

Department of Surgery, Section of Vascular 
Surgery, University of Medicine and Dentistry 
of New Jersey - New Jersey Medical School, 
Newark, NJ, USA 



Peter A. Robless, MB ChB, FRCS, MD, FEBVS 
Department of Cardiac, Thoracic and Vascular 
Surgery, National University Hospital, 
Singapore, Republic of Singapore 

Eva M. Rzucidlo, MD 

Department of Vascular Surgery, Dartmouth- 
Hitchcock Medical Center, Lebanon, NH, 
USA 

Rajabrata Sarkar, MD, PhD 

Division of Vascular Surgery, University of 

California, San Francisco, CA, USA 

Sherry D. Scovell, MD 

Division of Vascular Surgery, Beth Israel 

Deaconess Medical Center, Boston, MA, USA 

William L. Smead, MD 

Division of General Vascular Surgery, Ohio 

State University, Columbus, OH, USA 

Frank C.T. Smith, BSc, MD, FRCS 

University Department of Surgery, University 

of Bristol, Bristol Royal Infirmary, Bristol, UK 

Laila Tabatabai 

Institute for Vascular Health and Disease, 

Albany Medical Center, Albany, NY, USA 

Kong T. Tan, MB ChB, BAO, FRCSI, FRCR 
Sheffield Vascular Institute, Vascular Office, 
Northern General Hospital, Sheffield, UK 

William G. Tennant, BSc, MB ChB, MD, FRCS 
Department of Vascular Surgery, Queens 
Medical Centre, Nottingham, UK 

Steven M. Thomas, MRCP, FRCR, MSc 
Sheffield Vascular Institute, Vascular Office, 
Northern General Hospital, Sheffield, UK 

Gilbert R. Upchurch, Jr., MD 
Surgery Department, Vascular Surgery 
Section, University of Michigan Health 
System, Ann Arbor, MI, USA 

Michael G. Wyatt, MB BS, MSc, MD, FRCS 
Department of Vascular Surgery, Freeman 
Hospital, Newcastle upon Tyne, UK 



1 



The Epidemiology and Etiology 
of Atherosclerosis 

Paul B. Kreienberg, R. Clement Darling III, 
and F.G.R. Fowkes 




The underlying disorder in the vast majority of 
cases of cardiovascular disease is atherosclero- 
sis, for which low-density lipoprotein (LDL) 
cholesterol is recognized as a major risk factor. 
Evidence from epidemiological and clinical 
studies continues to improve our understanding 
of the pathogenesis of atherosclerosis. Athero- 
sclerosis contributes to myocardial infarction, 
stroke, and peripheral vascular disease. Despite 
major advances in the development of diagnos- 
tic methods and effective treatments, cardiovas- 
cular disease remains the leading cause of 
mortality in the Western world. 

Vascular surgeons treat patients who have 
already developed end-stage cardiovascular 
disease. These surgeons have a unique opportu- 
nity to intervene not only in the arterial pathol- 
ogy itself, but also in the main factors that 
contribute to the development of atherosclero- 
sis. Atherosclerosis is a systemic disorder and all 
aspects of the disease must be addressed in 
treating these patients. 



Epidemiology 



Cardiovascular disease is responsible for 
approximately 30% of all mortality worldwide, 
amounting to approximately 15 million deaths 
(Sueta et al., 1999). Furthermore, cardiovascular 
disease is the principal cause of mortality in all 
developed countries, responsible for 50% of all 
deaths, and is also emerging as a prominent 
public health problem in developing countries, 



representing approximately 16% of all deaths. In 
the United States cardiovascular disease is the 
most common cause of mortality in both men 
and women and accounts for almost 500,000 
deaths/year. Additionally, 58,800,000 Americans 
have one or more types of cardiovascular 
disease according to current estimates (Table 
1.1). 

Coronary artery disease (CAD) has been rec- 
ognized as the leading cause of death since the 
late 1940s. Hypertension, hypercholesterolemia, 
cigarette smoking, and diabetes mellitus were 
identified as key contributors to atherosclerosis 
and the development of cardiovascular risk. 
These risk factors are also related to cere- 
brovascular and peripheral vascular disease 
(Table 1.2). 

The data support aggressive treatment of ath- 
erosclerosis in populations at risk. In patients 
with peripheral arterial disease (PAD), there 
is a high prevalence of myocardial infarction, 
stroke, and increased mortality. Lack of patient 
and physician awareness of peripheral vascular 
disease is associated with low atherosclerosis 
risk factor treatment intensity (Sueta et al., 
1999). In this study of patients with known 
cardiovascular disease, only 35% had smoking 
behavior treated, 73% had lipid abnormal- 
ities treated, and 71% were on antiplatelet 
therapy. Recognition and treatment in patients 
with symptomatic or asymptomatic [ante- 
brachial index (ABI) <0.9] PAD were 
significantly lower than the rates for patients 
with CAD. 



1 




> 



VASCULAR SURGERY 



Table 1.1. Cardiovascular Disease in the United States: 
58.8 million Americans have one or more types of 
cardiovascular disease 



Type 


Number (in millions) 


Hypertension 


50 


Coronary artery disease 


12 


Stroke 


4.4 


Congestive heart failure 


4.6 


Peripheral arterial disease 


8.4 



Table 1 .2. Selected risk factors for atherosclerosis 

Age 
Diabetes 

I Smoking 
Hyperlipidemia 
Hypertension 
Hyperhomocystinemia 
Hyperfibrinogenemia 



Risk Factors 

Smoking 

Nearly 440,000 Americans die each year of 
smoking-related illness, at a cost of about $50 
billion annually. In general, smoking is associ- 
ated with a threefold increase in the risk for 
peripheral atherosclerosis (Hiatt et al., 1995). 
Two large follow-up studies of patients with 
intermittent claudication demonstrate the 
benefits of smoking cessation (Jonason and 
Bergstrom, 1987; Smith et al, 1996). In these 
studies, 11% to 27% of the patients complied 
with the advice to stop smoking. Within 3 years 
of stopping, there was no reduction in limb- 
threatening complications of the vascular 
disease. However, after 7 years, rest pain had 
developed in 16% of persistent smokers, but in 
none of those who had stopped smoking. After 
10 years, 53% of persistent smokers suffered a 
myocardial infarction compared to only 11% of 
stopped smokers; 54% of persistent smokers 
died compared to 18% of stopped smokers. In a 
comprehensive review of the literature, absti- 
nence from smoking was found to be associated 
consistently with better outcomes following 
revascularization, lower amputation rates, and 
improved survival (Hirsch et al., 1997). 
However, smoking cessation had probably only 



a minimal effect in improving walking distance 
in claudicants. 



Hyperlipidemia 

An estimated 50% of American adults have total 
blood cholesterol levels of 200mg/dL and 
higher, and about 20% of American adults 
have levels of 240mg/dL or higher. Levels of 
240mg/dL or higher are considered high risk 
and levels from 200 to 239mg/dL are considered 
borderline high risk. Evidence linking lipids to 
atherosclerosis has grown, suggesting that low- 
ering serum cholesterol, whether through diet 
and lifestyle modification alone or in combina- 
tion with cholesterol-lowering pharmacother- 
apy, decreases the incidence of vascular events. 
For example a 1 mg/dL increase in high-density 
lipoprotein (HDL) cholesterol concentration is 
associated with a 2% to 3% decrease in CAD and 
a 4% to 5% decrease in cardiovascular mortal- 
ity. Data from the Multiple Risk Factor Inter- 
vention Trial (MRFIT) demonstrate a strong, 
graded, positive correlation between serum 
cholesterol and cardiovascular mortality rate 
(Neaton and Wentworth, 1992). Elevations in 
lipoprotein (a) [Lp(a)] constitute a more 
recently recognized independent risk factor 
for cardiovascular disease. Elevations of LP(a) 
greater than 30 mg/dL increase the risk of CAD 
approximately twofold (Beckman et al., 2002). 

Diabetes 

Diabetes mellitus magnifies the risk of cardio- 
vascular morbidity and mortality. Diabetics 
have a two- to fourfold increase in the risk 
of CAD. Diabetics, particularly those with 
non-insulin-dependent diabetes mellitus 
(NIDDM) are at high risk of vascular disease 
because of high levels of triglycerides, LDL, and 
very low-density lipoprotein (VLDL) particles. 
Patients with NIDDM tend to produce small, 
dense LDL particles that are more vulnerable to 
oxidation. Other mechanisms for the adverse 
effects of diabetes that promote vascular disease 
include glycation of arterial wall proteins, 
enhancement of LDL oxidation, microvascular 
disease of the vasa vasorum, change in cellular 
function, promotion of thrombogenesis, and the 
development of renal disease and hypertension 
(Beckman et al, 2002). 



THE EPIDEMIOLOGY AND ETIOLOGY OF ATHEROSCLEROSIS 




Hypertension 

Hypertension is a well-recognized risk factor 
for atherosclerotic disease, particularly stroke 
and to a lesser extent ischemic heart disease and 
peripheral vascular disease. There are several 
possible mechanisms for the underlying poten- 
tiation of atherogenesis by hypertension, 
including direct mechanical disruptive effects, 
actions on vasoactive hormones, and changes in 
the response characteristics of the arterial wall. 
It is thought that, although hypertension may 
potentiate or enhance atherogenesis, hyperten- 
sion alone is probably not sufficient for athero- 
genesis (Valentine et al., 1996). 

Homocysteine 

Alterations in homocysteine metabolism are an 
independent risk factor for the development of 
vascular disease. Elevations of plasma homo- 
cysteine levels are associated with increased 
risks of all forms of atherosclerotic vascular 
disease. Homocysteine can react with LDL cho- 
lesterol to form oxidized LDL, which is found 
in early atherosclerotic lesions. Through this 
mechanism homocysteine can promote 
endothelial dysfunction, lipid peroxidation, and 
oxidation of LDL cholesterol. 

Atherogenesis and Lipid Metabolism 

Central to the discussion of atherogenesis is the 
metabolism of the peripheral blood lipopro- 
teins. These are a complex macromolecule of 
lipid and protein in which the nonpolar lipid 
core is surrounded by a polar monolayer of 
phospholipids and heads of free cholesterol 
and apolipoproteins. This structure allows for 
the transport of the relatively insoluble lipids 
through the liquid plasma. The lipoproteins 
differ in their proportions of lipid content and 
proteins found on their surface. Lipid disorders 
alter the composition and structure of the 
lipoprotein. For example, as mentioned earlier, 
patients with high triglycerides produce LDL 
with a higher protein-to-lipid ratio, yielding a 
small dense LDL. 

Cholesterol 

The body uses cholesterol for numerous func- 
tions including cell membrane biogenesis, 



steroid synthesis, and formation of bile acids. 
The human body can produce all the cholesterol 
it needs. The liver is the primary producer of 
endogenous cholesterol. Cholesterol is derived 
from the in vivo form acetate by a mechanism 
characterized by a rate-limiting step in which 3- 
hydroxy-3-methylglutaryl coenzyme A (HMG 
CoA) is converted into melavalonic acid by 
HMG CoA reductase. Statin-type drugs inhibits 
this step. This action decreases endogenous 
cholesterol production. 

Lipoproteins 

The major lipoproteins are chylomicrons, 
VLDL, intermediate-density lipoprotein (IDL), 
LDL, and HDL. 

Chylomicrons are larger triglyceride carrying 
particles formed after ingestion of a meal. They 
result from the processing of ingested fat by 
intestinal mucosal cells. They are transported to 
the thoracic duct via the intestinal lymphatics to 
eventually end up in the peripheral circulation. 
At peripheral sites the chylomicrons are acted 
upon by lipoprotein lipase bound to capillary 
endothelium. Chylomicrons have the lowest 
density of any of the lipoproteins. 

Very low-density lipoproteins are not as large 
as chylomicrons and are slightly more dense. 
They carry triglycerides and other fats synthe- 
sized in the liver. The IDLs are formed when 
some of the triglyceride is removed from VLDL. 
Under normal circumstances it is removed so 
rapidly from plasma that its concentration is 
quite low. 

Low-density lipoprotein is normally pro- 
duced from the catabolism of VLDL. It is the 
major carrier of cholesterol in the plasma. 
It is cleared by both receptor-mediated and 
non-receptor-mediated processes from the 
plasma. Low-density lipoprotein may be modi- 
fied through acetylation, oxidation, or both. 
These modified forms are particularly impor- 
tant in the development of atherogenesis. 
First, these molecules are cytotoxic and may 
damage the vascular endothelium, initiating 
atherosclerosis. They may also aggregate in the 
intima of the vessel wall and are chemotactic 
for inflammatory cells such as monocytes. 
These modified LDL particles have a decreased 
affinity for the normal LDL receptor and thus 
require clearance through other scavenger 
pathways. 




> 



VASCULAR SURGERY 



Lipoprotein (a) 

Lipoprotein (a) [Lp(a)] is a particle comparable 
in size to LDL. It is assembled from LDL and a 
large glycoprotein called apolipoprotein (a). It 
has a decreased affinity for the LDL receptor 
compared with LDL itself. Evidence links ele- 
vated levels of Lp(a) with increased risk of vas- 
cular disease. Lipoprotein (a) may also be taken 
up by scavenger pathways and thus accumulates 
in foam cells in the early atherosclerotic lesion. 

Small-Density LDL 

Alterations of LDL metabolism may create 
species of LDL particles with higher protein- 
lipid concentrations. Increased levels of these 
types of particles (termed small, dense LDL) are 
associated with increased risk of atherosclerotic 
disease and with elevated triglycerides (TGs) 
and low levels of HDL cholesterol. The mecha- 
nism linking these particles and hypertriglyc- 
eridemia relates to the production of 
chylomicrons and VLDL that are particularly 
rich in TGs. Catabolism of these TG-saturated 
VLDL particles produce LDL particles that have 
higher than normal TG content. These TG-rich 
LDLs are susceptible to further lipolysis by 
hepatic lipase, producing a decrease in size and 
increase in the density of the particle. Small, 
dense LDLs are believed to be more susceptible 
to oxidative modification and hence are thought 
to be highly atherogenic. Metabolic disorders 
such as diabetes and insulin resistance syn- 
drome often produce this lipid particle. 

High-Density Lipoprotein 

High-density lipoprotein is the lipoprotein 
responsible for the transport of cholesterol from 
cells to other lipoprotein or catabolic sites. 
High-density lipoprotein may be formed de 
novo in the liver and intestines, and intravascu- 
larly from the redundant surface material of 
chylomicrons and VLDL. Newly formed HDL 
consists of free cholesterol phospholipids and 
apoproteins. Free cholesterol from cells and 
perhaps from other lipoproteins reacts in the 
plasma with a complex containing the enzyme 
lecithin-cholesterol acetyltransferase (LCAT), 
apoprotein A-l, and an HDL-associated apopro- 
tein D. This complex attaches to, and the cho- 
lesterol is esterified by, LCAT. As nonpolar 



esterified cholesterol migrates to the interior of 
the particle, a sphere with an outer coating of 
free cholesterol and an inner core of esterified 
cholesterol and small amounts of triglyceride is 
formed. 

In epidemiological studies elevated HDL 
levels are associated with a reduced risk of ath- 
erosclerotic vascular disease. It is thought that 
HDL mediates this benefit through reverse cho- 
lesterol transport, which does not involve a 
direct route from peripheral tissues to the liver 
but rather depends on repeated transfer of cho- 
lesterol esters among lipoproteins before excre- 
tion through the liver. 

Lipid Metabolism 

Chylomicrons are formed in the intestine from 
ingested fat and taken by the intestinal lym- 
phatics to peripheral blood and then to adipose 
and other tissues. There, most of the triglyceride 
is acted upon by the enzyme lipoprotein lipase, 
transported across the cell membrane as fatty 
acid and monoglyceride, resynthesized into 
triglyceride, and stored. When necessary, intra- 
cellular triglyceride can undergo lipolysis. The 
released fatty acid is then transported out of the 
cell and bound to albumin to be transported in 
the plasma. After lipolysis a remnant of the chy- 
lomicron is transported to the liver and catabo- 
lized as a portion of the particle apolipoprotein 
A [Apo(A)] free cholesterol, and phospholipid is 
transferred to HDL formed in the liver; HDL 
may also pick up free cholesterol from cells. The 
cholesterol from other cells is esterified under 
the influence of LCAT. This ester is then avail- 
able for storage or transport. 

Very low-density lipoprotein is synthesized in 
the liver from fatty acids obtained from the pro- 
cessing of chylomicrons or from endogenously 
produced triglyceride. These particles are 
smaller and more dense than chylomicrons. The 
apolipoproteins associated with VLDL are Apos 
B-100, C-l, C-II, C-III, and E. 

Very low-density lipoprotein exchanges 
triglycerides for cholesterol esters from HDL. 
Like chylomicrons, lipoprotein lipase catalyzes 
the hydrolysis of triglyceride in VLDL to fatty 
acids that are used by muscle or stored as fat 
in adipose tissue. This hydrolysis step reduces 
VLDL to IDL. Intermediate-density lipoprotein 
can be taken up by the LDL receptor or be 
reduced to LDL by hepatic lipase. Intermediate- 



THE EPIDEMIOLOGY AND ETIOLOGY OF ATHEROSCLEROSIS 




density lipoprotein clearance is mediated by 
Apo E, which has a higher affinity for the LDL 
receptor than Apo BB. Low-density lipoprotein 
contains only the apolipoprotein B-100. Two 
thirds of the LDL is cleared through the LDL 
receptor, 60% to 70% of which is located in the 
liver. Peripheral cells can also take up LDL for 
membrane biogenesis and steroid synthesis. 

Low-density lipoprotein is removed from 
plasma by binding to these specific receptors 
located in many tissues, including the liver. After 
binding, LDL is internalized and metabolized to 
free cholesterol and other products. Cholesterol 
is stored in cells as the ester. Saturation of LDL 
receptors inhibits intracellular cholesterol syn- 
thesis by inhibiting HMG CoA reductase. This 
negative feedback system operates so that intra- 
cellular cholesterol synthesis varies inversely 
with the availability of intracellular LDL. 

Theories of Atherogenesis 

Numerous theories exist regarding the patho- 
genesis of atherosclerosis (Table 1.3). One uni- 
fying hypothesis linking the various theories is 
the "response to injury" model that in a broad 
context embraces many aspects of the theories 
(Ross, 1999). 

The Atheroma 

Whatever the initiating process, the first lesion 
of arteriosclerosis occurs with the entry of LDL 
through the intima and into the arterial wall. 
The lipids of human plasma are similar to what 
can be found in these early lesions. These early 
lesions, termed fatty streaks, are minimally 
raised yellow lesions found in the aorta of 
infants and children. The lipid deposits in these 
lesions are found within macrophages and 
smooth muscle cells. Foam cells, which are 
macrophages containing lipid particles, are 



Table 1.3. Pathogenesis of atherogenesis 

Response to injury 
Monoclonal hypothesis 
Lipid hypothesis 
Inflammatory 
Lesion regression 
Unstable plaque 




Figure 1.1. Atherosclerotic plaque. The appearance of 
complex atherosclerotic plaque removed during a carotid 
endarterectomy. 



characteristic of these earlylesions.lt is believed 
that these lesions represent the precursor to 
more advanced atherosclerotic lesions. As these 
lesions grow they then intrude into the arterial 
lumen. 

Fibrous plaques are composed of large 
numbers of smooth muscle cells and connective 
tissue forming a cap over an inner core con- 
taining mainly lipid cholesterol esters believed 
to be from disrupted foam cells. The fibrous cap 
may provide structural support or may function 
as a barrier to sequester thrombogenic debris in 
the underlying plaque from the arterial lumen. 
These plaques can show evidence of uneven and 
episodic growth. Intermittent ulceration and 
healing may occur, and there is evidence that 
thrombi formed on lesions are incorporated 
into them and resurfaced with a fibro cellular 
cap and an intact endothelial layer. Whether all 
fibrous plaques are characteristic of advanced 
atherosclerosis evolving from the fatty streak 
is uncertain. However, fibrous plaques often 
appear chronologically after fatty streaks in the 
same anatomical locations and characterize 
clinically apparent atherosclerosis. 

Complicated plaques comprise the end stage 
of atherosclerosis and cause clinical symptoms 
(Fig. 1.1). These are fibrous plaques that have 
become calcified, ulcerated, or necrotic. The 
consensus, at least for coronary ischemic events, 
is that they are thrombotic in origin, resulting 
from the rupture of the complicated atheroscle- 
rotic lesions. In most patients myocardial 
infarction occurs as a result of erosion or 




> 



VASCULAR SURGERY 



uneven thinning and rupture of the fibrous cap, 
often at the shoulders of the lesion where 
macrophages enter and accumulate. Degrada- 
tion of the fibrous cap may occur by release of 
metalloproteinases, collagenases, and elastases 
by these cells. 

Response to Injury 

The response-to-injury hypothesis initially 
proposed that endothelial denudation was the 
first step in atherosclerosis. More recent data 
suggest that endothelial dysfunction rather 
denudation is the primary problem. According 
to this model, atherogenesis is a response to 
injury of the vascular endothelium. At its 
mildest, atherosclerosis may represent a repara- 
tive process that leads to thrombus formation 
and smooth muscle cell proliferation at the site 
of endothelial injury. The production of the 
endothelial injury may be from hypertension, 
cytotoxic molecules, or blood flow changes. Ath- 
erosclerosis develops at sites exposed to unusual 
shear stress, such as in the abdominal aortic 
bifurcation. Hypercholesterolemia, hyperhomo- 
cystinemia, and smoking may all contribute to 
endothelial injury. 

Lipid Hypothesis 

Cholesterol accumulation in atherosclerotic 
lesions was initially considered an incidental 
accompaniment of the degenerative changes in 
the arterial wall. Forty years ago the normal 
range of blood cholesterol encompassed every- 
one within two standard deviations from the 
mean. Cardiologists and cardiovascular sur- 
geons concluded that serum cholesterol must be 
unimportant because most myocardial infarc- 
tions occurred in patients well below this arbi- 
trary normal level. The notion that the mean 
cholesterol level in the average American was 
high enough to cause serious clinical illness 
seemed improbable, and thus the lipid hypoth- 
esis had few advocates. 

However, the most important study to 
demonstrate that blood cholesterol is a risk 
factor for CAD is the Framingham study. The 
results of this study demonstrated the risk for 
developing clinically evident CAD was a contin- 
uous curvilinear function of blood cholesterol 
level. Larger trials would follow substantiating 
the link between cholesterol level and clinical 



risk, so that eventually experts agreed on the 
causal link between blood cholesterol levels and 
CAD risk. 

As mentioned previously, the endothelial 
injury hypothesis postulated the loss of 
endothelial cell integrity; however, in areas of 
atheroma often the endothelium remains intact. 
These results suggested that monocytes pene- 
trate the intact endothelium, settle in the intima, 
and then take up cholesterol particles to become 
foam cells. The lesion is initiated by elevated 
blood cholesterol characterized by lipid accu- 
mulation in foam cells. 

This process is accelerated in vivo under con- 
ditions in which the circulating LDL is modified 
to oxidized LDL. Many biological properties of 
oxidized LDL make it more atherogenic than 
native LDL including cytotoxicity. These facts 
are supported by data that demonstrate benefits 
of antioxidants in preventing oxidation of LDL 
on lesion progression. 

Inflammatory Theory 

This theory stems from the observations that 
atherosclerosis represents a different stage in 
chronic inflammatory process in the artery. 
Unchecked, this process may eventually result in 
the advanced complicated lesion. 

The different forms of injury increase the 
adhesiveness of the endothelium to leukocytes 
and platelets. It also induces the endothelium 
to have procoagulant activities and to form 
vasoactive cytokines and growth factors. The 
response then triggers the migration and prolif- 
eration of smooth muscle cells that form the 
fibrous lesion. Macrophages and T lymphocytes 
regulate the majority of the inflammatory com- 
ponent of this process. 

Macrophages have the ability to produce 
cytokines (such as tumor necrosis factor-oc, 
interleukin-1, and transforming growth factor- 
P), proteolytic enzymes, and growth factors 
such as platelet-derived growth factor and 
insulin-like growth factor. In addition, they 
express class II histocompatibility antigens 
that allow them to present antigens to T 
lymphocytes. 

Plaque Regression 

Plaque regression refers to a discernible 
decrease in intimal plaque. Apparent regression 



THE EPIDEMIOLOGY AND ETIOLOGY OF ATHEROSCLEROSIS 




of atherosclerosis has been documented by 
serial contrast arteriography in both coronary 
and peripheral vascular beds. Although plaque 
regression is usually thought of as a decrease in 
plaque bulk, it may proceed by other means. 
This lessening of luminal intrusion on sequen- 
tial angiography coincides experimentally with 
decreased plaque size and lipid content. 
However, as intimal plaques enlarge, a closely 
associated enlargement of the affected artery 
segment tends to limit the stenosing effect of the 
enlarging intimal plaque (Glagov et al., 1987). 
In the human left main coronary artery such 
enlargement keeps pace with increases in 
intimal plaque and is effective in preventing 
lumen stenosis until plaque area occupies on 
the average approximately 40% of the cross- 
sectional area. Continued plaque enlargement 
or complication apparently exceeds the ability 
of the artery to enlarge and stenosis may then 
develop. Thus the development of critical lumen 
stenosis, the maintenance of normal cross- 
sectional area, and the development of an 
increase in luminal diameter are dependent on 
the respective rates of plaque growth and arte- 
rial enlargement. 

Unstable Plaque 

Plaque rupture is the major cause of acute 
coronary syndromes (Table 1.4). Often, however, 
plaque rupture may be asymptomatic but con- 
tributes to the rapid growth of lesions as throm- 
bus fibroses. 

A number of characteristics distinguish 
stable plaque from the unstable plaque that 
might produce acute symptoms. The common 
underlying feature of the unstable plaque is 
thinning of the fibrous cap, which is composed 
mainly of vascular smooth muscle cells and 
matrix. In plaques that have ruptured, the 
fibrous cap at the shoulders of lesion where the 
cap meets the normal segment of the arterial 
wall is where this thinning occurs. Another 
typical feature of the unstable plaque is a large 



Table 1.4. Characteristics of the unstable plaque 

Thinning of fibrous cap 
Lipid core 

Intraplaque thrombosis 
Macrophage infiltration 



necrotic core filled with lipid and cellular debris 
with intraplaque and intraluminal thrombosis. 
The final feature is that of intense macrophage 
infiltration. Proteases and elastases released 
form inflammatory cells may contribute to the 
thinning of the fibrous cap seen in these lesions. 
Additionally, they contribute to the thrombotic 
nature of the unstable plaque through the elab- 
oration of tissue factor. 

Prevention 

Hypotheses of pathogenesis and etiology of 
atherosclerosis have been tested through the 
manipulation of risk factors associated with this 
disease process. Among the various strategies 
tried, only those strategies that promote a 
decrease in LDL or an increase in HDL have 
been associated with favorable changes in the 
plaque itself. 

Additionally, large epidemiological studies 
have demonstrated that lower cholesterol levels 
are associated with a lower overall risk of mor- 
bidity and mortality due to CAD (Martin et al., 
1986). Numerous clinical trials support these 
epidemiological data, and show that cholesterol 
lowering therapies lead to a significant reduc- 
tion in morbidity and mortality associated with 
CAD. Additionally, these benefits extend to a 
population presenting with peripheral arterial 
disease as well. The benefits of statin therapy to 
decrease risk is seen as early as the first year of 
treatment and extend not only to prevention 
of cardiovascular disease but also to the quality 
of life. In this era of evidence-based medicine, it 
would be difficult not to treat patients identified 
at risk with statin therapy based on these data. 
Recommended treatment guidelines are given 
in Table 1.5. 

However, one must understand that the treat- 
ment to prevent or stabilize atherosclerotic 
plaques extends not just to those patients with 
demonstrable severely stenotic lesions. In fact, 
it seems to be that most myocardial infarctions 
occur at sites that did not have prior angio- 
graphically recognized severe lesions. These 
facts are supported by the finding that thallium 
studies in stable CAD show that the site of 
stress-induced myocardial ischemia is fre- 
quently not the site of myocardial infarction. To 
extend this concept, it would seem reasonable to 
start statin therapy in patients at risk before the 




VASCULAR SURGERY 



Table 1 .5. Risk factors that modify low-density lipoprotein 

Cigarette smoking 

Hypertension (>140/90) or on antihypertensive 

I medication 
Low HDL cholesterol (<40mg/dL) 
Family history of premature coronary artery disease in 
male first-degree relative or female <65 years of 
age 
Age (men >45 or women >55 years of age) 

Risk categories that modify LDL cholesterol goals 
Risk category LDL goal 

Coronary artery disease <100mg/dL 

Multiple (2+) risk factors <130mg/dL 

0-1 risk factors <160mg/dL 



development of these unstable plaques or to 
stabilize the ones already present. 



Controversy 



The importance of treating patients to lower the 
cholesterol levels and to lessen the risk of devel- 
oping atherosclerosis is well accepted. However, 
the question remains whether there is a thresh- 
old below which cholesterol reduction may 
translate into clinical benefit. 

On average drug therapy with simvastatin 
loweres LDL cholesterol levels by 35% and 
reduces heart risk by 34% (Pedersen et al., 
1998). The goal of this study was to reduce total 
cholesterol below 200. However, many patients 
achieved reductions greater than this and were 
associated with continuing but progressively 
smaller reductions in heart attack risk. This 
subgroup analysis estimated a 1% reduction in 
LDL, reducing the risk of major coronary events 
by 1.7%. However, at what point this benefit can 



be extrapolated to remains to be determined. 
Another active debate is whether the treatment 
for acute myocardial infarction in high-risk 
patients should be lipid-lowering therapy rather 
than revascularization (Forrester and Shah, 
1997). 

Additional therapeutic approaches that are 
receiving attention include antioxidant treat- 
ment such as vitamin E. In a situation where 
oxidation of LDL is a major target in atheroge- 
nesis, antioxidant therapy obviously might play 
a role. To what extent it may be of benefit is still 
under investigation. 

Fundamental to the treatment of atheroscle- 
rosis is recognizing it as a systemic disease with 
the potential to affect a variety of end organs. 
Therefore, when patients are identified it 
appears advantageous to screen, counsel, and 
treat patients as soon as possible. 

References 



Beckman JA, Creager MA, Libby P. (2002) JAMA 287: 

2570-81. 
Forrester JS, Shah PK. (1997) Circulation 96:1360-2. 
Glagov S, Weisenberg E, Zarins CK, Stankunavicius R, 

Kolettis GJ. (1987) N Engl J Med 316:1371-5. 
Hiatt WR, Hoag S, Hamman RE (1995) Circulation 91: 

1472-9. 
Hirsch AT, Treat-Jacobson D, Lando HA, Hatsukami DK. 

(1997) Vase Med 2:243-51. 
Jonason T.Bergstrom R. (1987) Acta Med Scand 221:253-60. 
Martin MJ, Hulley SB, Browner WS, Kuller LH, Wentworth 

D. (1986) Lancet 2:933-6. 
Neaton JD, Wentworth D. (1992) Arch Intern Med 152:56-64. 
Pedersen TR, Olsson AG, Faergeman O, et al. (1998) Circu- 
lation 97:1453-60. 
Ross R. (1999) N Engl J Med 340:115-26. 
Smith I, Franks PJ, Greenhalgh RM, Poulter NR, Powell JT. 

(1996) Eur J Vase Endovasc Surg 11:402-8. 
Sueta CA, Chowdhury M, Boccuzzi SJ, et al. (1999) Am J 

Cardiol 83:1303-7. 
Valentine RJ, Kaplan HS, Green R, Jacobsen DW, Myers SI, 

Clagett GP. ( 1996) J Vase Surg 23:53-61, discussion 61-3. 



2 



Clinical Evaluation of Patients with 
Vascular Disease 

William G. Tennant 




The primary goal of the clinical evaluation of 
patients with vascular disease is to decide which 
tests will help the surgeon treat the patients' 
problem while at the same time minimizing 
patient discomfort. Investigation of patients 
with vascular disease differs from that of other 
surgical patients and depends mainly on the 
underlying disease process. For instance, 
patients with lower extremity occlusive vascular 
disease suffer not only from their index problem 
(claudication, ischemic rest pain, gangrenous 
ulcers) but also from some of the conditions that 
have predisposed them to vascular disease in the 
first place (diabetes, hypercholesterolemia, etc.). 
In addition, they are likely to require a number 
of medications for these predisposing condi- 
tions, some of which require consideration in 
diagnosing and treating vascular disease. It is 
also important to keep in mind that the presence 
of occlusive vascular disease in the lower limbs 
indicates the likely involvement of other vessels 
(coronary, carotid, cerebral, renal, mesenteric, 
etc.). Because the underlying pathology is very 
different, the clinical evaluation of patients with 
aneurysmal disease is strikingly different. These 
patients are healthier overall and their clinical 
evaluation is less intense. 



Patients' Characteristics 

Vascular surgery patients suffer many of the 
fears and anxieties of other surgical patients. 
Added to this are fears of gangrene, amputa- 



tion, and aneurysm rupture. Many elderly 
patients suffer other severe illness or disability. 
They may, as a result, have limited aims and 
aspirations when seeking investigation and 
treatment. In contrast, younger patients' family 
life or career may be threatened by their disease, 
and very high expectations of investigation and 
treatment have to be realistically modified. It is 
these human characteristics that deserve our 
consideration when deciding on an investigative 
pathway. Although it is important to gain all the 
information required to execute an effective 
treatment plan, it is equally important to do this 
in as noninvasive and humane a way as possi- 
ble. Fortunately, the technology is on our side in 
this regard, and the days of highly invasive 
investigations are probably numbered. 



The History 



The value of a good clinical history is increas- 
ingly overlooked as techno-diagnosis advances. 
One should remember that the history is usually 
the first interaction that takes place between the 
doctor and patient. It is at this time that the 
therapeutic relationship is forged. With skill and 
practice it is possible to elicit not only symp- 
toms but also their significance to the patients, 
the patients' expectations and fears, and their 
attitudes toward treatment. It is possible to 
avoid unnecessary diagnostic tests and limit 
the investigative mill that the patient is put 
through. 




> 



10 



VASCULAR SURGERY 



There are some general points in the clinical 
history that warrant mention: 

1. Lifestyle. Risk factors that can lead to 
the progress of vascular disease such as 
smoking, diabetes, hypertension, and 
hyperlipidemia are ascertained in the 
history. Additionally, an adequate exercise 
history should be elicited. One question 
that elucidates the rate-limiting organ 
system is how far patients can walk, and 
what stops them (leg pain, shortness of 
breath, chest pain, etc.). It is also important 
to know if the patient is taking hormonal 
medications such as oral contraceptives 
or hormone replacement therapy. These 
medications can predispose to venous 
and occasionally arterial thrombosis. It is 
during the history taking that a physician 
can begin to address many of these risk 
factors. By recruiting antismoking clinics 
or eliciting the help of diabetes and cardiac 
specialist physicians, a surgeon can 
improve a patient's overall health both pre- 
and postoperatively. 

2. Family history. It is especially important to 
question the patient about the prevalence 
of early cardiovascular disease or throm- 
bosis (i.e., stroke, occlusive limb disease, or 
cardiac disease) that manifests before age 
50. Aneurysm disease has a clear familial 
association, and an incidence approaching 
20% in first-degree relatives. 

3. Atherosclerosis. Atherosclerosis is a sys- 
temic disorder, so inclusion of a discussion 
of stroke/transient ischemic attacks and 
coronary artery disease/myocardial infarc- 
tion/angina is important. 

Although the points covered above may elicit 
factors predisposing the patient to vascular 
occlusive or aneurysm disease, they are non- 
specific and nondiagnostic. Because the symp- 
toms of occlusive vascular, aneurysmal, and 
venous disease differ, they will be dealt with sep- 
arately below. It should be remembered, 
however, that they may occur in combination. 

Chronic Limb Ischemia 

The principal symptom of chronic limb 
ischemia (CLI) is that of claudication (claudi- 
care, to limp). This is effort-related muscular 



pain relieved by rest. In the lower limb, patients 
in the initial stages of disease complain of calf, 
thigh, or buttock pain brought on by walking, 
which is relieved after a few minutes of rest. 
This is commonly a condition that follows a 
variable course with periods of remission and 
relapse, often according to changes in lifestyle, 
medications, or the progress of a comorbid con- 
dition such as diabetes mellitus. With worsen- 
ing ischemia, the patient begins to feel pain 
at night usually in the distal forefoot, toes, 
and instep (rest pain). As the patient becomes 
horizontal in bed (removing the effect of gravity 
on blood flow), and the blood pressure drops 
with the onset of sleep, perfusion of the lower 
limbs worsens. Patients often wake up in the 
middle of the night with pain that they can 
relieve only by getting out of bed and, paradox- 
ically, walking around the bedroom. Some 
patients with rest pain learn to sleep with the 
affected leg hanging over the side of the bed to 
regain the assistance of gravity (Fig. 2.1). When 
patients sleep with ischemic limbs dependent, 
there is a gradual onset of edema and worsen- 
ing tissue perfusion, which create a vicious 
circle of pathologies. 



Acute Critical Limb Ischemia 

Acute critical limb ischemia (ACLI) can be 
defined as sudden onset of severe limb 
ischaemia of less than 24 hours' duration. The 
principal causes are arterial embolism and 
thrombosis. A history should be taken to 
include the common sources of emboli (Table 
2.1). A history suggestive of claudication in 
the affected limb makes thrombosis in situ of 
a chronic arterial stenosis more likely than 
embolus. 

The symptoms of ACLI include paresthesia, 
pain in the limb at rest, numbness, coldness, and 
paralysis. Symptoms are likely to be more severe 
in cases of embolus than in cases of thrombosis 
because thrombosis usually occurs at the site of 
a chronic stenosis, completely occluding the 
vessel. Where a stenosis has existed, it is likely 
that a collateral circulation has developed that 
will continue to function even when the main 
vessel is occluded. In cases where an embolus 
has suddenly occluded a previously normal 
limb artery, there are no collaterals to support 
adequate perfusion. 



11 



CLINICAL EVALUATION OF PATIENTS WITH VASCULAR DISEASE 





Figure 2.1. Rest pain. This elderly woman is adopting a classic 
posture, which gives gravity assistance to blood flow while she 
is recumbent. 



Upper Limb Vascular 
Occlusive Disease 

Chronic occlusive vascular disease in the arm is 
considerably less common than that in the leg. 

Table 2.1. Common sources of emboli 

Cardiac arrhythmias (commonly atrial fibrillation) 
Cardiac mural thrombus from recent myocardial 

infarction 
Diseased heart valves 

Atheroma of aortic arch or more distal aorta 
Areas of chronic arterial damage (cervical rib, thoracic 

outlet syndrome) 
Aortic aneurysm (rare) 
Broken catheter tips 

Bullets and other materials introduced violently 
Air 

Amniotic fluid 
Fat (long bone fractures) 



Perhaps because of its rarity, the diagnosis is 
often made late and by exclusion. Arm claudi- 
cation presents with effort-induced heaviness 
or tiredness that is relieved by rest. The patient 
may also complain of relative pallor and an 
impression of coldness of the affected limb, 
exacerbated by cold exposure. 

Subclavian occlusive disease may also cause 
cerebrovascular symptoms because of the 
anatomical relationship between the vertebral 
arteries that arise off of the subclavian arteries. 
This is best exemplified by the subclavian steal 
syndrome. Tight stenosis or occlusion of the 
subclavian artery proximal to the origin of the 
vertebral artery leads to effort-induced reversal 
of flow in the vertebral artery that contributes 
to the arterial supply of the arm (Fig. 2.2). When 
the arm is exercised, increased (reversed) flow 
from the vertebral artery to the subclavian can 
lead to marked but transient symptoms of 
brainstem ischemia in addition to arm claudi- 
cation. Although most patients with this condi- 
tion have no symptoms of cerebrovascular steal 
at rest, symptoms can occur during exercise, 
including dizziness, ataxia, diplopia, and bilat- 
eral blurred vision. 

Acute upper limb vascular disease is usually 
due to embolism. Trauma is a less common 




Severe stenosis 
or occlusion 
of proximal 
subclavian artery 



Figure 2.2. Subclavian stenosis can lead to reversal of flow in 
the vertebral artery and vertebrobasilar symptoms. 




> 



12 



VASCULAR SURGERY 



cause. The symptoms are the same as for acute 
lower limb disease: pain or numbness, paralysis, 
and pallor. A history of recent myocardial 
infarction or of known atrial fibrillation should 
be taken. Traumatic causes can range from a 
"simple" supracondylar humeral fracture to the 
massive bone and soft tissue disruption caused 
by motorcycle accidents. It is also important to 
find out how the symptoms have developed over 
time. For instance, many elderly patients with 
brachial emboli give a history of severe pain ini- 
tially followed by gradual resolution over the 
succeeding hours. These patients then present 
to the vascular surgeon with viable limbs and 
minor symptoms. Other patients may complain 
of worsening symptoms, and the need for 
urgent intervention becomes obvious. Where an 
artery has been damaged by trauma, there may 
be a clear history of resolution of symptoms 
and restoration of pulses after, for instance, the 
reduction of a fracture. 

Aortic Aneurysm 

Whether the aneurysm affects the thoracic or 
abdominal aorta, there are usually very few 
symptoms. Chronic symptoms that do occur are 
usually due to pressure effects on the surround- 
ing structures. Even in quite small abdominal 
aneurysms, erosion of adjacent vertebral bodies 
can occur, leading to back pain. One of the com- 
monest symptoms of large thoracic aneurysms 
is dysphagia from direct pressure on the esoph- 
agus. Patients who notice abnormal abdominal 
pulsation (frequently while bathing, or in bed) 
often present with amusing self-diagnoses that 
belie the serious nature of the condition. This 
has been called "slipped-heart syndrome." 

In the special case of inflammatory abdomi- 
nal aortic aneurysm (vide infra), fibrosis can 
extend laterally in the retroperitoneum to 
include the ureters, which can result in ureteral 
stenosis. The presentation of such aneurysms 
is often via the urologist, the patient having 
presented with symptoms due to obstructive 
uropathy and hydronephrosis or even renal 
failure. 

The acute presentation of abdominal aortic 
aneurysm is usually as a differential diagnosis 
of acute abdominal pain. Symptoms are not 
always due to rupture, and the aneurysm may 
be intact but acutely symptomatic. Symptoms in 
an intact aneurysm, though the etiology is 



unknown, are principally severe abdominal and 
back pain of sudden onset. The pain may radiate 
into the groin, flanks, or genitalia and can 
closely mimic renal colic. When the aneurysm is 
ruptured, there is also collapse and hypov- 
olemia. The distinction between acutely symp- 
tomatic intact aneurysms and ruptured 
aneurysms is impossible to make on history 
alone. 



Superficial Venous Disease 

Patients frequently complain about the 
unsightly nature of varicose veins, and imbue 
them with many symptoms. These include 
aching, itching, and swelling. Symptoms, 
however, correlate poorly with the apparent 
severity of the disease. When superficial venous 
disease is extensive and severe, symptoms are 
common and include those above with the addi- 
tion of ulceration. 



Deep Venous Disease 

Symptoms are usually of swelling, heaviness, 
and occasionally severe discomfort. The symp- 
toms are usually worse after prolonged stand- 
ing. There may be a history of deep venous 
thrombosis or previous abdominal or pelvic 
surgery with venous damage. Symptoms result 
from venous hypertension in the limb affected, 
and this is the final common pathway of both 
occlusion and incompetence of the deep veins. 
There may be a history of ulceration even if 
none is present at the time of examination. 



Clinical Examination 

Inspection 

The general signs of a predisposition to occlu- 
sive vascular disease include deposits of fat in 
the thin skin around the eyes (xanthelasma), 
and in the corneas themselves (arcus senilis). 
Patients may have white hair; the fingertips may 
be tinted yellow with tar from cigarettes, and 
patients may smell strongly of cigarette smoke. 
Some of these patients assert that they have 
stopped smoking. Patients may be short of 
breath at rest or on minimal exertion because of 
coexistent cardiac or respiratory disease. 



13 



CLINICAL EVALUATION OF PATIENTS WITH VASCULAR DISEASE 




The specific effects of occlusive vascular 
disease may produce clinical signs apparent 
on general examination, such as limb swelling 
ulceration or gangrene. There may be signs of a 
previous stroke or of severe loss of weight. 

Aneurysms may appear as a localized 
swelling if present in the periphery, for instance, 
traumatic aneurysms of the femoral, popliteal, 
or radial artery (Fig. 2.3). There are often very 
few signs of abdominal aortic aneurysms on 
general examination, unless the patient is very 
slim and the abdominal wall may be "draped" 
across the aneurysm with the patient supine and 
relaxed. The pressure caused by an aneurysm on 
adjacent structures may rarely cause related 
clinical signs (Fig. 2.4). 

Palpation: Examination 
of the Pulses 

Lower Limbs 

Pulses are normally palpable in the femoral tri- 
angle at the midinguinal point, in the popliteal 
fossa, posterior to the medial malleolus, and on 
the dorsum of the foot between the first and 
second metatarsals. In the normal subject, the 
popliteal pulse is felt by compressing the artery 
against the tibial plateau anteriorly. This is best 
done with the patient's leg flexed at the knee. 
It is particularly important to distinguish aor- 
toiliac disease from infrainguinal disease. In 
aortoiliac disease the femoral pulses are dimin- 
ished, whereas in infrainguinal disease the 
femoral pulses are normal. 





Figure 2.3. This aneurysm of the superficial femoral artery was 
caused by previous trauma. It presented as a pulsatile swelling 
of the mid thigh. 



Figure 2.4. Swelling due to venous congestion from a popliteal 
aneurysm. A popliteal aneurysm caused the swelling of this 
patient's left leg by pressure on the adjacent popliteal vein. 
There are also multiple small skin infarcts caused by emboliza- 
tion from the aneurysm. 



Upper Limbs 

The subclavian pulse is present in the supra- 
clavicular fossa and the axillary artery in the 
infraclavicular fossa. Thereafter, the brachial 
artery is usually palpable in the cubital fossa 
deep to the bicipital aponeurosis. The ulnar 
pulse is palpable just medial to the tendon of the 
flexor carpi ulnaris and the radial lateral to the 
tendon of the flexor carpi radialis on the radial 
styloid process. A pulse is usually also palpable 
in the "anatomical snuff box" between the 
tendons of the extensor pollicis longus and 
brevis, where it overlies the scaphoid bone. 

Neck 

The carotid pulse can be felt medial to the 
muscle belly of sternomastoid. Occasionally 




> 



14 



VASCULAR SURGERY 



there may be marked tortuosity of the carotid 
artery in the neck, giving the impression of an 
aneurysm. 

Abdomen 

It is usually difficult to feel the normal abdom- 
inal aorta without causing the patient discom- 
fort. The aorta is best palpated between the 
xiphoid and umbilicus. Below the umbilicus, 
the aorta bifurcates. When aneurysmally dilated 
the pulse is easier to feel, and may in fact be a 
presenting symptom. 

Auscultation 

In each case, palpation of the pulses should 
be followed by auscultation in the same sites. 
Where there is a stenosis either at or immedi- 
ately proximal to the point of examination, a 
bruit will be heard in time with the cardiac 
systole. This is particularly relevant in carotid 
stenosis. Carotid bruits are not very sensitive 
or specific for carotid stenosis and require 
confirmation by carotid ultrasound (Magyar 
et al., 2002). However, a carotid bruit is often 
indicative of systemic atherosclerosis. Rarely, 
the bruit of a stenosed renal artery can be heard 
during auscultation of the abdomen. 

Differential Diagnosis of Leg Ulcers 

Many vascular patients present with ulcera- 
tions. There are three major types of leg ulcers: 
venous, ischemic, and neuropathic (Table 2.2). 
Ischemic ulcers tend to be very distal in the vas- 
cular tree and painful. Venous stasis ulcers tend 
to occur in the region of the medial malleolus 
and have associated brownish discoloration of 
the skin (lipatodermatosclerosis) and edema. 
Neuropathic ulcers tend to occur in diabetic 
patients under pressure points. Patients with 
diabetes present particular challenges in terms 



Table 2.2. Differential diagnosis ol 


leg ulcers 




Type of 


Location 


Pain 


Associated 


ulcer 






findings 


Ischemic 


Distal foot 


Yes 


No pulses 


Venous stasis 


Medial 


Maybe 


Stasis 




malleolus 




dermatitis 


Neurotropic 


Pressure 


No 


Diabetes 




points 








Figure 2.5. Pallor on elevation. The patient's leg is elevated,and 
the foot displays profound pallor. 



of diagnosis and management (Sumpio et al., 
2003). 

Other Clinical Tests 

Capillary Refill 

With the patient supine and the great toes 
together, both toes are gripped by the examiner 
using one hand and compressed. On release, the 
toes should change symmetrically from white to 
pink in less than 5 seconds. Asymmetry suggests 
arterial disease on the slowest side. 

Buerger's Test 

This is a test for severe chronic arterial occlu- 
sive disease. With the patient supine the straight 
legs are raised as far as possible. In arterial 
disease, there is extreme pallor of the feet in this 
position (Fig. 2.5). The legs are then placed on 
the examination bench and the patient is told to 
sit with the legs dependent over the side of the 
bench. Where there is severe chronic arterial 
disease, the feet become suffused with a deep 
ruddy red color, commonly described as "sunset 
foot." This is caused by ischemic maximal dila- 
tion of the arteriolar bed of the skin, allowing 
the skin to fill with partially oxygenated blood 
(Fig. 2.6). 



Trendelenburg Test 

In cases where incompetence of the saphe- 
nofemoral junction is suspected as a major 



15 



CLINICAL EVALUATION OF PATIENTS WITH VASCULAR DISEASE 





Figure 2.6. Dependent rubor. The leg has been placed depend- 
ent over the side of the bed, and is extremely hyperemic. 



cause of superficial varicose veins, the patient is 
asked to lie supine and raise the affected limb to 
about 45 degrees. Venous blood is "milked" 
proximally by firm stroking of the leg to empty 
all of the superficial veins. A tourniquet is 
applied as proximally as possible to occlude the 
superficial venous system. The patient is then 
asked first to sit up and swing the legs over the 
side of the examination couch, and then to 
stand. Where saphenofemoral incompetence is 
the major cause of superficial varicosities, the 
varicosities will remain collapsed. It is usual for 
the superficial veins to fill slowly, but rapid 
filling of the varicosities with the tourniquet in 
place indicates significant perforator disease 
distal to the tourniquet. It is possible to localize 
incompetent perforating veins by repeating the 
test with the tourniquet just above the knee. In 
this case calf varicosities will remain collapsed 
if the guilty perforating vein is between the 
saphenofemoral junction and the tourniquet. If 
the incompetent perforating vein is below the 
knee, the below knee varices will fill rapidly. 
Although the Trendelenburg method is some- 
what insensitive in localizing incompetent per- 
forating veins, it can provide useful clinical 
information. For more accurate localization of 
incompetent thigh perforators, and for all those 
in the calf, it is best to use duplex examination. 

Fixed Wave Doppler Examination 

A number of small and portable battery- 
operated machines are available, operating at 



frequencies between 5 and 10 MHz depending 
on the depth of penetration required (Fig. 2.7). 
In each case the signal from the insonation of 
the examined artery is converted into an audible 
sound from a built-in speaker. Normally the 
signal has a "triphasic" sound. Although it is 
possible to use the Doppler simply to locate an 
artery, the most common use is to measure the 
blood pressure at the periphery of a limb. For 
this, the Doppler machine is used in the same 
way as a stethoscope when measuring the blood 
pressure using Korotkoff sounds. A blood pres- 
sure cuff is placed around the limb proximal to 
the artery to be examined. The artery is then 
insonated and the cuff inflated above the sys- 
tolic pressure. As the cuff is deflated, the signal 
returns, and the pressure at which this happens 
is noted. When the pressure in all the required 
arteries has been measured, the pressure in the 
brachial artery is measured using the same tech- 
nique. The ratio between the ankle pressure 
and the brachial pressure is known as the 
ankle-brachial index (ABI). The ABI in normal 
patients without arterial occlusive disease is 
greater than 1. 

Handheld Doppler examination is also useful 
in the diagnosis of superficial venous disease 




Figure 2.7. An example of the type of handheld Doppler device 
suitable for use in the clinic. 




> 



16 



VASCULAR SURGERY 



to confirm the incompetence of the saphe- 
nofemoral or saphenopopliteal junctions, and to 
localize incompetent perforating veins. At each 
of the saphenous junctions, there is physiologi- 
cal retrograde flow into the superficial system of 
under 1 second' duration, which is audible using 
a handheld Doppler machine. If the reflux is of 
longer duration, it is indicative of pathological 
incompetence of the junction. With an experi- 
enced operator, it is possible to localize incom- 
petent perforating veins. 



Clinical Examination 
of Specific Conditions 

Chronic Lower Limb Ischemia 

In mild disease the legs may appear normal to 
inspection, but capillary return is delayed and 
the feet may be cool to touch. Distal pulses are 
be either weak and difficult to feel or absent. 
With increasing severity, the legs may be hair- 
less below the knee, and the toes cyanotic. As 
ischemia progresses, more proximal pulses may 
disappear and ulcers may appear on or between 
the toes. Buerger's test becomes positive. Even- 
tually more proximal painful ulcers over the 
lower leg, and digital gangrene signals very 
severe occlusive disease. 

Acute Lower Limb Ischemia 

Where there is thrombosis of a collateralized 
chronic stenosis, the signs of acute ischemia 
may be less severe than in cases of embolus 
(vide supra). In these less severe cases, the acute 
onset of the symptoms maybe the most obvious 
clue. The limb may appear normal to inspection 
but have reduced capillary return, pale rapidly 
on elevation, and have slightly altered sensation 
on formal testing. There may be some weakness, 
principally of the anterior muscle groups of the 
lower leg. With increasing ischemia, the signs 
of pallor and weakness increase, and there may 
be complete paralysis of the foot and toe 
dorsiflexor muscle groups. In these cases, the 
foot usually lies at rest in equinovarus due to 
paralysis of the peroneal muscles. In severe 
acute ischemia, such as that caused by 
embolism, the skin becomes mottled with blue 
blotches on a background of sallow white. If the 



blotches blanche on finger pressure (unfixed 
mottling), it may still be possible to save the leg 
if immediate action is taken to revascularize it. 
Where the mottling is fixed, that is, it fails to 
blanche on pressure, it is too late to save the 
limb. Muscle ischemia and impending necrosis 
in these severe cases cause the muscles to swell 
and become tender. This is often best seen in the 
anterior muscle compartment where the ten- 
derness is often exquisite and the compartment 
is almost stone hard to palpation. Examination 
of the pulses allows approximate localization of 
the level of disease. The presence of normal 
pulses on the opposite extremity supports the 
diagnosis of acute embolic disease. 

Mesenteric Ischemia 

In chronic cases there are often few signs on 
examination of the abdomen, but signs of 
weight loss and systemic vascular disease 
are present. Where the disease is acute, the 
abdomen can feel curiously doughy in the early 
stage and is diffusely tender. Bowel sounds may 
still be present. As time passes and transmural 
infarction supervenes, peritoneal signs develop. 

Upper Limb Ischemia 

Chronic arterial occlusive disease seldom affects 
the upper limb except as part of rare conditions 
affecting the aortic arch and subclavian ar- 
teries. Upper limb ischemia is usually due to 
embolism. The commonest embolic source in 
these cases is the myocardium in atrial fibrilla- 
tion or following myocardial infarction. Other 
sources include proximal stenoses in the aortic 
arch and subclavian arteries (Fig. 2.8). The clin- 
ical signs of acute upper limb ischemia are the 
same as in the lower limb: sensory alteration, 
paresthesia, weakness, and muscle tenderness. It 
is uncommon for the ischemia to be so severe as 
to lead to irreversible change because of the rich 
collateral supply in the arm. In intravenous drug 
abusers, intraarterial injection of illicit medica- 
tions "cut" with insoluble excipients may lead to 
extensive acute occlusion of small distal vessels 
(Fig. 2.9). Patients often present following delib- 
erate or accidental intraarterial injection, with 
a short history of almost overwhelming pain 
together with exquisite muscle tenderness and 
forearm muscles stone hard to touch. The skin 



17 



CLINICAL EVALUATION OF PATIENTS WITH VASCULAR DISEASE 





Figure 2.8. Gangrene from an arterial embolus. The distal 
aspect of the digit is gangrenous secondary to an embolus 
arising from a subclavian stenosis. 



of the forearm and hand is fixed and mottled 
and the hand clawed. 

Chronic Venous Disease 

Inspection with the patient standing is one of 
the most important aspects of the examination 
of chronic venous disease. The dilated veins of 
superficial disease are frequently obvious. Other 
signs of importance include swelling, hemo- 
siderosis of the skin of the malleolar area, 
lipodermatosclerosis, atrophie blanche, and 
ulceration (Fig. 2.10). Deep venous disease may 
be less obvious and present simply with chronic 
swelling of the limb. In later stages, all of the 
above signs may be present. 




Figure 2.9. Gangrene from drug injection. Injection into 
the radial artery led to gangrene to the thumb and thenar 
eminence. 



Figure 2.10. Chronic venous insufficiency. The limbs demon- 
strate the brownish discoloration associated with lipatoder- 
matosclerosis. Varicosities are also present. 




> 



18 



VASCULAR SURGERY 



Conclusion 

Clinical evaluation of the patient with vascular 
disease is of the utmost importance. Many clues 
about a patient's temperament, the disease, and 
expectations of treatment can be obtained from 
a thorough interview. Assessing a patient's risk 
factors for vascular disease not only helps the 
physician better understand the patient's chief 
complaint but also directs the preoperative 
workup of the patient. Despite the recent 
advances in vascular radiology, nothing replaces 
an excellent physical examination, which can 



shed light on the clinical extent of a patient's 
disease process. Finally, when radiological 
studies may take valuable time, there are several 
bedside tests that can be performed rapidly, 
allowing a vascular surgeon to make immediate 
treatment decisions. 



References 



Magyar MT, Nam EM, Csiba L, Ritter MA, Ringelstein EB, 

Droste DW. (2002) Neurol Res 24:705-8. 
Sumpio BE, Lee T, Blume PA. (2003) Clin Podiatr Med Surg 

20:689-708. 



3 



Noninvasive Vascular Examination 

Colleen M. Brophy 




The noninvasive vascular laboratory assists 
with the diagnosis of peripheral arterial disease. 
In general, two basic approaches are used: (1) 
indirect measures that characterize the func- 
tional severity of the disease, such as segmental 
pressures, Doppler waveform analyses, plethys- 
mography, and skin perfusion pressures; and (2) 
direct measures that characterize the anatomy 
of the disease using color duplex imaging. These 
studies are used to accurately diagnose the loca- 
tion and extent of peripheral arterial disease, 
assist with the planning of therapeutic options 
for the disease, and follow the outcomes of 
peripheral vascular interventions. 

Indirect Evaluation of 
Arterial Disease 

Ankle-Brachial Index 

In patients without palpable pedal pulses, the 
next step in the clinical evaluation is to perform 
a Doppler analysis. This is usually performed in 
the clinic or at the bedside using a handheld 
Doppler device. The probe is placed at an angle 
(Fig. 3.1) over the dorsalis pedis and posterior 
tibial arteries to determine if a signal can be 
obtained. The complete absence of Doppler 
signals suggests significant peripheral arterial 
disease. Approximately 10% of the normal pop- 
ulation does not have a dorsalis pedis artery; 
hence, the absence of a dorsalis pedis signal 
alone is not a significant finding. It is important 



to place the probe directly over the artery but at 
a 45- to 60-degree angle to obtain the best 
signal. Doppler signals can be used to assess the 
severity of the disease. A blood pressure cuff 
is inflated just above the ankle and a Doppler 
signal is listened for while the cuff is deflated. 
The highest pressure in which a signal is heard 
(dorsalis pedis compared to posterior tibial) is 
the ankle index. A similar approach with a blood 
pressure cuff on the upper arm and a Doppler 
probe on the brachial artery is used to deter- 
mine the brachial index. The ratio of the 
ankle to the brachial index represents the 
ankle-brachial index (ABI). The ABI is usually 
1 or greater. An ABI of 0.5 to 0.8 is consistent 
with claudication, and less than 0.4 is consistent 
with critical limb ischemia. Diabetic patients 
often develop medial calcinosis. Simply stated, 
the medial wall of the vessels become calcified 
("bone-like") and cannot be compressed by a 
blood pressure cuff. Thus, the ABI in a diabetic 
patient may be falsely elevated, and other 
studies are needed to accurately assess the 
peripheral vascular status. In general, however, 
the ABI is a useful screening test for peripheral 
vascular disease. 

Segmental Limb Pressures 

Segmental limb pressures assist with determin- 
ing the location of disease by measuring the 
pressure in the upper thigh, lower thigh, below 
the knee, just above the ankle, and at the trans- 
metatarsal level (Figs. 3.2 and 3.3). Again using 



19 




» 



20 



VASCULAR SURGERY 




Figure 3.1. Doppler signals are best obtained by holding the 
Doppler probe at an angle over the artery that is being 
insonated. 



a Doppler, an arterial signal is found at the dor- 
salis pedis or posterior tibial artery, the cuff 
is inflated until no signal is heard, and then 
deflated until the signal resumes. The systolic 
pressure is recorded for each cuff at each loca- 
tion. A gradient (decrease) in pressure greater 
than 20 mm Hg between adjacent levels suggests 
arterial occlusive disease in the vessel between 
the two cuffs (Fig. 3.3). An arterial pressure in 
the thigh that is less than that in the arm sug- 
gests aortoiliac occlusive disease. A drop in 
pressure between the upper thigh and lower 
thigh cuffs suggests superficial femoral artery 



disease. It is important to make a distinction 
between aortoiliac and infrainguinal disease 
because the overall approach is somewhat 
different. 

An additional useful measurement, particu- 
larly in diabetic patients, is to measure the 
digital pressures. The digital arteries are less 
likely to be affected by medial calcification and 
hence provide useful information when the ABI 
is falsely elevated. Digital pressures are meas- 
ured by placing a pneumatic cuff around the 
digit and measuring a plethysmographic arte- 
rial waveform using a photoelectrode on the 
end of the digit. In general, a normal toe pres- 
sure is 80% to 90% of the brachial pressure 
(normal toe-brachial index is 0.8 to 0.9). A toe 
pressure less than 38 mm Hg has been correlated 
with impaired forefoot wound healing (Vitti 
et al, 1994). 

In patients with symptoms consistent with 
claudication but relatively normal indices, exer- 
cise testing should be performed. A standard 
exercise test involves measuring the ABI at rest 
followed by walking on a treadmill at 2 miles/ 
hour at a 10% to 15% incline for 5 minutes. 
The ABI is recorded 1 minute after exercise. 
Normally, after exercise, there is a slight in- 
crease or no difference in the ABI. If the ABI 
decreases after exercise, this is consistent with 
claudication. 

Pulse Volume Recording 

Pulse volume recordings (PVRs) are a plethys- 
mographic measure of limb perfusion. 




Figure 3.2. Segmental limb pressures are 
measured by placing the cuffs along the 
extremity. 



21 



NONINVASIVE VASCULAR EXAMINATION 




SEGMENTAL PRESSURE 
AND PVR STUDY 

Brachial 
RIGHT LEFT 

134 | 



1.25 y i7i 



1.23 
0.81 

RIGHT 




0.74 


101 
110 


[dp 

PT 


65 


0.80 


86 








11 





ABI: 0.80 



ABI 



31 

0.63G, 






Plethysmography is essentially a measurement 
of volume. In general, the same cuffs are used to 
obtain segmental limb pressures and PVRs. The 
cuff is inflated and the volume shift that occurs 
with the cardiac cycle in that limb segment is 
recorded as a waveform. The PVRs do not 
provide quantitative data but can be analyzed in 
a qualitative manner. In normal patients there is 
a brisk upstroke, rapid decline, and dicrotic 
notch in the waveform. In the presence of 
peripheral arterial disease, the waveform 
becomes broader with a decreased amplitude. A 
PVR tracing that is flat at the forefoot (trans- 
metatarsal) level is consistent with significant 
peripheral vascular diseases and suggests that 
revascularization will be required for any fore- 
foot lesion to heal. 

Doppler Waveform Analysis 

Similar to PVRs, the Doppler waveform can be 
analyzed to determine if there is disease. Instead 
of an audio signal, a digital signal is converted 
to a tracing. Normally the waveform is triphasic 
(Fig. 3.4). With moderate disease the reverse 
flow component is lost and the waveform is 
monophasic. With severe disease, the waveform 
is blunted and/or absent. 

Skin Perfusion Pressure 

The skin perfusion pressure (SPP) is an addi- 
tional noninvasive vascular evaluation that is 
useful to determine which foot ulcers will heal 
with local wound care or minor amputation and 
which will require revascularization or major 
amputation. An SPP of less than 30mmHg can 
predict failure of forefoot wound healing with 



Figure 3.3. Segmental limb pressures show a low high-thigh 
pressure, indicating iliac disease, and a drop-off between the 
high-thigh and low-thigh pressures, indicating superficial 
femoral artery disease on the left side. On the right side there is 
a drop-off between the pressures in the low thigh and popliteal, 
indicating distal superficial femoral artery/popliteal artery 
disease. ABI, ankle-brachial index; Dorsaus Pedis (DP), Posterior 
Tibial (PT), prothrombin time; PVR, pulse volume recording. 






Figure 3.4. Normal triphasic Doppler waveform. 




22 



VASCULAR SURGERY 



Table 3.1 . Noninvasive measurements consistent with 



im 


Daired wound healing 






Ankle-brachial index (ABI) 


<0.3-0.4 




Toe pressure 


<35-40mmHg 


Skin perfusion pressure 


<30mmHg 


Pulse volume recording (PVR) 


Flat forefoot tracing 



an accuracy of 80% (Castronuovo et al., 1997). 
Skin perfusion pressure has largely replaced 
transcutaneous oximetry. 

What Wounds Will Heal? 

The aggregate measurements that are consistent 
with impaired wound healing are listed in 
Table 3.1. However, it is ultimately the clinical 
response to wound care that is the most impor- 
tant factor. If debridement and wound care leads 
to consistently unhealthy appearing wounds, 
angiography and revascularization should be 
considered. If good wound care leads to healthy 
granulation tissue, further evaluation is likely 
unnecessary. 

Direct Evaluation of 
Arterial Disease 

Color duplex imaging or "duplex" imaging 
incorporates real-time B-mode imaging and 
pulsed Doppler spectral analysis (Fig. 3.5). B- 
mode imaging directly views the blood vessel 
and provides anatomical detail of the vessel. 
This modality can analyze plaque characteris- 
tics, identify thrombus and the intimal flaps, 
and measure vessel diameter (to detect 
aneurysms and pseudoaneurysms). 

B-mode imaging is also used to localize areas 
of stenosis so that Doppler velocities can be 
determined. Velocity analyses are an indirect 
measure of the degree of stenosis. For carotid 
artery analyses, the degree of stenosis is esti- 
mated based on the peak systolic velocity (PSV), 
end-diastolic velocity (EDV), and the ratio of 
the PSV at the stenosis to the velocity in the 
common carotid artery (V r ). In general a V r 
greater than 4 is indicative of clinically 
significant stenosis (Table 3.2). 

Infrainguinal graft surveillance can be per- 
formed with duplex imaging. Because most 




Figure 3.5. Color duplex image of a high-grade carotid 
stenosis with a peak systolic velocity of 319 and end-diastolic 
velocity of 123. 



abnormalities occur in the first 2 years after 
implantation, graft surveillance is most impor- 
tant in this time frame. A PSV greater than 150 
to 200 cm/second is considered abnormal. In 
addition, the ratio of the velocity at the stenosis 
to the normal proximal velocity is used to 
determine the degree of stenosis (V r ). In addi- 
tion, high EDVs (>100 cm/second) are also sug- 
gestive of high-grade stenoses. Although the 
high-velocity criteria (PSV, EDV, V r ) are the 
most accurate to assess the risk of graft throm- 
bosis, low-velocity criteria are also helpful. The 
graft flow velocity (GFV) should normally 
exceed 45 cm/second. The velocities are used to 
stratify the risk of thrombosis (Table 3.3) (Mills, 
2001). Prophylactic graft revision should 
be considered in grafts with a high risk of 
thrombosis. 

Duplex examination is also useful for the 
diagnosis of popliteal artery aneurysm. This 
condition should be suspected if a widened 
popliteal pulse is palpated. An arterial diameter 



Table 3.2. Doppler velocity criteria for carotid stenosis 



Stenosis (%) 


PSV 


EDV (cm/sec) 


v- 


Normal 


<123 


<140 


<4.0 


1-15 


<123 


<140 


<4.0 


16-49 


<123 


<140 


<4.0 


50-79 


>123 


<140 


<4.0 


80-99 


>123 


>140 


>4.0 



EDV, end-diastolic velocity; PSV, peak systolic velocity; V r , velocity 
ratio. 



23 



NONINVASIVE VASCULAR EXAMINATION 




Table 3.3. Risk stratification for graft thrombosis 



Category 


PSV 


V, 


EDV 


GFV 




(cm/sec) 




(cm/sec) 


(cm/sec) 


Highest risk 


>300 


>3.5 


>100 


<45 


High risk 


>300 


>3.5 


<100 


>45 


Intermediate 


180-300 


>2.0 


<100 


>45 


risk 










Low risk 


<180 


<2.0 


<100 


>45 



EDV,end-diastolicvelocity;GFV,graft flow velocity; PSV, peak systolic 
velocity; V„ velocity ratio. 



Conclusion 

Noninvasive vascular laboratory tests comple- 
ment the clinical evaluation in determining 
the appropriate therapeutic approaches to 
patients with peripheral vascular disease. The 
specific laboratory tests should be tailored to 
the individual patient's diagnoses. The labora- 
tory provides noninvasive information on both 
functional and anatomical aspects of vascular 
disease. 



greater than 10 mm is considered abnormal. 
Bilateral aneurysms are seen in about 50% of 
cases, and proximal aneurysms occur in approx- 
imately 30% to 50% of patients. Consequently, it 
is imperative that aneurysms at other locations 
be excluded, with abdominal aortic aneurysms 
and femoral aneurysms the most common. 



References 



Castronuovo JJ Jr, Adera HM, Smiell JM, Price RM. (1997) 

J Vase Surg 26:629-37. 
Mills JL Sr. (2001) Semin Vase Surg 14:169-76. 
Vitti MJ, Robinson DV, Hauer-Jensen M, et al. (1994) Ann 

Vase Surg 8:99-106. 



4 

Radiological Investigations 

Steven M. Thomas, Kong T. Tan, and Mark F. Fillinger 




This chapter offers an overview of the available 
imaging techniques used in vascular radiological 
practice for the investigation of vascular diseases 
and how they can be used for a range of common 
vascular conditions. This discussion emphasizes 
the move away from invasive techniques and 
toward noninvasive techniques for the diagnosis 
of vascular diseases. We then describe the range 
of endovascular techniques currently required 
for the investigation and treatment of vascular 
disease. The most important contemporary 
approaches andtherapies are described,showing 
the important role these techniques now play in 
the management of a range of vascular disease 
processes. This discussion demonstrates how the 
area of endovascular intervention remains at the 
forefront of developments in minimally invasive 
techniques to treat vascular disease. Approaches 
for specific manifestations of peripheral vascular 
disease are not discussed, but the Trans Atlantic 
Inter-Society Consensus (TASC) document is 
recommended as an overview of endovascular 
intervention in the management of specific as- 
pects of peripheral arterial disease (Dormandy 
andRutherford,2000). 

Controversies 

The following controversies currently exist in 
the field of vascular radiology: 

• Conventional diagnostic catheter angiog- 
raphy is likely to be made obsolete by non- 
invasive vascular imaging. 



• When considering endovascular treatment 
of carotid artery stenosis, the current lim- 
itations of noninvasive testing mean that 
catheter arch angiography is required to 
assess the arch vessels and the whole of the 
carotid artery. 

• Computed tomography (CT) pulmonary 
angiography has resulted in little need 
for conventional pulmonary angiography 
in the diagnosis of pulmonary embolic 
disease. 

■ Arterial closure devices enhance patient 
throughput, and this may offset the 
increase in cost from their use. However, 
they can produce serious complications. 

• There is little evidence that primary stent- 
ing is superior to primary angioplasty 
alone in the treatment of arterial occlusive 
disease in the peripheral circulation. 

• Following endovascular aneurysm repair 
(EVAR) for abdominal aortic aneurysms 
(AAAs), long-term costs of stent-graft 
surveillance, and secondary treatment, may 
outweigh the short-term benefits of EVAR. 

Radiological Investigations 
and Endovascular Approaches 

The first vascular imaging technique was 
described at the end of the 19th century shortly 
after the discovery of the x-ray. This technique, 
described by Haschek and Lindenthal, involved 



25 




> 



26 



VASCULAR SURGERY 



the injection of a chalk-based contrast agent 
into the vein of an amputated hand. In vivo 
vascular imaging was first described in the 
1920s using agents such as lipiodol, strontium 
bromide, sodium iodide, and Selectan. The 
contrast agents were usually introduced by a 
catheter, following exposure of the vessel used 
for catheterization, or by percutaneous needle 
puncture to directly inject contrast into the 
vessel. Translumbar aortography used this type 
of technique as early as the 1930s. However, it 
was developments from the mid-20th century 
that brought angiography and subsequently 
vascular intervention to the forefront in vascu- 
lar diagnosis and treatment. The first of these 
was the introduction of the Seldinger technique 
in the 1950s that allowed safe percutaneous 
access to vascular structures. There then fol- 
lowed significant improvements in the range of 
available catheters to aid in gaining access to the 
different vascular beds, and rapid film changers 
allowed high-quality images to be obtained of 
all the major vascular beds. As a result, vascular 
imaging became a major branch of diagnostic 
medicine. However, arteriography remained 
an uncomfortable, if not painful, experience 
because of the use of conventional ionic contrast 
agents. The introduction of digital subtraction 
angiography (DSA) and nonionic contrast 
agents in the last two decades of the 20th 
century made arteriography much more accept- 
able to patients and their doctors. Digital sub- 
traction angiography allowed smaller amounts 
of contrast to be used, and the newer contrast 
agents rarely produced intense heat or pain. The 
combination produced fewer systemic side 
effects as well. 

At the same time, attention was directed 
toward less invasive therapeutic interventions, 
such as Dotter's technique of sequential dilation 
for atherosclerotic occlusive lesions. The devel- 
opment of the balloon angioplasty catheter, first 
described by Gruntzig and Hopff in 1974, her- 
alded an era in which a range of manifestations 
of arterial occlusive disease became amenable 
to treatment using percutaneous vascular inter- 
ventional techniques. There followed develop- 
ments of a range of techniques for treating 
arterial occlusive disease such as arterial stents, 
atherectomy catheters, lasers, and thrombolysis. 
From these, implantable devices such as inferior 
vena cava (IVC) filters and stent grafts for 
aneurysmal disease were developed. All these 



devices broadened the range of conditions 
amenable to treatment using interventional 
vascular radiological techniques. 

Improvements in the design of catheters and 
other devices have resulted in smaller caliber 
catheters and introduction sheaths. These along 
with the introduction of closure devices to seal 
the access site puncture allowed the develop- 
ment of day case ("same day") arterial proce- 
dures for both diagnosis and intervention. 
Despite these improvements, developments in 
other noninvasive imaging modalities meant 
that for arterial imaging and diagnosis, the use 
of what remains an invasive technique is 
required less frequently. Whereas arteriography 
was once a first-line investigation, noninvasive 
techniques such as ultrasound, radionuclide 
imaging, CT, and magnetic resonance imaging 
(MRI) now have a much wider role to play in 
both general diagnostic radiology and in the 
radiological investigation of vascular disease. 
Not only are these techniques safer, they also 
have the potential for a greater diagnostic yield 
because of the additional information that can 
often be obtained using these techniques. As a 
result of improvements in these noninvasive 
vascular imaging techniques, it is possible that 
the need for conventional diagnostic catheter 
angiography will become obsolete. However, at 
present, despite the reduced requirement for 
angiography for general diagnostic work, there 
remains a role for angiography in the investiga- 
tion of vascular disease, either as a first-line 
investigation or following other imaging inves- 
tigations. Also angiography remains paramount 
as part of all percutaneous endovascular inter- 
ventional techniques. 

Imaging Modalities 

The imaging techniques available for the inves- 
tigation of vascular diseases are best divided 
into invasive (i.e., catheter angiography) and 
noninvasive techniques. The following is a brief 
description of the commonly used modalities. 

Catheter Angiography 

Catheter angiography is an x-ray investigation 
in which a contrast agent is injected into the vas- 
cular system via a catheter, and sequential x-ray 
exposures are performed. The x-ray receiver can 



27 



RADIOLOGICAL INVESTIGATIONS 




be plain x-ray film (older machines), but are 
now almost universally digital systems, using an 
image intensifier or a flat panel detector. Older 
machines using cut plain x-ray films were cum- 
bersome and difficult to operate. In addition, 
the postprocessing facilities were very limited. 
In contrast, digital systems are easy to operate, 
require lower contrast dosage, produce better 
contrast resolution, have real-time display, 
and have facilities for image manipulations, 
such as pixel shifting and digital subtraction 
angiography. 

Contrast Agents 

The primary aim of the contrast agent is to allow 
imaging of vessel anatomy and morphology. 
During fluoroscopy, x-ray contrast agents act 
by changing the attenuation between tissues. 
Iodinated agents and gadolinium have a higher 
density than surrounding tissue, whereas 
carbon dioxide has a lower density. 

Iodinated Contrast Agents 

The most widely used contrast agents for 
catheter angiography are the water-soluble 
iodine-based agents. They can be divided into 
ionic and the newer nonionic agents. Nonionic 
agents have the advantage of considerably 
reducing the risk of adverse reactions to con- 
trast, but are more expensive. In the United 
Kingdom the low-osmolar nonionic agents are 
used almost exclusively for intravascular proce- 
dures. These agents are denser than blood, and 
the commonly used strength for diagnostic and 
interventional work is 300 mg of iodine per 
milliliter of contrast. For hand injections, this 
strength of contrast is usually diluted 50 : 50 
with saline. Patients with a previous history of 
adverse reactions to these contrast media, a 
strong history of allergic disease, or a hyper- 
sensitivity to iodine are at risk of developing a 
severe allergic reaction to these contrast agents. 
Other investigative modalities that do not 
require iodinated contrast media such as MRI 
or ultrasound should be considered, or alterna- 
tive non-iodine-based contrast such as carbon 
dioxide (C0 2 ) or gadolinium, as described 
below. The other major problem with iodinated 
contrast is nephrotoxicity. Overall, contrast- 
induced nephropathy occurs in approximately 
5% of all procedures requiring iodinated con- 



trast media. For the majority, the effect is 
temporary. However, in high-risk patients the 
incidence of contrast-induced nephropathy is 
around 20% to 30%, and in a significant number 
of cases the effect is permanent and nonre- 
versible. High-risk groups are those patients 
with preexisting renal impairment, diabetes 
mellitus, dehydration, on nephrotoxic drugs, 
and a history of multiple myeloma. These 
patients should be well hydrated prior to the 
procedure, if necessary by the administration of 
intravenous saline, and their renal function 
should be checked 48 hours postprocedure. 
Recently, some studies involving a small 
number of patients suggest that acetylcysteine 
and calcium channel blockers may reduce the 
incidence of contrast-induced nephropathy 
(Kay et al., 2003). There is also evidence that at- 
risk patients, particularly those with diabetes 
mellitus, have a lower incidence of contrast- 
induced nephropathy following the use of 
recently introduced iso-osmolar contrast agents 
(Aspelin et al., 2003). There is also a risk of 
potentially fatal lactic acidosis if a patient 
taking metformin develops acute renal impair- 
ment following the administration of iodinated 
contrast. Ideally metformin should be discon- 
tinued at the time of, or prior to, the procedure 
and withheld for 48 hours subsequent to the 
procedure and reinstituted only after renal 
function has been reevaluated and found to be 
normal. 

Other Agents 

Mainly because of the risk of nephrotoxicity 
with iodine-based agents, C0 2 has recently 
been advocated as avascular contrast media for 
patients with renal impairment. It can also be 
used when there are contraindications to iodi- 
nated contrast such as a history of iodine or 
contrast allergy. Minor alterations in techniques 
and software are required for C0 2 angiography. 
Although dedicated C0 2 pump injectors are 
available, these are expensive to purchase. 
Cheaper alternative techniques, not requiring a 
dedicated pump injector, have been described 
and used with complete safety if appropriate 
measures are utilized (Snow and Rice, 1999). 
Stacking software is required to overcome frag- 
mentation of the C0 2 gas column by the flowing 
blood (Fig. 4.1). For adequate imaging of the 
tibial and distal arteries, elevation of the feet by 




> 



28 



VASCULAR SURGERY 




Figure 4.1. Carbon dioxide angiography showing the application of stacking software to remove the beading effect seen in panels 
AandB. 



10 to 20 degrees may be necessary. Unlike 
iodine-based agents, C0 2 can be administered in 
large quantity, even in patients with pulmonary 
disease, without any adverse effect. The C0 2 gas 
displaces the blood and appears radiolucent in 
relation to surround tissue (negative contrast 
agent). Unfortunately, due to its low density, C0 2 
angiography has a lower contrast resolution 
compared to iodine-based agents and hence 
produces a poorer image quality, particularly 
in the small peripheral vessels. Furthermore, 
C0 2 gas can cause the vapor lock phenom- 
enon, a condition in which the gas is collected 
over the most anterior aspect of an artery, 
in particular the aorta, causing poor visualiza- 
tion of the mesenteric arteries and producing 
temporary abdominal pain, thought to be sec- 
ondary to bowel ischemia. Finally, C0 2 angiog- 
raphy is contraindicated for "above-diaphragm" 
angiography, because of the risk of cerebral 
embolization. 

Gadolinium chelate, an MRI contrast agent, 
can also be used as an intravascular contrast 
media. However, it is a poor radiographic con- 
trast agent, and it should not be used in patients 
with renal impairment, as there is evidence that 
at the doses required for conventional angiog- 



raphy (as opposed to MRI) it is potentially more 
nephrotoxic than iodinated contrast (Thomsen 
et al., 2002). It can be useful in patients with 
other contraindications to iodinated agents, uti- 
lized as a problem solver to answer specific 
questions that cannot adequately be defined by 
C0 2 angiography. Because of dose limitations 
(maximum 0.3mmol/kg), careful planning is 
required prior to its use. 

Even with the introduction of smaller 
catheters and safer contrast agent, the role of 
contrast catheter angiography as the primary 
investigation tool of vascular diseases is dimin- 
ishing due to its many drawbacks (Table 4.1). In 
the future, it will be employed mainly as part of 
an interventional procedure or in special diag- 
nostic situations. 

Computed Tomography 

Until relatively recently, the images obtained 
from CT examinations were of suboptimal 
quality for the assessment of most vascular dis- 
eases except aneurysmal conditions. This was 
due to the long examination time (minutes) and 
thick slice width (5 to 10 mm); hence, images 
were subjected to movement artifacts and were 



29 



RADIOLOGICAL INVESTIGATIONS 




Table 4.1. Complications of angiography 

Contrast medium-related complications 
Minor adverse reactions: common, but rarely life 

threatening (e.g., urticaria, nausea) 
Major adverse reactions: rare, but serious 

(idiosyncratic anaphylactoid reactions) 
Local vascular changes (e.g., effects on red 

cells/coagulation) 
Systemic vascular changes (e.g., increases in blood 

volume) 
Individual organ toxicity (e.g., renal) 
Access site complications 
Hemorrhage 

Intramural or perivascular injection of contrast 
Vascular thrombosis (following dissection or local 

trauma) 
Peripheral embolization 
Vascular stenosis or occlusion 
Pseudoaneurysm formation 
Arteriovenous fistula 
Local infection 
Nerve damage 

Damage to other local structures 
Catheter and general complications 
Air embolism 

Catheter thrombus embolization 
Dissection or perforation of vessels 
Organ ischemia or infarction secondary to spasm, 

dissection, or embolization 
Fracture and loss of guidewire or catheter fragments 
Catheter knot formation 
Mistaken injection of toxic material (e.g., skin 

cleansing agent) 
Vasovagal reactions 
Adverse reaction to local anesthetic or other drugs 



of low spatial resolution. These shortcomings 
prohibited the high-quality reconstruction 
often necessary for vascular studies. However, 
the introduction of spiral/helical CT shortened 
the examination time dramatically to less than 
30 seconds and allowed the use of thin collima- 
tion (i.e., slice thickness). This short acquisition 
time meant the examination could be per- 
formed in a single breath-hold, reducing the 
motion artifact that was frequently encountered 
with older scanners. Thin collimation in turn 
improved spatial resolution, further enhancing 
image quality. These factors were further 
improved with the introduction of multislice/ 
detector spiral CT. In comparison to the first- 
generation spiral CT scanners that have a single 
bank of x-ray detector, multidetector scanners 



have multiple banks of detectors (up to 64), 
allowing several slices to be acquired with 
each revolution of the scanner. The volumetric 
data obtained from recent scanners can be 
processed and displayed as multiplanar refor- 
mats (MPRs), maximum intensity projections 
(MIPs), or surface shaded displayed (SSD) (Fig. 
4.2). Details about these reconstructions are 
beyond the scope of this chapter, but it is prob- 
ably fair to say that MPR is most frequently used 
clinically. 

With the new generation of multidetector 
scanners, the best spatial resolution of the 
images obtained is around 0.5 mm, which is ade- 
quate for assessing most vascular systems. 
However, as with contrast angiography, the 
main limitation of CT examinations is the 
requirement of large volumes (100 to 150mL) of 
potentially nephrotoxic iodine-based contrast 
agent even with the new generation machines. 
Alternative contrast agents such as gadolinium 
can be used for CT, but produce less contrast 
and are more expensive. 

Currently, the main roles of CT in relation to 
vascular diseases are (1) diagnosis of aortic 




Figure 4.2. Surface shaded reconstruction showing the rela- 
tionship of an infrarenal abdominal aortic aneurysm to the bony 
landmarks. 




30 



VASCULAR SURGERY 



aneurysms, (2) assessment for open or en- 
dovascular treatment of aortic aneurysm, (3) 
endovascular stent graft surveillance, and (4) 
vascular trauma. Although CT provides accurate 
imaging of visceral and peripheral arteries, 
magnetic resonance angiography (MRA), which 
has none of the disadvantages of CT (i.e., 
nephrotoxic contrast agent and ionizing radia- 
tion), is more widely accepted by clinicians. 
Because magnetic resonance provides poor 
visualization of calcified arteries, a combination 
of MRA and noncontrast CT is often required in 
these cases. 

Magnetic Resonance Angiography 

Prior to the advent of gadolinium-enhanced 
three-dimensional (3D) MRA by Prince and 
coworkers, MRA played a small role in the inves- 
tigations of vascular diseases, mainly because 
the images obtained were of poor quality due to 
movement and turbulence-induced artifacts 
(Prince et al., 1993). The use of gadolinium 
chelate as a contrast agent shortens the exami- 
nation time significantly, allowing the examina- 
tion to be performed in a single breath-hold, 
reducing movement artifacts. In addition, 
gadolinium-enhanced MRA is not susceptible to 
the flow artifacts that plague noncontrast MRA. 
The data range in 3D MRA is acquired as a 
continuous volume, and this allows 3D recon- 
struction of the images, producing images com- 
parable to catheter angiography. The resolution 
limit of current MR scanners is approximately 1 
mm, which is adequate for the imaging of most 
arteries, including the pedal vessels. Visualiza- 
tion of smaller vessels is software dependent, 
however, and MR images of distal vessels are 
limited by contrast issues, as the gadolinium 
dilutes as it travels distally and enhances the 
veins soon after the distal arteries. Despite these 
issues, MRA of the infrainguinal vessels can be 
performed with adequate quality for planning 
distal bypasses in centers with very high quality 
MRI. 

The main advantage of MRA, in relation to 
CT, is the use of gadolinium as a contrast agent, 
which is safe and does not cause renal impair- 
ment, except in rare cases. The current limita- 
tions of MRA are high cost, lack of availability, 
patient claustrophobia, narrow range of field 
of view (for example, it is unable to image the 
entire aorta in one continuous acquisition), and 



poor display of intraluminal thrombus and 
calcification of the arterial wall. The latter two 
limitations are important to consider in 
endovascular procedures, such as aneurysm 
stent grafting. In addition, MRA has the ten- 
dency to over-grade the degree of stenosis. 
Hence, in most units, confirmatory catheter 
angiography is routinely performed prior to 
intervention in cases where the MRA has shown 
significant stenotic disease. 

Currently, the main roles of MR are for the 
investigations of aortoiliac disease, visceral 
arteries, and extent of arteriovenous malforma- 
tions. In addition, as it involves no ionizing 
radiation, MR is the most appropriate imaging 
modality for young patients requiring long- 
term follow-up, such as those with aortic dis- 
section, aortic root replacement surgery, or 
aortic coarctation. Although MR is not widely 
used at the moment as the primary imaging 
modality for vascular disease, with further 
advances in software and hardware designs 
expected in the future, it will become an ideal 
imaging tool. 



Radiation Safety 



X-rays are an ionizing radiation, interacting 
with water molecules, producing radicals that 
cause cellular damage and death. The effect can 
be immediate, for example, skin necrosis, or late, 
such as genetic mutation or cancer formation. It 
is important to bear in mind that radiation 
exposure is cumulative and permanent; hence, 
it is prudent that the exposure to x-rays be 
minimized. There are several simple methods 
to reduce the exposure to both patients and 
operators: 

1. Radiation exposure is proportional to 
fluoroscopy time, and hence the most 
effective way to reduce exposure is to 
reduce the fluoroscopy time. Use pulse 
mode rather than continuous mode when 
possible and plan cases in advance to 
obtain only the necessary views. 

2. Increase the distance from the source. 
Exposure decreases with the square of the 
distance from the source (inverse square 
law). 

3. Use lower magnification and careful 
collimation. 



31 



RADIOLOGICAL INVESTIGATIONS 




4. Use posterior-anterior imaging, that is, the 
x-ray source is from the posterior of a 
supine patient. 

5. Position the image intensifier as close as 
possible to the patient to reduce scatter. 

6. Use protective barriers such as table aprons, 
lead glass shields, as well as the usual lead 
apron, thyroid shield, andglasses. 

The use of dosimeter badges by all persons 
working with ionizing radiation is mandatory. 
In the United Kingdom, the badge must be posi- 
tioned at waist level under the lead apron. Addi- 
tional badges can be worn (such as on the 
fingers or forehead) for specific exposed areas. 
In the United States, badges are required either 
outside the apron at the neck level, beneath the 
lead apron, as in the U.K., or in both locations 
(with finger badge an option). In the U.K. and 
U.S., no individual working in the controlled 
area should receive doses in excess of (1) 
20mSv/year to the body, (2) 150mSv/year to the 
lens, and (3) 500mSv/year to hands or forearm. 
Finally, although the use of dosimeters is man- 
dated, it is the responsibility of the operator to 
wear and use them correctly. 



Investigations of 
Vascular Diseases 

This section is divided into system-based cate- 
gories. Common conditions and appropriate 
investigations for each system are discussed. 

Peripheral Vascular Disease 
(Lower Limb) 

This is the most common of all vascular condi- 
tions, accounting for the majority of the work 
load in any vascular unit. Clinically, this condi- 
tion is best divided into two subcategories: (1) 
patients with symptoms of claudication without 
any tissue loss or rest pain; and (2) patients 
with rest pain or tissue loss, suggesting limb- 
threatening ischemia. In those patients with 
symptoms that are severe enough to warrant 
intervention, further investigation is justified. 
However, most claudicants do not require inter- 
vention and are treated conservatively, and thus 
further imaging is not necessary. In most cases, 



duplex examination is adequate as a first-line 
investigation for the demonstration of the exter- 
nal iliac arteries, femoral arteries, and even as 
distally as the tibial trifurcation. Conventional 
catheter angiography, MRA, or CT angiography 
can be performed if the examination is of 
suboptimal quality or if the assessment of 
distal arteries is required, for example in cases 
where distal bypass surgery is considered. This 
depends on staff expertise and equipment avail- 
ability in individual centers. In some centers 
distal bypass decisions can be based on duplex 
arterial mapping, MRA or even CT angiography 
(CTA), but currently most units still depend 
on catheter angiography. In patients with sus- 
pected aorto iliac disease, duplex examination is 
more difficult due to anatomical constraints. In 
this situation, MRA or CTA is generally pre- 
ferred. If these two options are not available, an 
angiogram can be performed via a catheter that 
is positioned in the distal abdominal aorta. If 
brachial or radial artery access is used, access 
should be obtained from the left arm to mini- 
mize the risk of embolic complications to the 
cerebral circulation. 

Catheter angiography is useful to diagnose 
vasculitic conditions such as thromboangiitis 
obliterans (Buerger disease). This condition 
classically affects small and medium-size arter- 
ies and veins of the lower limbs in young 
smokers with a typical angiographic appear- 
ance. In patients with blue digit syndrome, 
catheter angiography may show the source of 
emboli, typically atherosclerotic plaques in the 
aorta or iliac arteries. However, CT or other 
noninvasive modalities are preferred for the 
initial evaluation in blue digit syndrome, 
because the catheter-based modalities carry the 
risk of further athero emboli. 

Finally, although conventional catheter 
angiography plays an ever-diminishing role as 
the primary diagnostic tool in the investigation 
of peripheral vascular disease (PVD), it still has 
a few advantages in comparison to noninvasive 
techniques. Direct pressure measurement can 
be performed in equivocal stenosis (>10mmHg 
systolic gradient is considered significant), with 
the use of enhanced gradients in cases of possi- 
ble "subcritical" stenosis [by the use of distal 
vasodilators such as glyceryl trinitrate (nitro- 
glycerin in the U.S.) or papaverine], as well as 
allowing intervention to be performed in the 
same sitting. 




» 



32 



VASCULAR SURGERY 



Upper Limb Vascular Diseases 

Upper limb vascular diseases are far less fre- 
quently encountered than those of the lower 
limb. These can usually be attributed to subcla- 
vian steal syndrome, embolic disease, or blue 
digit syndrome (distal embolization), and rarely 
due to diffuse atherosclerotic disease commonly 
seen in the lower limbs. Duplex ultrasonogra- 
phy is useful as a preliminary test. Although it is 
anatomically not possible to visualize the first 
part of the subclavian artery directly, waveform 
analysis of flow in the distal segments may 
reveal changes (such as damped signal or spec- 
tral broadening) that are suggestive of proximal 
occlusion or stenosis. In addition, duplex exam- 
ination may detect flow reversal in the vertebral 
artery, which suggests a steal phenomenon and 
is a good indication of significant ipsilateral 
proximal subclavian artery stenosis or occlu- 
sion. Magnetic resonance angiography is widely 
accepted for the visualization of arch vessels, but 
aortic pulsation artifact may obscure anatomi- 
cal detail at the origins of the great vessels. Com- 
puted tomography angiography is useful but 
subjected to beam-hardening artifacts from the 
thoracic cage and contrast in the venous system, 
both of which are in close proximity to the 
arteries of interest. Ultimately, catheter angiog- 
raphy is still the definitive test for the investiga- 
tions of patients with upper limb ischemia. It is 
pertinent that a flush aortic arch angiogram is 
performed in two planes (left and right anterior 
oblique), as origin disease is frequently missed 
if only one projection is obtained. 

A rare cause of upper limb ischemia is 
Takayasu arteritis (pulseless disease), a condi- 
tion most commonly seen in young female 
Asians. It is a granulomatous disease involving 
major arteries, in particular the aortic branches 
and pulmonary arteries. Clinically, the patient is 
systemically unwell with symptoms and signs of 
limb ischemia or renovascular hypertension. 
Radiological features suggestive of the condi- 
tion are enhancing thickened arterial wall on 
CT, segmental stenotic or occlusive disease of 
major arteries, and aneurysm formations. 

Carotid Arteries 

Duplex ultrasonography examination is the pre- 
liminary test for patients with suspected carotid 
artery stenosis in whom intervention is consid- 



ered appropriate (see Chapter 3). The procedure 
is relatively easy to perform with good accuracy 
in experienced hands. In addition, it provides 
morphological assessment of the plaque. Hence, 
in many vascular centers, duplex examination 
is the sole radiological investigation prior to 
carotid endarterectomy However, when endo- 
vascular treatment is an option, it is important 
that the entire carotid artery is assessed for 
synchronous stenotic disease, tortuosity, and 
a favorable angle of origin of the vessel to be 
treated (Fig. 4.3). These factors determine if 
endovascular treatment is feasible. Unfortu- 
nately, duplex examination cannot answer all 
these questions, and in our unit we presently 
rely on conventional arch aortography for full 
assessment (selective catheterization should not 
be required). However, it is likely that as 
improvements in MRA and CTA continue to be 
made, the full characterization of carotid steno- 
sis, the arch vessels, and cerebral circulations 
will be of sufficient quality to eliminate the 
requirements for catheter angiography to deter- 
mine whether a patient is a good candidate for 
endovascular intervention. 

Carotid body tumor is a rare condition that 
classically presents as a painless pulsatile mass 
below the angle of the jaw, and is laterally 
mobile but fixed vertically. Approximately 10% 
of cases are bilateral. The typical ultrasonic 
feature is an oval mass splaying the carotid 
internal and external carotid arteries. This mass 
enhances avidly on CT or MR examinations. 

Pulmonary Arteries 

There are few medical conditions that require 
the imaging of the pulmonary vasculature. Most 
frequently this is required for suspected acute 
pulmonary embolic (PE) disease or as part of 
the investigation of pulmonary hypertension, to 
exclude chronic thromboembolism. In the 
majority of cases with suspected acute PE, the 
initial investigations should consist of a chest 
radiograph and a radionuclide ventilation- 
perfusion (V/Q) scan or a CTA. Unfortunately, 
up to 50% of V/Q scans are of no diagnostic 
value, and in these circumstances supplemen- 
tary investigations are required. Previously, 
catheter angiography was the imaging modality 
of choice, but with the advent of spiral CT the 
role of catheter angiography is limited to situa- 
tions such as massive PE, where immediate 



33 



RADIOLOGICAL INVESTIGATIONS 




Figure 4.3. Tortuous common and 
internal carotid arteries (arrow), a 
contraindication to carotid artery 
stenting. 




intervention may be required. Although CTA is 
undoubtedly accurate for the diagnosis of mod- 
erate to large emboli, it is limited by its inabil- 
ity to reliably diagnose subsegmental embolism, 
particularly with the older single-slice spiral 
scanners. However, the clinical significance of 
subsegmental PE is uncertain and may have 
only minimal impact in the long-term outcome. 
The results of ongoing outcome studies where 
patients are managed on the basis of multide- 
tector/slice helical CT findings are eagerly 
awaited. 

Magnetic resonance angiography has not 
been thoroughly evaluated as a tool for the diag- 
nosis of PE,but a recent study showed that it has 
reasonable accuracy in comparison to catheter 
angiography (Haage et al., 2003). However MRA, 
as with CTA, is limited by the poor visualization 
of subsegmental arteries, and resolution tends 
to be approximately half that of CT in most 
institutions. Magnetic resonance angiography is 
unlikely to be widely used for the diagnosis of 
PE until further data are available (Haage et al., 
2003). 

In certain situations, the catheter pulmonary 
angiogram remains the investigation of choice. 
Patients with acute massive PE and cardiorespi- 
ratory instability should proceed directly to 
catheter angiogram, where treatment such as 
thrombectomy or thrombolysis can be adminis- 



tered immediately following the confirmation of 
the diagnosis. Furthermore, it is possible to 
insert an IVC filter at the same time to prevent 
further embolism, if this remains a risk. 
Pulmonary angiography is also indicated in 
patients in whom noninvasive diagnostic tests 
remain inconclusive and a reasonable suspicion 
for PE exists. In this situation, pulmonary 
angiography is aimed at excluding PE, thus 
allowing withdrawal of anticoagulant therapy 
with its inherent bleeding risk. In patients in 
whom the consequence of bleeding is unfor- 
giving, for instance following recent neuro- 
surgery, pulmonary angiography can provide a 
definitive diagnosis. The disadvantages of pul- 
monary angiography are that it is invasive and 
uses nephrotoxic contrast agents. Nevertheless, 
the safety of this technique has improved dra- 
matically over the past 10 years. Current esti- 
mates of mortality and morbidity are around 
0.03% and 0.47%, respectively, in experienced 
hands. 

Pulmonary arteriovenous malformation 
(AVM) is a rare condition in which there is an 
abnormal large vascular communication 
between the pulmonary artery and vein. This 
allows shunting of blood from the pulmonary 
circulation to the systemic circulation and 
hence the risk of complications such as cere- 
brovascular accident and cerebral abscess. The 




» 



34 



VASCULAR SURGERY 




Figure 4.4. Coil embolization of pulmonary arteriovenous malformation (AVM). A: Pulmonary angiogram shows an AVM in the 
periphery of left lobe (arrow). B: Selective angiogram demonstrates the characteristic early draining vein. C: Angiographic appear- 
ance after successful embolization. 



AVM can be solitary (40% of cases) or multiple, 
often associated with hereditary hemorrhagic 
telangiectasia (Rendu-Osler-Weber syndrome). 
Diagnosis can be reliably confirmed by CT or 
MR examinations, showing a vascular mass with 
a large feeding artery and draining vein (Fig. 
4.4). Catheter angiogram is indicated only as a 
part of the current treatment of choice, i.e., 
embolotherapy 

Mesenteric Arteries 

The main indications for the investigation of 
mesenteric arteries are gastrointestinal (GI) 
bleeding, intestinal angina, and mesenteric 
artery aneurysms or pseudoaneurysms. For GI 
bleeding, endoscopy is the first-line investiga- 
tion. If the bleeding site or cause is not identified 
(up to 40% of endoscopic examinations), the 
patient should have a radionuclide test if the 
hemorrhage is not life threatening, or urgent 
selective catheter angiography if the patient is 
hemodynamically unstable and actively bleed- 
ing. Radionuclide investigations are more 
sensitive than catheter angiography for the 
investigation of GI bleeding (isotope remains in 
the vascular system far longer than contrast 
agent), but less accurate in identifying the exact 
anatomical site or the nature of the disease. 



Technetium 99m ( 99m Tc) sulfur colloid or 99m Tc- 
labeled erythrocytes are used to localize mild- 
to-moderate intermittent bleeding, particularly 
in the lower GI tract, and 99m Tc pertechnetate is 
used to detect Meckel's diverticulum. Selective 
catheter angiography may reveal the exact site 
of hemorrhage but this is highly dependent on 
the timing of the procedure. Many investiga- 
tions turn out to be false negatives (reported 
accuracy of 40% to 90%), as the patients are not 
actively bleeding at the time of investigation. 
Nonetheless, the tests may still show areas of 
abnormal vascularity such as angio dysplasia. In 
addition, endovascular treatment such as 
embolization or vasopressin infusion may be 
initiated, and, it is hoped, avoid the high mor- 
bidity and mortality (up to 30%) of emergency 
surgery. The treatment may permanently stop 
the bleeding or allow a window of opportunity 
to stabilize the patient prior to definitive surgi- 
cal resection if deemed necessary. 

Although atheromatous disease of the mesen- 
teric arteries is common, mesenteric angina is a 
rare entity. This is because there are numerous 
large collaterals between the mesenteric arter- 
ies, and hence the symptoms of mesenteric 
angina only manifest if there is severe stenosis 
or occlusion in two of the three mesenteric 
arteries (celiac, superior mesenteric, and infe- 



35 



RADIOLOGICAL INVESTIGATIONS 




rior mesenteric arteries). The first-line investi- 
gation should be duplex ultrasonography, as it 
has excellent diagnostic value in many institu- 
tions. The origins of celiac and superior mesen- 
teric artery are relatively straightforward to 
assess. However, the inferior mesenteric artery 
is frequently not seen due to overlying bowel 
gas. Because of this, many radiologists prefer 
MRA as the primary investigational modality 
for this condition. The 3D acquisition allows 
accurate assessment of the ostial disease with 
sensitivity and specificity comparable to that of 
catheter angiography. Computed tomography 
angiography is also being used as an adjunctive 
diagnostic modality in some institutions, pri- 
marily to aid in evaluation of calcific aortic 
plaque that is not seen well on MRA, but may 
have a substantial impact on the approach to 
surgical intervention. Currently, catheter angio- 
graphy is usually reserved for cases in which 
other imaging modalities are equivocal, or when 
endovascular intervention is considered. 

Mesenteric artery aneurysm is an uncommon 
condition, and is best assessed with cross- 
sectional studies such as CT. This allows the 
identification of the vessel of origin, the size of 
the aneurysm, and the mass effect on the sur- 
rounding structure. Catheter angiography can 
then be performed as part of the embolization 
treatment or for evaluation of potential 
aneurysms in the mesenteric arcades. 



Renal Arteries 

The main indications for imaging of the renal 
arteries are (1) diagnosis of renal artery steno- 
sis, most commonly atherosclerotic or fibro- 
muscular dysplasia (FMD); and (2) evaluation 
of potential live renal donors. Although conven- 
tional catheter angiography is still considered 
the gold standard, it involves the use of nephro- 
toxic contrast agent in a group of patients with 
a high incidence of preexisting renal impair- 
ment, and hence is employed only in cases 
where other tests are unavailable or inconclu- 
sive. In many institutions MRA is currently the 
investigation of choice in patients with sus- 
pected renovascular disease, primarily due to 
the lack of nephrotoxicity with gadolinium- 
chelate. It has high sensitivity and specificity for 
the assessment of ostial disease, but is unproven 
for distal stenosis such as in FMD. In our unit, 



patients with suspected FMD have MRA as the 
first-line investigation, followed by catheter 
angiography if the MRA is of suboptimal 
quality or there is strong clinical suspicion of 
FMD. Although CTA is accurate for assessing 
renal artery stenosis, it is subjected to a similar 
drawback as catheter angiography, that is, 
requiring iodinated contrast media and ionizing 
radiation. Duplex examination is the initial 
imaging modality of choice in many institutions 
due to its low cost and relatively high accuracy 
for renal artery stenosis, but it is more prone to 
missing duplicate renal arteries, and it is highly 
operator- and patient-dependent, with up to a 
15% technical failure rate. 

With the current shortage of cadaver renal 
donors, living-related renal transplant surgery 
has become a common procedure. Previously, 
the donors had to undergo catheter angiogra- 
phy, ultrasonography, and intravenous urogra- 
phy as part of the assessment. Currently, MR is 
the imaging method of choice as it provides the 
most comprehensive and accurate assessment of 
the donor in one sitting. It allows anatomical 
and morphological evaluation of the renal 
parenchyma, veins, arteries, and ureters with 
confidence. In addition, it may show other 
intraabdominal pathologies such as adrenal or 
pelvic masses. Computed tomography angiog- 
raphy with 3D reconstruction can also be used 
in patients who are known to have normal renal 
function, as is true for most potential renal 
donors. 

Aortic Aneurysms and Dissections 

The conventional treatment for patients with 
aneurysmal disease of the aorta is surgical. 
Patients with abdominal aortic aneurysms 
(AAAs) or thoracic aortic aneurysms of greater 
than 5.5 or 6.5 cm in diameter, respectively, 
are generally best treated, depending on the 
patient's fitness for surgery, because of the 
increasing risk of rupture above these sizes. In 
most cases, CT examination alone is adequate 
to provide all the information needed prior to 
open surgical repair. Computed tomography 
shows the relation of visceral arteries to the 
aneurysm sac, the size and extent of the 
aneurysm, and other conditions that may pre- 
clude surgical repair, such as horseshoe kidney. 
In the early 1990s endovascular aneurysm 
repair (EVAR) revived the role of catheter 




» 



36 



VASCULAR SURGERY 



angiography for the assessment of aortic 
aneurysms. Catheter angiography began to 
diminish again, however, as spiral CT with 3D 
reconstruction and specialized software in the 
mid-1990s allowed high-quality reconstructions 
not previously possible. Accurate preoperative 
imaging is crucial to ensure proper selection of 
patients, stent-graft types, and potential intra- 
operative adjuncts such as coil embolization, 
iliac angioplasty, renal stenting, femoral 
endarterectomy, and iliac conduits. To eliminate 
the need for catheter angiography preopera- 
tively, work focused on 3D reconstruction of CT 
and MR data, with specialized software for 
detailed measurements and even endograft 
simulations preoperatively Currently, CT or MR 
with 3D reconstruction and computer-aided 
measurement, planning, and simulation is the 
standard for imaging prior to EVAR, with meas- 
urement accuracy and outcomes equal or supe- 
rior to catheter angiography and without the 
morbidity or expense (Broeders et al., 1997; 
Fillinger, 1999, 2000). The main disadvantages of 
MR are the difficulty in visualizing thrombus 
and calcification, important points to be con- 
sidered, as thrombus in the neck of the 
aneurysm may preclude endovascular repair. 
Nevertheless, future advances in MR technolo- 
gies are expected to resolve these problems. 

Although the technical success of stent-graft 
implantation is well established, only modest 
data are available for midterm results, and data 
on long-term efficacy and safety are not yet 
established. The goals of postprocedure imaging 
are to (1) confirm appropriate stent-graft place- 
ment, (2) assess the effectiveness of aneurysm 
exclusion, (3) follow the aneurysm sac size, and 
(4) detect device failure and/or migration. 
Because late complications have been observed, 
lifelong follow-up is felt to be essential at the 
present time. The current imaging strategy 
should include plain radiographs of the stent 
graft in four projections (anterior-posterior, 
lateral, and two obliques) and CT angiography 
at 1, 6, and 12 months postoperative, and then 
annually. More or less frequent imaging may 
be recommended depending on the imaging 
results (e.g., endoleak or migration), type of 
stent graft, and manufacturer recommenda- 
tions. Plain radiographs are an excellent and 
inexpensive means to assess the metallic stent- 
graft frame for structural failure, angulation, 
and kinking. Plain radiographs have typically 



been recommended for establishing migration, 
but this is best performed on CT. Triple-phase 
CT examinations (nonenhanced, arterial phase, 
and delayed "venous" phase) allow serial meas- 
urements of aneurysm sac diameters, monitor- 
ing of the integrity and position of the device, 
and detection of an endoleak (reperfusion of 
the aneurysm sac) (Fillinger, 1999). Three- 
dimensional reconstruction with volume 
measurements is the most sensitive method 
for detecting aneurysm sac size changes, but 
requires expertise in image segmentation or 
outsourcing to a commercial entity. It may allow 
earlier detection of problems that require inter- 
vention after EVAR or earlier reassurance to the 
patient and less frequent surveillance in suc- 
cessful repairs (Fillinger, 1999; Kay et al., 2003). 
Duplex ultrasound is a useful adjunct for the 
detection of endoleak, without the risk of con- 
trast nephrotoxicity and ionizing radiation. 
However, it may not show the source of 
endoleak and is unable to assess stent-graft 
integrity accurately. Three-phase contrast- 
enhanced MRA serves as an attractive alterna- 
tive to CTA in patients with renal impairment 
and relative contraindications to iodinated con- 
trast media. There is evidence that it is superior 
to CT in depicting small type 2 (collateral) 
endoleaks. Unfortunately, it is not suitable for 
stainless steel devices, which cause severe image 
degradation secondary to metal artifacts. Cur- 
rently, the role of catheter angiography is to 
characterize endoleaks (inflow and outflow 
channels) detected by the noninvasive tests or to 
further evaluate sacs that appear to have 
enlarged without an endoleak. 

Aortic dissection is a condition in which a 
spontaneous tear of the tunica intima allows cir- 
culating blood to gain access to the tunica 
media, splitting it longitudinally. The objective 
of imaging is not only to diagnose the condition 
but also to localize the site of entry, assess the 
extent of dissection, and identify associated 
complications. The potential complications that 
may occur are occlusion of major aortic 
branches such as coronary and visceral arteries; 
aortic valve insufficiency; rupture into the peri- 
cardial sac, mediastinum, or pleural cavity; 
and aneurysm formation in the long-term. At 
present, the imaging modality of choice for 
acute dissection is CT angiography. It has excel- 
lent sensitivity and specificity in comparison to 
catheter angiography, which is considered the 



37 



RADIOLOGICAL INVESTIGATIONS 




standard of reference. However, it is vital that 
the examination be performed in the arterial 
phase of contrast enhancement, as failure to do 
so may lead to misinterpretation of a dissection 
as an aneurysm. Transesophageal ultrasonic 
examination is a useful adjunct, showing the 
presence or absence of aortic valve incompe- 
tence and pericardial effusion, and confirming 
the presence of the intimal flap. Magnetic reso- 
nance is as accurate as CT, if not more so, in 
diagnosing acute dissection. However, it is 
rarely employed because of the lack of immedi- 
ate availability, the delay from bedside to 
scanner, the long examination time, the limited 
access to the patient, and the restricted moni- 
toring of vital signs, which is especially prob- 
lematic in these often hemodynamically 
unstable patients. 

Vascular Trauma 

Vascular injuries can occur in a number of ways, 
such as motor vehicle accidents, knife or 
gunshot wounds, or iatrogenically In most 
cases, the history and clinical features provide 
the diagnosis. Hemodynamically unstable 
patients usually proceed immediately to the 
operating room for control of the hemorrhage. 
If the patient is stable enough, imaging can be 
used to confirm hemorrhage or injury, localize 
the site, and assess the severity. In addition, 
treatment options can be planned from the 
information acquired, and in some cases 
catheter-based therapeutic intervention can be 
performed. Direct vascular injuries in a stable 
patient are often best investigated initially by 
CT examination. One major advantage of con- 
trast-enhanced CT over ultrasound is the ability 
to identify the exact site of bleeding and poten- 
tially help plan the approach for surgical or 
endovascular intervention. 

Aortic injury occurs most often following a 
rapid deceleration injury, and the vast majority 
of cases are due to motor vehicle accidents. 
Complete rupture accounts for 85% of cases, 
and most patients do not survive. The remain- 
ing 15% have incomplete rupture (contained 
rupture) and they require immediate treatment, 
as half of them will progress to complete 
rupture within 24 hours. The most common site 
of injury (90%) is at the aortic isthmus, just 
distal to the left subclavian artery. The prelimi- 



nary investigation is a chest radiograph. It may 
show features suggestive of transection, such as 
a widened mediastinum, apical cap, and dis- 
placement of the trachea, left main bronchus, or 
nasogastric tube. A normal chest x-ray does not 
exclude transection, but will diagnose condi- 
tions such as pneumothorax or hydrothorax. 
Arterial phase contrast-enhanced spiral CT with 
fine collimation is the most widely used modal- 
ity for the imaging of aortic transection. Fea- 
tures of aortic injury are the presence of an 
abrupt change in aortic contour, false aneurysm, 
intimal flap, or extravasation of contrast. Medi- 
astinal hematoma is a frequent finding in trau- 
matic chest injury, but in the majority of cases, 
the source of hematoma is the azygos or hemi- 
azygos veins and paraspinal and intercostals 
vessels rather than aortic injuries. Although 
catheter angiography is still considered the 
standard of reference for investigation of aortic 
transection, it is employed only when the CT 
examination is equivocal or as part of the 
endovascular treatment. When performing 
catheter angiography, it is vital that at least two 
projections of the arch and descending aorta be 
obtained prior to pronouncing the investigation 
normal. In addition, an awareness of ductus 
diverticulum is required. This is the vestigial 
remnant of the ductus arteriosus and is present 
in approximately 10% of the population. This 
anatomical variant appears as a small bulge on 
the medial wall of the aorta, just inferior to the 
origin of the left subclavian artery, and is fre- 
quently misinterpreted as a false aneurysm. 

Conclusion 

We have presented an overview of available 
imaging techniques in use in vascular radiolog- 
ical practice, emphasizing the move away from 
invasive techniques and toward noninvasive 
techniques for diagnosis of vascular diseases. 
The most important contemporary approaches 
and therapies have also been described, 
showing the important role these techniques 
now play in the management of a range of vas- 
cular disease processes. 

References 

Aspelin P, Aubry P, Fransson SG, Strasser R, Willenbrock R, 
Berg KJ. (2003) N Engl J Med 348:491-9. 




> 



38 



VASCULAR SURGERY 



Broeders IA, Blankensteijn JD, Olree M, Mali W, Eikelboom 

BC. (1997) J Endovasc Surg 4:252-61. 
Dormandy JA, Rutherford RB. (2000) J Vase Surg 31:S1- 

S296. 
Fillinger ME (1999) Surg Clin North Am 79:451-75. 
Fillinger ME (2000) Semin Vase Surg 13:247-63. 
Haage P, Piroth W, Krombach G, et al. (2003) Am J Respir 

Crit Care Med 167:729-34. 



Kay J, Chow WH, Chan TM, et al. (2003) JAMA 289:553-8. 
Prince MR, Yucel EK, Kaufman JA, Harrison DC, Geller SC. 

(1993) J Magn Reson Imaging 3:877-81. 
Snow TM, Rice HA. (1999) Clin Radiol 54:842-4. 
Thomsen HS, Almen T, Morcos SK. (2002) Eur Radiol 12: 

2600-5. 



5 



Bleeding and Clotting Disorders 

Vivienne J. Halpern and Frank C.T. Smith 




Bleeding and clotting disorders have major 
implications for the effective management of 
vascular patients. Such disorders can influence 
disease progression, perioperative complica- 
tions, graft patency, limb salvage, and wound 
healing. Most of these disorders are relatively 
rare, and diagnosis requires an index of clinical 
suspicion combined with a need to obtain a 
relevant medical history and appropriate 
specialized investigations. Correct and timely 
treatment may help prevent some of the com- 
plications associated with these disorders. This 
chapter reviews the presentation, diagnosis, 
and management of some of the more common 
disorders. Medications frequently associated 
with abnormal bleeding or clotting are also 
discussed. 



History, Physical and 
Laboratory Evaluation 

Recognition of a patient with a bleeding or 
clotting disorder involves taking an adequate 
history. A screening questionnaire, such as that 
provided in Table 5.1 is useful and may help to 
direct further investigation. Obtaining a history 
of mucosal bleeding involving epistaxis, gum 
bleeding, or menorrhagia may be more consis- 
tent with platelet disorders (thrombocytopenia, 
von Willebrand disease, etc.) than, for instance, 
bleeding into a joint or muscle, which occurs 
more commonly with hemophilia. Patients with 



a history of myeloproliferative, myelodysplastic, 
and lymphoproliferative disorders may also 
have increased bleeding through several mech- 
anisms, which may not appear in routine pre- 
operative testing. Renal failure predisposes to 
bleeding tendencies based on platelet dysfunc- 
tion, whereas, for instance, recent splenectomy 
may induce thrombocythemia predisposing to 
abnormal clotting. An abnormal history of clot- 
ting, such as multiple episodes of deep venous 
thrombosis (DVT), may warrant screening for 
thrombophilia. 

Medications can influence both bleeding 
and clotting. Commonly used drugs such as 
aspirin and other nonsteroidal antiinflamma- 
tory drugs (NSAIDs) affect platelet function, as 
do Aggrenox and Plavix. Herbal remedies and 
various vitamin combinations may also increase 
bleeding risks (Table 5.2). Furthermore, malnu- 
trition and vitamin deficiencies, such as vitamin 
C deficiency, may contribute to abnormal bleed- 
ing tendencies. Estrogen or estrogen-like med- 
ications including phytoestrogens predispose 
to thrombotic episodes. PC-SPES is an herbal 
preparation with several components used for 
treatment of prostate carcinoma. It contains 
phytoestrogens, which may induce thrombosis. 
However, it also contains Baikal skull cap 
(Scutellaria baicalensis georgi), which is a 
coumarin (a naturally occurring group of 
substances, structurally similar to warfarin). 
These examples illustrate the importance of 
querying the use of herbal medications as well 
as more conventional pharmacotherapies when 



39 




40 



VASCULAR SURGERY 



Table 5.1 . Screening survey for abnormal bleeding or clotting 

Do you suffer from a bleeding disorder? 

Do you have bleeding from the gums or from the nose? 

Have you ever coughed up or vomited blood? 

Do you notice easy or spontaneous bruising or does it take you a long time to stop bleeding when cut? 

Do you have excessive bleeding with menstrual cycles? 

Have you had any blood in the urine or with stools? 

Have you had any bleeding into muscles or joints? 

Have you had a tooth extraction or any other procedure after which bleeding has taken a long time to stop? 

Have you needed to receive any blood products, plasma, or vitamin K to help stop bleeding? 

Do you have any problems with your liver or kidneys? 

Has anyone in your family had any of the above problems? 

Have you or anyone in your family had a history of clots in the blood vessels, either artery or veins? 

Do you take oral contraceptives? 

Do you take aspirin or any medications for pain or arthritis? 

Do you take any steroid medications? 

Do you take any herbal medications or vitamins? 

Do you take Coumadin or other blood thinners? 

Do you take any medications to prevent stroke or heart attacks? 

Do you have any blood diseases? 



Table 5.2. Common medications, herbs, and vitamins associated with increased bleeding 



Medication 


Mechanism of action 


When to stop preoperatively 


Medications that may increase bleeding 




Aspirin 


Inhibits platelet aggregation via 


5-7 days before major surgery 




thromboxane B 2 


3-4 days before minor surgery 


Persantine 


Inhibits phosphodiesterase to increase 
cyclic AMP 


Omit dose before surgery 


Aggrenox 


Combined Persantine and aspirin: increased 
effects of aspirin 


As aspirin 


Plavix 


Irreversible binding to platelet inhibits ADP 
binding to platelet 


Stop 7-10 days before surgery 


NSAIDs 


Various mechanisms 


24 hours prior to surgery 


Herbs/vitamins that may 


increase bleeding 




Feverfew 


Used for migraines, ? inhibits platelet 
aggregation via thromboxane B 2 ,may be 
irreversible 


Stop at least 1 week before surgery 


Garlic 


Inhibits platelet function by inhibiting 
thromboxane synthesis; may be 
irreversible 


Stop at least 7-10 days before surgery 


Gingko 


Inhibition of platelet activating factor 


Stop at least 36 hours prior to surgery 


Ginseng 


Inhibits platelet aggregation; prolongs PT 
and PTT; may be irreversible 


Minimum of 24 hours prior to surgery 


Vitamin E 


May decrease platelet adhesiveness; may 


Unclear; should stop around 




effect vascular endothelium 


5 days before surgery 


Willow bark 


Salicylate precursors 


Stop 7-10 days before surgery, similar to aspirin 


Oil of wintergreen 


Affects platelet function 




Meadowsweet flower 







ADP, adenosine diphosphate; AMP, adenosine monophosphate; NSAID, nonsteroidal antiinflammatory drug; PT, prothrombin time; PTT, partial 
thromboplastin time. 



41 



BLEEDING AND CLOTTING DISORDERS 




assessing the patient with a bleeding or clotting 
disorder. 



Investigations 



Standard preoperative investigations for most 
vascular surgery patients include prothrombin 
time (PT), international normalized ratio (INR), 
activated partial thromboplastin time (aPTT), 
platelet count, and activated clotting time 
(ACT). A platelet count of 50,000/uL should 
ensure adequate hemostasis, whereas a count of 
10,000/|J,L or less may result in spontaneous 
bleeding. The aPTT evaluates the intrinsic and 
contact activation pathways of coagulation 
with the exception of factors VII and XIII. This 
investigation is used to monitor the effects of 
treatment with heparin. The extrinsic pathway 
(factors II, V, VII, and X, and fibrinogen) is 
evaluated with the PT and INR. These tests 
are employed to check the effectiveness of oral 
anticoagulation with warfarin. Additional tests 
should be directed by the history and clinical 
picture. 

The prevalence of inherited bleeding and 
clotting disorders is relatively rare. Patients with 
a recurrent, familial, or juvenile history of DVT; 
arterial thrombosis at a young age or without 
evidence of atherosclerosis; or thrombosis in an 
unusual location, such as mesenteric or cerebral 
veins, should be assessed for a hypercoagulable 
state. Recurrent graft failure whether in a bypass 
or an arteriovenous fistula, when not explained 
by the presence of an anatomical lesion, may 
also imply a prothrombotic state. Components 
of a thrombophilia screen include those listed in 
Table 5.3 (Donaldson et al, 1990). 

Patients with a history of abnormal bleeding, 
but not requiring medication, should undergo 
routine preoperative investigations as above. 
Excessive or prolonged bleeding during surgical 
procedures where routine preoperative testing 
of PT, aPTT, and platelet counts was normal may 
represent a platelet functional abnormality, a 
dysfibrinogenemia, factor XIII deficiency, vas- 
cular endothelial disorders, other mild factor 
deficiencies (if >25% of factor present), or oc- 
antiplasmin deficiency. Surreptitious use of 
medication might also be considered. Where 
abnormal preoperative investigations are 
encountered, there is a rationale for further 
tests. A protocol is suggested in Table 5.4. 




Table 5.3. Thrombophilia screen 

Protein C levels 
Protein S levels 
Antithrombin III 
Factor V Leiden 
Activated protein C 
Lupus anticoagulant 
Anticardiolipin antibody 
Antiphospholipid antibody 
Homocysteine levels 
Prothrombin 20210A mutation 
Factor VIII and XI levels 



Bleeding Disorders 

Bleeding disorders may be secondary to abnor- 
malities of plasma clotting factors, blood 
vessels, or platelets. Some hemostatic defects 
involve more than one of these systems. 



Table 5.4. Protocol for further investigation of abnormal 
preoperative blood tests 

Repeat abnormal PT/INR,aPTT with 50:50 mix with 

normal plasma (mixing study) 
If then normal, undertake the following investigations: 
Normal PT/INR, increased aPTT: 

Test for factor deficiency: usually isolated XI, IX, VIII 
Increased PT/INR, normal aPTT: 

Test for factor deficiency: isolated VII or can be 
multiple 

Test for liver abnormalities 

Look for vitamin K deficiency (malnourished patient, 
patient on prolonged antibiotics) 
Increased PT/INR and aPTT: 

Test for factor deficiency: isolated X, V, prothrombin, 
fibrinogen, or can be multiple 
If abnormal mixing study, undertake the following 

investigations: 
Normal PT/INR, increased aPTT: 

Test for inhibitor activity, especially for XI, IX, VIII 

Test for nonspecific inhibitors, e.g., antiphospholipid 
antibodies 
Increased PT/INR, normal aPTT: 

Test for factor VII inhibitor 

Test for nonspecific inhibitors (rarely cause isolated 
increase in PT/INR) 
Increased PT/INR and aPTT 

Test for inhibitors of X,V, prothrombin, fibrinogen 

Test for nonspecific inhibitors 

aPTT, activated partial thromboplastin time; INR, international nor- 
malized ratio; PT, prothrombin time. 




> 



42 



VASCULAR SURGERY 



In normal hemostasis, the blood vessel con- 
stricts in response to injury to reduce bleeding. 
Circulating platelets adhere to sub endothelial 
collagen that is exposed by injury, promoted by 
release of tissue factor (TF) from the damaged 
vessel wall. Platelets bind von Willebrand factor 
(vWF) at the glycoprotein lb receptor, stabiliz- 
ing adhesion. Fibrinogen binds to platelet gly- 
coprotein Ilb/IIIa receptors forming bridges 
between adjacent platelets and causing aggre- 
gation. Activated platelets also release potent 
aggregating agents to recruit more platelets. 

Coagulation is initiated through the extrinsic 
pathway. Exposed endothelium releases TF, 
which complexes with and activates factor VII. 
Factor Vila then activates both the common 
pathway and the intrinsic pathway. The intrin- 
sic pathway requires factor VIII and factor IX to 
proceed to the common pathway. Von Wille- 
brand factor also forms a noncovalent bond 
with factor VIII and is essential for its survival 
in the circulation. Von Willebrand factor also 
potentiates factor VIII activity in clot formation 
and protects it from proteolysis. In the final step 
of the coagulation pathway, thrombin cleaves 



fibrinogen to generate fibrin monomers, which 
then polymerize and link to one another to form 
a chemically stable clot. Thrombin also feeds 
back to activate cofactors VIII and V, thereby 
amplifying the coagulation mechanism. 
Together platelet aggregates and fibrin form the 
clot that achieves hemostasis. The coagulation 
cascade is illustrated in Figure 5.1. Any disrup- 
tion of these pathways may lead to increased 
bleeding. 

Features in the history and physical examina- 
tion that may help to differentiate among factor 
deficiencies, platelet disorders, and endothe- 
lial (blood vessel) dysfunction are listed in 
Table 5.5. 

Inherited Disorders 
of Coagulation 

Factor Deficiencies 

Hemophilia A and B represent 80% of inherited 
bleeding diatheses. These are both sex-linked 
recessive deficiencies affecting mostly males. 



Intrinsic 
Pathway 



XII 



XI I, i 



▼ *• 



Villa 



Vila 



Common 

Pathway 



T *' 



Xa 



I In 



-► Va 



Prolbrombin (II) 



Thrombin (I la) 



Extrinsic 

Pathway 



Tissue Factor 



Fibrinogen 



Fibrin 



■ VII 



Figure 5.1. The coagulation cascade. 
Under physiological conditions, tissue 
factor is not exposed to blood. However, 
after injury, tissue factor is exposed to 
blood and activates the extrinsic pathway 
by acting in concert with activated VII and 
phospholipids to convert factor IX to IXa 
and factor X to Xa. The "intrinsic pathway" 
includes activation of factor XII to Xlla, 
which activates factor XI to Xla, IX to IXa, 
and X to Xa. Factor Xa is the active cat- 
alytic ingredient of the prothrombinase 
complex, which includes factor Va and 
phospholipase and converts prothrombin 
to thrombin. Thrombin is a protease that 
cleaves fibrinogen to fibrin. The result- 
ing fibrin monomers polymerize, forming 
a clot. 



43 



BLEEDING AND CLOTTING DISORDERS 




Table 5.5. The relationship of factor disorders, platelet and blood vessel (endothelial) dysfunction to aspects of clinical 
presentation 



Clinical picture 


Factor disorder 


Platelet dysfunction 


Endothelial 
dysfunction 


Onset of bleeding 


Delayed 


Immediate 


Immediate 


Duration of bleeding 


Prolonged 


Short 


Variable 


Precipitant of bleeding 


Often spontaneous 


Trauma 


Variable 


Site 


Joints, muscle, viscera 


Skin, mucous membranes, Gl tract 


Skin, Gl tract 


Family history 


Usually present (unless factor 
inhibitor) 


Absent 


Usually absent 


Drug-related 


Rarely 


Often 


Sometimes 


Sex predominance 


Usually male 


Often female 


Usually 
female 


Response to focal pressure 


No response 


Sometimes responds 


Responds 


Platelet count 


Normal 


Normal, low, or high 


Normal 


Prothrombin time 


Abnormal in cases of factor II, 
VII, IX, and X deficiency 
(and inhibitors) 


Normal 


" 


Partial Thromboplastin 


Abnormal with factor VIII 


Normal 


Normal 


time 


or IX deficiency (and 








inhibitors) 







Gl, gastrointestinal. 



They are caused by factor VIII (classic hemo- 
philia) and factor IX (Christmas disease) 
deficiencies, respectively. Hemophilia A is four 
to six times more common than hemophilia 
B. Together they have a prevalence of about 
20/100,000 in the United States and 9/100,000 to 
15/100,000 in Great Britain. 

The risk for bleeding depends on the degree 
of factor deficiency. At levels less than 1% to 
2% of normal (severe disease), spontaneous 
bleeding is likely to occur and the aPTT is pro- 
longed. Patients may present with spontane- 
ous soft tissue or intramuscular hemorrhage, 
hemarthroses, hematuria, and spontaneous 
retroperitoneal bleeding. Patients with mild 
(>5%) and moderate (1% to 5%) disease usually 
have bleeding only with injury or surgery. 
Patients may have increased bleeding with levels 
above 25%, but these deficiencies are hard to 
detect as the aPTT will be normal. 

Treatment for hemophilia A involves infusion 
of recombinant factor VIII concentrates of 
which several are commercially available, the 
amount depending on the deficiency present. 
This includes a loading dose and then a main- 
tenance dose for the appropriate period 
depending on the location of the bleed if spon- 
taneous or perioperative. For milder hemophilia 
or cases of mild hemorrhage, or if factor VIII 



concentrates are not available, cryoprecipitate 
or fresh frozen plasma (FFP) may be used. 

The treatment for hemophilia B is similar, 
using specific factor IX concentrates or factor IX 
complex transfusion. The factor IX complex 
contains one unit of factor IX per milligram of 
protein with varying amounts of other vitamin 
K-dependent proteins. The concentrates are 
preferred treatment. Again, loading dose and 
maintenance doses are administered according 
to the given situation. 

Another factor deficiency (vWF), which can 
present like hemophilia, occurs in von Wille- 
brand disease (vWD). Overall, this is the most 
common bleeding disorder with an incidence of 
up to 1% in some populations. There are three 
subtypes of VWD, the most severe and uncom- 
mon being type 3. In this form, vWF in plasma 
and platelets is markedly reduced or absent, 
thereby reducing factor VIII activity to 1% to 
10% of normal. Patients can present with spon- 
taneous or severe bleeding. Inheritance is auto- 
somal recessive. Type 1 is the most common 
type, representing around 70% of cases. vWF is 
reduced to the range of 20% to 50% with a con- 
comitant reduction of factor VIII activity. The 
structure of vWF is generally normal in this 
type, and inheritance is autosomal dominant. 
Patients usually present with mild to moderate 




> 



44 



VASCULAR SURGERY 



bleeding diatheses, and the levels of vWF do not 
necessarily correlate with clinical symptoms. 
This may be because some patients with type 1 
VWD have reduced levels of both plasma and 
platelet vWF, whereas some just have reduced 
plasma levels. 

Type 2 VWD is divided into further subtypes 
2A, 2B, 2M, and 2N based on the site of the 
genetic mutation. Type 2A is the most common 
and accounts for around 10% of all vWF 
deficiency cases. Inheritance is autosomal dom- 
inant. The largest multimers of vWF are absent, 
and this leads to a lack of platelet binding. Activ- 
ity of factor VIII, however, can be near normal 
in this subgroup. Clinical presentations in this 
group are therefore variable. In type 2B, patients 
lack multimers in the plasma but have near- 
normal levels in platelets. However, these 
patients are usually thrombocytopenic, which 
can be made worse with exercise, stress, preg- 
nancy, or advanced age. Levels of factor VIII are 
low to normal. Presentation is again variable 
and inheritance is autosomal dominant. Type 
2M is very rare and has abnormal multimers as 
its cause of abnormal coagulation. This has not 
been well defined in terms of inheritance. Type 
2N is inherited as an autosomal-recessive con- 
dition and sometimes mimics mild hemophilia. 
These patients have normal vWF multimers and 
normal vWF activity but reduced factor VIII 
activity due to poor binding between vWF and 
factor VIII. This variant should be considered 
in the differential diagnosis of factor VIII 
deficiency, especially if the patient is female 
and other aspects of the pedigree support 
autosomal-recessive inheritance rather than 
sex-linked. Von Willebrand disease may be 
treated with desmopressin acetate [deamino- 
8-D-arginine vasopressin (DDAVP)], which 
probably increases vWF, tissue plasminogen 
activator (tPA), and factor VIII secretion from 
stored sources. Some of the subtypes may also 
require factor replacement. More recent treat- 
ments include recombinant factor VIII/vWF 
concentrates, which reduce risks of transmis- 
sion of infection. 

Other factor deficiencies are quite rare. Factor 
V deficiency is inherited as an autosomal- 
recessive trait. It has a wide range of clinical 
manifestations but seems to have less bleed- 
ing associated with it than hemophilia A. Severe 
factor V deficiency (levels <1% of normal) pres- 



ents with abnormal bruising, soft tissue hemor- 
rhage, and epistaxis. Bleeding from the umbili- 
cal stump at the time of birth is common. 
Women commonly have abnormally heavy 
menstrual bleeding and postpartum bleeding. 
Most cases present in adulthood. Fresh frozen 
plasma is the mainstay of treatment. 

Factor VII deficiency is inherited as an auto- 
somal-recessive disorder and affects males and 
females equally with an incidence of about 1 in 
500,000. Clinical manifestations can be similar 
to those of hemophilia with severe deficiencies 
(<1% level) as the factor Va/tissue factor com- 
plex is the key initiator of coagulation in vivo. 
However, about half of patients are asymp- 
tomatic, and levels of factor V do not correlate 
well with clinical manifestations. Interestingly, 
diminished or defective function of tissue 
factor has not been documented as a cause of 
decreased factor VII activity. The most common 
presentations include easy bruising, soft tissue 
hemorrhage, and menorrhagia in women. 
Patients with levels less than 1% of normal may 
present like hemophiliacs with pathology 
including intracranial bleeds and hemarthroses. 
Current treatment in the United States is with 
FFP, although factor VII concentrates and 
recombinant factor VII are available in Europe. 

Factor X deficiency is transmitted as an auto- 
somal-recessive trait and has an estimated inci- 
dence of 1 in 500,000. Usually those with greater 
than 15% of normal levels do not have severe 
bleeding, although bleeding with major surgery 
or trauma may occur. However, with more 
severe deficiencies, severe bleeding episodes 
similar to those seen in hemophilia may occur, 
including hemarthroses, retroperitoneal 
hematomas, hematuria, pseudotumors, and 
menorrhagia. Another cause of factor X 
deficiency is amyloidosis. In this situation, 
transfusion of factor X is not helpful because of 
its absorption by the extracellular amyloid. 
Improvement does not occur unless the amyloi- 
dosis resolves, although splenectomy may help 
by debulking splenic amyloid. Treatment for 
standard factor X deficiency is with FFP or pro- 
thrombin complex concentrates. Pure factor X 
concentrates are not available for commercial 
use yet. Concentrations of 10% to 15% give 
adequate hemostasis. Overtransfusion can lead 
to thromboembolic events and disseminated 
intravascular coagulation (DIC). 



45 



BLEEDING AND CLOTTING DISORDERS 




Factor XI deficiency occurs mostly in 
Ashkenazi Jews with a gene frequency in this 
population of 4.3%. Bleeding usually occurs 
with levels <20% of normal and usually only 
after major trauma or surgery. Rarely, there is 
spontaneous bleeding as seen with hemophilia, 
although soft tissue hemorrhage, epistaxis, and 
bleeding after dental extraction and with major 
surgical procedures may occur. Menorrhagia 
may occur in females. Bleeding risk is signifi- 
cantly increased in patients taking aspirin. 
Treatment when necessary is with FFP and with 
cryoprecipitate-poor plasma. Factor XI concen- 
trates are available. 

Fibrinogen Abnormalities 

The dysfibrinogenemia are mostly inheritable 
abnormalities of fibrinogen structure and func- 
tion. Clinically, many patients are asymptomatic 
but some present with either bleeding or a 
thromboembolic event or both. There are mul- 
tiple different abnormalities too diverse to dis- 
cuss in detail in this chapter because they have 
variable inheritance, affect different portions of 
the molecule, and have different manifestations. 
Suffice it to say they should be considered in 
patients with a history of bleeding and abnor- 
mal coagulation testing. Inheritable afibrino- 
genemias and hypofibrinogenemias also exist. 
Patients present with bleeding episodes in the 
afibrinogenemias and with hypofibrinogene- 
mias with levels above 50mg/dL. Afibrinogene- 
mias are inherited as an autosomal-recessive 
trait but hypofibrinogenemias are less pre- 
dictable. Bleeding complications when they 
occur can be severe, with a high incidence of 
bleeding in the neonatal period. This may result 
in death in about one third of patients with 
severe hypo- and afibrinogenemias. 

Platelet Disorders 

Bernard-Soulier syndrome (glycoprotein Ib-IX 
deficiency) is a disorder of platelet adhesion and 
is a relatively rare cause of bleeding. It is char- 
acterized by a prolonged bleeding time, large 
platelets, and thrombocytopenia due to an 
inability to adhere to vWF in the subendothelial 
matrix. It presents in infancy or childhood with 
epistaxis, ecchymosis, and bleeding gums. Treat- 
ment is with platelet transfusion. Hormonal 



control may be helpful with menorrhagia, and 
DDAVP may be useful for prophylaxis prior to 
procedures. 

Deficiencies of platelet collagen receptors 
also exist but do not cause significant bleeding, 
although they may prolong bleeding time. 

Disorders of platelet aggregation include a 
deficiency of Ilb/IIIa, known as Glanzmann's 
thrombasthenia, characterized by a prolonged 
bleeding time and abnormal clot retraction. It is 
inherited as an autosomal-recessive trait in 
clusters of disease. It presents with mucocuta- 
neous bleeding in the neonatal period or in 
infancy and occasionally as bleeding following 
circumcision. Epistaxis and purpura are the 
most common presentation. Severe bleeding 
with menses may be encountered. If it is unrec- 
ognized, significant bleeding with surgery or 
trauma will occur if the patient is not transfused 
with normal platelets. Platelet counts and 
smears are normal, but bleeding times are very 
prolonged. Platelet aggregation is absent in 
normal testing except with epinephrine, where 
it is weak. Platelet secretion is normal with 
strong agonists like thrombin but is absent with 
weak stimulators like epinephrine and adeno- 
sine diphosphate (ADP). Clot retraction is 
either absent or reduced. Treatment is under- 
taken with platelet transfusion if bleeding is 
present. Leukocyte-depleted platelets may 
reduce risks with future transfusions. Hormonal 
therapy is useful with menorrhagia. Regular 
dental care is important to reduce the risk of 
gingival bleeding but Amicar (e-aminocaproic 
acid) may be valuable in helping to control 
bleeding after dental extractions. 

Disorders of platelet secretion are related to 
deficiencies in one or more of the four types 
of platelet granules or to abnormalities in the 
secretory mechanism. Platelets have four types 
of granules: 

1. Dense or 8 granules containing ADP, 
adenosine triphosphate (ATP), calcium, 
serotonin, and pyrophosphates 

2. a-granules containing a variety of pro- 
teins, some of which are obtained from 
the plasma and others synthesized by 
megakaryocytes; these include fibrinogen, 
vWF, albumin, factor V, immunoglobulin 
G (IgG), fibronectin, and proteinase 
inhibitors from the plasma and platelet 




> 



46 



VASCULAR SURGERY 



factor-4, (3-thromboglobulin, platelet- 
derived growth factor, and throm- 
bospondin from megakaryocytes 

3. Lysosomes containing acid hydrolase 

4. Microperoxisomes containing peroxidase 
activity 

Secretory dysfunction usually results in mild 
to moderate bleeding manifested by easy bruis- 
ing, menorrhagia, and excessive postoperative 
or peripartum bleeding. Testing reveals a pro- 
longed bleeding time, a decreased second wave 
of aggregation with ADP and epinephrine 
stimulation, and decreased aggregation with 
collagen. Secretory dysfunction should be dif- 
ferentiated from acquired disorders with acetyl- 
salicylic acid (ASA) use, uremia, and multiple 
myeloma, and from VWD. 

Gray platelet syndrome is an a-granule 
deficiency. Platelets occur without these gran- 
ules but with vacuoles and small a-granule pre- 
cursors containing material that stain positive 
for vWF and fibrinogen. Additionally, the vac- 
uoles and these precursors contain P-selectin 
and Gllb-IIIa. These factors indicate the pres- 
ence of a-granules, but the normal proteins they 
contain cannot be packaged. Other granules 
are present in normal quantity. Patients have 
a history of mild to moderate mucocutaneous 
bleeding. They have prolonged bleeding times 
with moderate thrombocytopenia (60,000 to 
100,000), reticular fibrosis of the bone marrow, 
and large platelets that appear gray on Wright- 
stained blood smears-hence the name. Platelet 
aggregation studies are variable. Treatment 
requires transfusion of normal platelets and at 
least one patient responded to DDAVP. The 
Quebec platelet disorder is extremely rare and 
again involves a-granules that appear grossly 
normal but are deficient in many of the proteins 
normally seen including factor V, fibrinogen, 
vWF, and fibronectin. Patients present similarly 
to the gray platelet syndrome and are treated the 
same way. 

Dense granule deficiency or 8-storage pool 
disease is a heterogeneous group of disorders, 
which can be divided into deficiency states asso- 
ciated with albinism and those in otherwise 
normal patients. With albinism, the disease is 
related to a qualitative deficiency in these gran- 
ules. In nonalbinos, the number of granules 
is near normal. In some of these nonalbino 
patients it is associated with a variable 



deficiency of a-granules. The content of lysoso- 
mal hydrolase is normal, but thrombin-induced 
acid hydrolase secretion is impaired, which can 
only be corrected with ADP. Patients present 
with mild to moderate bleeding. Platelet counts 
are usually normal, but bleeding times are 
prolonged. The quantity of thromboxane B 2 , 
a metabolite of thromboxane A^ is reduced. 
Secondary aggregation induced by ADP and 
epinephrine is reduced. Collagen-induced 
aggregation is abnormal at low concentrations 
of collagen but is normal with high concentra- 
tions. Therapy requires transfusions of normal 
platelets if there is massive bleeding. Otherwise, 
DDAVP is the initial treatment and cryoprecip- 
itate can also be used. In one study, pred- 
nisolone seemed to reduce bleeding in patients 
with inherited platelet disorders but not in 
patients with thrombasthenia or ASA use. 

Platelet function in patients with inherited 
disorders of platelet secretion resembles that of 
patients receiving platelet function inhibitors 
like ASA. These disorders are a heterogeneous 
collection of abnormalities of secretion- 
response adhesion. In families where these dis- 
orders have been noted, the pattern appears to 
be autosomal dominant. A prolonged bleeding 
time and marked impairment of aggregation 
and secretion in response to ADP, epinephrine, 
and low concentrations of collagen occur. 
Stronger agonists like high levels of collagen, 
however, may induce a near-normal or normal 
response. Treatment is the same as that for 
patients with platelet storage disorders. 

Vascular Defects 

Inheritable conditions that lead to defects in the 
vascular bed include Marfan's syndrome and 
Osier- Weber-Rendu disease. These may increase 
bleeding through mechanisms that are not 
entirely clear. 

Acquired Disorders 
of Coagulation 

This group of disorders includes factor 
inhibitors as well as acquired diseases that affect 
platelet and endothelial function. Factor 
inhibitory proteins can be classified as neutral- 
izing, nonneutralizing, or altering. They are 



47 



BLEEDING AND CLOTTING DISORDERS 




usually autoantibodies. The most common 
factor inhibitor is factor VIII, often referred 
to as acquired hemophilia, but inhibitors have 
been found to thrombin and prothrombin, 
fibrinogen, thrombin and prothrombin, fibrino- 
gen, vWF, and factors V, VII, IX, X, and XI as 
well (Fig. 5.1). These are seen in response 
to inflammatory diseases such as rheumatoid 
arthritis and other autoimmune diseases like 
systemic lupus erythematosus (SLE) and 
Sjogren's syndrome and even inflammatory 
bowel syndrome. They also appear during preg- 
nancy and the puerperium, as well as with 
various tumors. They may be associated with a 
variety of medications including aminoglyco- 
sides, penicillins, valorous acid, etc. They are 
also seen with cirrhosis and major surgery. 

Interestingly, factors V and X and thrombin 
inhibitors have been seen following the use of 
topical thrombin and fibrin glue, usually after 
multiple exposures. Not only can this lead to 
perioperative bleeding, but also inhibitors of 
factor X can interfere with monitoring of low- 
molecular-weight heparin (LMWH) anticoagu- 
lation with the antifactor Xa assay. 

Factor inhibitors may lead to increased bleed- 
ing and, rarely, spontaneous bleeding. Most 
conditions are initially detected as a prolonga- 
tion of one or more of the coagulation screen- 
ing tests — PT/INR/aPTT — or thrombin time. 
To differentiate between the different types of 
inhibitors, a clotting study should be done on a 
1 : 1 mixture of the patient's plasma with normal 
plasma. A lack of correction of clotting time 
indicates a neutralizing antibody. Vigilance in 
testing is essential because at body temperature 
some studies initially correct and then return to 
a prolonged state after an hour. This is particu- 
larly true of factor V and VIII inhibitors. 

If the mixing study corrects, this suggests the 
presence of a nonneutralizing antibody that 
may facilitate clearance of the clotting factor 
from circulation. The antibody should be 
isolated to differentiate its presence from a 
deficiency of the factor. 

Treatment of bleeding includes infusion of 
factors to levels that overwhelm the antibodies 
and return the coagulation profile to normal. 
Long-term treatment includes immunosup- 
pression with steroids or other agents and 
plasmapheresis with immuno absorption with 
Ig-Therasorb. The latter can be used in con- 
junction with transfusion. 



Hypercoagulable States 

Like bleeding disorders, these states can be 
categorized as inherited or acquired (Table 5.6). 
Hyperhomocystinemia, however, can be either 
inherited or acquired. Indications for throm- 
bophilia screening have already been outlined 
and the components of such a screen are 
referred to in Table 5.3. For most of the hyper- 
coagulable states, treatment involves the use of 
LMWH or unfractionated heparin with conver- 
sion to oral warfarin. Often, if the patient has 
already had a thrombotic episode, anticoagula- 
tion is undertaken for life. 



Table 5.6. Differential diagnosis for hypercoagulable states 

Inherited (primary) 
Protein C and S deficiencies 
Antithrombin III deficiency 
Factor V Leiden mutation leading to activated 

protein C resistance 
Prothrombin 20210A gene mutation 
Heparin cofactor II deficiency and other heparin 

binding proteins 
Cystathionine synthase deficiency 

(hyperhomocystinemia) 
Dysfibrinogenemia 
Dys- and hypoplasminogenemia 

Acquired (secondary) 
Cancer 
Pregnancy 
Oral contraceptives and other hormone replacement 

therapy (HRT) 
Myeloproliferative disorders 
Hyperlipidemia 
Diabetes mellitus 
Vasculitis 
Antiphospholipid syndrome (lupus anticoagulant, 

anticardiolipin antibodies) 
Postoperative states/trauma 
Immobilization 
Nephrotic syndrome 
Congestive heart failure 
Increased levels of factor VII and fibrinogen 
Obesity 

Heparin thrombocytopenia 
Anticancer drugs (bleomycin, vinca alkaloids, 

mitomycin, etc.) 
Paroxysmal nocturnal hemoglobinuria 
Age 




Undetermined 
Elevated factor XI levels and \ 



I levels 




> 



48 



VASCULAR SURGERY 



Inherited 

Inherited hypercoagulable states account for 5% 
to 15% of patients with venous thromboem- 
bolism (VTE). The most common appear to be 
the factor V Leiden mutation followed by the 
prothrombin G20210A mutation of the pro- 
thrombin gene among the white population, but 
these are rare in Asians and Africans. Table 5.7 
shows the overall incidence of the various inher- 
ited hypercoagulable states. These are estimates 
from several studies and vary with ethnic back- 
ground and location. 

Factor V Leiden, also known as activated pro- 
tein C resistance (APCR), results from a point 
mutation in the factor V gene, leading to a loss 
of protein C cleavage sites (Ouriel et al., 1996). 
The consequence of this is impaired activation 
of protein C. The most common presentations 
are VTE and fetal loss. It usually does not result 
in arterial thrombosis unless other risk factors 
are also present, for example smoking. 
Homozygotes have an 80-fold increased risk of 
DVT, and heterozygotes have much less. Many 
patients with factor V Leiden remain asympto- 
matic, and about 60% of those who present with 
thrombosis have another risk factor such as use 
of oral contraceptives (OCs) or hormone 
replacement therapy (HRT). The overall risk of 
VTE is 3% to 7%. 

The prothrombin 202 10A gene mutation 
appears in 2.3% of healthy control patients. The 
incidence is twice as high in southern Euro- 
peans compared to northern Europeans and is 
rare in Asians and Africans. The mutation 
increases prothrombin levels and activity. The 
relative risk of clotting is two to three times that 
of normal individuals. 



Table 5.7. Incidence of inherited disorders 



Disorder 


Incidence 


Factor V Leiden (activated protein 


25% 


C resistance) 




Sticky platelet syndrome 


14% 


Protein C deficiency 


10% 


Protein S deficiency 


10% 


Prothrombin G20210A 


5-10% 


Increased homocysteine 


5-10% 


Dysfibrinogenemia 


1.5% 


Antithrombin III 


<1% (1/250-500) 


Dys- or hypoplasminogenemia 


1-3% 



Protein C and S deficiencies have relatively 
the same incidence. They both result from 
numerous different mutations. A heterozygote 
pattern is much more frequent as homozygotes 
present soon after birth with purpura fulminans 
or massive venous thrombosis. Protein C has 
type I and type II deficiencies. Type I deficien- 
cies are those mutations causing a decrease in 
levels and in activity; most of these are missense 
mutations. Type II deficiencies are those in 
which levels are normal but activity is affected; 
most of these are point mutations. Patients with 
protein C deficiency tend to present with VTE 
and fetal loss and rarely with arterial thrombo- 
sis. Protein S deficiency is harder to define. 
Many studies have shown the coexistence of 
protein S deficiency with APCR in as high as 
40% of the patient population studied. This 
makes it hard to determine which is more 
important. However, there are more numerous 
reports of arterial thrombosis with protein S 
deficiency, including stokes, compared to 
protein C deficiency. Many conditions may 
result in lowered levels of these proteins, partic- 
ularly protein S (for instance in liver disease, 
nephrotic syndrome, pregnancy, sepsis, etc.) 
and some medications like HRT may have the 
same effect. Therefore, measurement of levels of 
protein S may need to be repeated to ensure 
accuracy as well as to allow checking for specific 
genetic defects. Overall, as many as 50% of 
heterozygotes for protein C and S deficiencies 
develop VTE up to the age of 50 years. 

Antithrombin III (AT III) deficiency occurs as 
an autosomal-dominant disorder and in 1/5000 
healthy blood donors. It may also have type I 
and type II deficiencies. The type I deficiencies 
are caused by both gene segment mutations as 
well as point mutations, whereas the type II 
deficiencies are caused mostly by point muta- 
tions. Most patients are heterozygotes, 
as homozygous deficiency is probably incom- 
patible with life unless it is a type II deficiency 
of the heparin-binding site. All types are at 
increased risk of VTE, with as many as 80% of 
heterozygotes by age 50 having an episode of 
VTE. Like protein C and S, AT III deficiency can 
be acquired in association with the medical con- 
ditions described above. 

Hyperhomocystinemia and homocystinuria 
have both been described as associated with 
VTE and arterial thrombotic events (Nehler 
et al., 1997). Homocysteine is an intermediate of 



49 



BLEEDING AND CLOTTING DISORDERS 




methionine metabolism. Elevated levels arise 
from both genetic defects affecting the transsul- 
furation or remethylation pathways as well as 
with folate, B 6 and B 12 deficiency, renal failure, 
hypothyroidism, increased age, and smoking. 
Homozygotes for cystathionine P-synthase 
deficiency and methylene-tetrahydrofolate re- 
ductase may have severe vascular disease and 
appear even in childhood. As many as 60% have 
thromboembolic events before age 40 and 50% 
by age 29. Heterozygotes have a high incidence 
of premature arterial occlusive disease, which 
may represent as much as 1/70 of the normal 
population. However, the most common cause is 
dietary folate and B 6 and B 12 deficiencies, which 
account for around two thirds of the cases 
of hyperhomocystinemia. Measuring fasting 
homocysteine plasma levels establishes the 
diagnosis. The mechanism of thrombosis is 
thought to involve several mechanisms includ- 
ing induction of endothelial cell tissue factor 
activity, inhibition of thrombomodulin, 
decreased AT III activity, decreased protein C 
activation, increased factor V activity, and 
increased affinity of lipoprotein (a) and fibrin. 
Dietary supplements with B 6 , B 12 , and folate help 
lower the homocysteine levels, but unfortu- 
nately this may not reduce the risk of thrombo- 
sis and therefore anticoagulation may also be 
indicated. 

The dysfibrinogenemias can cause thrombo- 
sis as well as bleeding in around 20% of patients. 
The conversion of fibrinogen to fibrin consists 
of three main steps: release of fibrinopeptides A 
and B from the alpha and beta chains to form 
fibrin monomers, followed by polymerization of 
these monomers to a visible fibrin gel, which is 
then stabilized by activated factor XIII. Fibrin 
may then be broken down via fibrinolysis path- 
ways. Inherited dysfibrinogenemias result from 
mutations that alter one or more of these steps. 
They are usually autosomal dominant. One may 
be suspicious if there is a prolongation of the 
PT and PTT. Thrombosis can occur secondary 
to either an abnormal fibrinogen with reduced 
binding to thrombin and increased thrombin 
levels, or decreased fibrinolysis. It is relatively 
rare for this to be the sole cause of VTE (around 
0.8% of VTE) and is frequently associated with 
precipitating risk factors such as HRT, pro- 
longed bed rest, etc. Treatment remains anti- 
coagulation, but it is preferable initially to use 
LMWH, as unfractionated heparin activity can 



be difficult to monitor with a baseline elevated 
aPTT. Acquired dysfibrinogenemia is seen with 
liver disease, multiple myeloma, Waldenstrom's 
macroglobulinemia, and autoimmune diseases; 
the end result of all of these conditions is altered 
polymerization or delayed fibrinopeptide 
release. 

Dys- or hypoplasminogenemias occur even 
more rarely. These are usually autosomal dom- 
inant. Patients usually present in their late teens, 
most commonly with VTE. Routine tests are 
normal. There are also rare congenital deficien- 
cies of tPA and congenital increases of plas- 
minogen activator inhibitor (PAI). These 
disorders are more commonly acquired with 
diabetes, inflammatory bowel disease, and coro- 
nary atherosclerosis. 

Factor XII is involved with plasmin genera- 
tion. There is an autosomal-dominant genetic 
deficiency, which is quite rare but results in both 
VTE and arterial thrombosis. About 8% of 
deficient patients have thrombotic episodes. An 
elevated PTT is present, but it corrects with the 
addition of normal plasma. Treatment is with 
LMWH and then warfarin. Standard unfrac- 
tionated heparin again is difficult to monitor 
with baseline elevated PTT. 

Heparin factor II and other heparin-binding 
molecules are found to have deficiencies; how- 
ever, the association with thrombotic events is 
weak. The same is true for thrombomodulin 
defects. 

The sticky platelet syndrome is a rare 
autosomal-dominant disorder that results in 
platelets that aggregate more readily with epi- 
nephrine or ADP. Venous and arterial thrombo- 
sis occur, and retinal vascular thrombosis 
appears to be associated with this entity. Treat- 
ment is initially with low-dose aspirin. If aggre- 
gation does not normalize, then this dose can be 
increased to 325 mg daily. Clopidogrel (Plavix) 
provides an alternative potential therapy. 
Another platelet defect is the Wein-Penzing 
deficit. This is a deficiency of the lipoxygenase 
metabolic pathway resulting in an increase in 
the cyclooxygenase pathway and therefore ele- 
vated thromboxane levels. Platelets are thus in a 
state of increased activation. 

Other abnormalities that may be genetically 
based include elevated factor XI and factor XIII 
levels. Both these groups seem to have increased 
risk of VTE, and the level of increased risk 
appears to be proportional to the increase in the 




> 



50 



VASCULAR SURGERY 



levels of these factors. Hormone replacement 
therapy and pregnancy are also implicated in 
raised levels of these factors. Patients on HRT or 
OC and with elevated factor VIII levels have a 
10-fold increased risk of VTE compared to those 
without these risk factors. 

Acquired 

The most common cause in this group of con- 
ditions is the antiphospholipid antibody (APA) 
syndrome (Fligelstone et al., 1995). These anti- 
bodies bind to plasma proteins that have a high 
affinity for phospholipid surfaces. The most 
common of these proteins are the lupus antico- 
agulant (LA), anticardiolipin (ACL) antibodies, 
and anti-(3 2 -glycoprotein-l antibodies (B 2 G). 
These are usually acquired. Lupus anticoagulant 
can be suspected if there is an elevated PTT, and 
ACL and B 2 G are detected only by immunoas- 
says. These conditions can be primary, that is, 
not associated with other autoimmune condi- 
tions. They can present with VTE or arterial 
thrombosis as well as fetal demise. Antiphos- 
pholipid antibodies may be associated with 
infections, cancer, and even certain drugs and 
hemodialysis, but these are usually IgM as 
opposed to IgG antibodies and are present in 
low levels. They do not seem to be associated 
with thrombotic events. 

The overall incidence is 1% to 5% of the 
normal population. The incidence increases 
with age and coexisting chronic disease. The 
incidence in patients with SLE is 12% to 30% for 
ACL and 15% to 34% for LA. The risk of throm- 
bosis in patients found to have these autoanti- 
bodies is unclear but seems to be increased in 
those with a history of previous thrombosis, 
with LA and with increased IgG ACL — each of 
which increases the risk of thrombosis fivefold. 
The persistent presence of APA also increases 
the risk of thrombosis. These patients have a 
high proportion of pregnancy losses as well as 
an increased incidence of premature births. 
These patients are best treated with ASA plus 
heparin to achieve live births. 

Interestingly, not all arterial episodes of 
ischemia or infarction are due to primary 
thrombosis. Up to 63% of patients with APA 
syndrome have coexisting valve abnormalities 
on echocardiography and 4% have vegetations 
of the mitral or aortic valve. These patients may 
also be thrombocytopenic (as many as 40-50%) 



and may have hemolytic anemia (14-23%) and 
livedo reticularis (11-22%). Renal involvement 
may occur, and when it does it usually results in 
hypertension. 

Catastrophic APA syndrome is acute, and 
multiple simultaneous vascular thrombotic 
events can occur throughout the body. Small 
vessels of multiple organs are often affected. The 
syndrome may result in death. In 78% of these 
patients the kidneys are involved, followed by 
the lungs in 66%, the central nervous system in 
56%, the heart in 50%, and the skin in 50%. Dis- 
seminated intravascular coagulation is a com- 
ponent of this event in 25% of patients. The 
mortality rate is 50%, usually due to multisys- 
tem organ failure. The optimal treatment is 
not well defined but includes, in various com- 
binations of anticoagulation, steroids and either 
plasmapheresis or intravenous immune globu- 
lin. The condition can be precipitated by 
surgery, infection, withdrawal of anticoagula- 
tion therapy, and drugs, including oral 
contraceptives. 

Heparin-induced thrombocytopenia (HIT) 
also warrants some discussion. The entity was 
first described by Towne in 1979 as the white 
clot syndrome and occurs in 1% to 30% of 
patients on heparin. Heparin-induced thrombo- 
cytopenia is diagnosed by one or more HIT- 
associated clinical events, such as thrombosis of 
a graft, and the presence of heparin antibodies. 
Various tests (Table 5.8) are now available to 
help in making the diagnosis (Warkentin and 
Greinacher, 2004). The problem is that the onset 
of HIT is often delayed or seen after multiple 
exposures to heparin, where the first few expo- 
sures resulted in minimal or no decrease in 
platelets. Additionally, the delayed onset may 
be seen only after the offending heparin has 
been removed. The initial exposure can be 
as inconsequential as heparin lock flushes to 
maintain the patency of intravenous lines or 
standard subcutaneous heparin prophylaxis. 
Treatment includes stopping the heparin. 
Coumadin alone is not a good treatment due to 
the hypercoagulable period before anticoagula- 
tion is achieved, leading to potential skin necro- 
sis. Additional therapy is switching to hirudin 
(Refludan), argatroban (Novastartan), or dana- 
parnol (Orgaran). Additionally, some of the 
Gllb-IIIa inhibitors have been successfully 
employed in this situation. Newer agents are 
under investigation including a recently Food 



51 



BLEEDING AND CLOTTING DISORDERS 




Table 5.8. Testing for heparin-induced thrombocytopenia 



Ig, immunoglobulin; PF4, platelet factor 4. 





Sensitivity 

(%) 


Specificity 

(%) 


Positive 
predictive 

(%) 


value 


Negative 
predictive value 


PF4 + heparin coated plate: anti IgG, IgA, 

and IgM 
Serotonin release 
Platelet aggregation 


97 

88 
91 


86 

100 
77 


93 

100 
89 




81 



and Drug Administration (FDA)-approved oral 
anti-factor Xa agent. 

Most malignancies can be associated with a 
higher incidence of thromboembolic events, in 
particular, adenocarcinoma and myeloprolifer- 
ative disorders. 



Conclusion 

Bleeding and clotting disorders are encountered 
sufficiently frequently in vascular surgical prac- 
tice to encourage an awareness of the range of 
disorders and of the different available thera- 
peutic modalities. A high index of suspicion, a 
comprehensive medical history, and judicious 
employment of investigations help guide the 
experienced clinician to a correct diagnosis. The 
bleeding disorders are varied, and each requires 
specific management. The majority of throm- 
bophilic tendencies may simply be treated with 



appropriate anticoagulation with the exception 
of catastrophic amyotrophic lateral sclerosis 
(ALS). The cumulative risks of different inher- 
ited and acquired thrombophilic factors have 
appreciable significance. Individual personal- 
ized risk profiles for thrombotic events may 
have a role to play for patients in the future. An 
awareness of the broad extent of these condi- 
tions helps vascular surgeons provide optimal 
management for their patients. 

References 

Donaldson MC, Weinberg DS, Belkin M, Whittemore AD, 

Mannick JA. (1990) J Vase Surg 11:825-31. 
Fligelstone LJ, Cachia PG, Ralis H, et al. (1995) Eur J Vase 

Endovasc Surg 9:277-83. 
Nehler MR, Taylor LM Jr, Porter JM. (1997) Cardiovasc Surg 

5:559-67. 
Ouriel K, Green RM, DeWeese JA, Cimino C. (1996) J Vase 

Surg 23:46-51, discussion 51-2. 
Warkentin TE, Greinacher A. (2004) Chest 126:3 11S-337S. 



6 



Medical Management of Peripheral 
Arterial Disease 

Jill J.R Belch and Andrew H. Muir 




Atherosclerosis 

Peripheral arterial disease (PAD) is a marker 
for multisystem vascular disease. Workers in 
this field recognized this over 150 years ago, 
noting the similarity between claudication 
symptoms and angina. Since then, published 
work has supported this view, but the com- 
monality of the link between PAD and coronary 
artery disease (CAD) has only now been fully 
recognized. Myocardial infarction (MI) and 
stroke are the biggest risks to life and health 
for the PAD patient rather than amputation 
or critical limb ischemia (CLI). The natural 
history of intermittent claudication (IC), the 
usual symptom of lower limb atherosclerosis, is 
often benign. Most patients improve or their 
disease remains stable. Less than 5% of patients 
require amputation. In contrast, however, the 
death rate in these patients is three to four 
times higher than in patients of similar age 
without IC (Fig. 6.1). This mortality occurs due 
to atherosclerosis at other sites within the body. 
Only one quarter of the mortality of these pa- 
tients is from nonvascular events. One half die 
from CAD, 15% as a result of stroke, and 10% 
from vascular pathology within the abdomen 
such as ruptured aortic aneurysm. The associa- 
tion between PAD and generalized vascular 
mortality is so strong that even in asymptomatic 
PAD [detected by a decrease in the ante- 
brachial index (ABI)] the patient's relative risk 
of a cardiac or cerebrovascular event is very 
much higher. 



Thus we now recognize that atherosclerosis is 
a systemic disorder affecting the entire vascular 
tree. Extracranial carotid disease can lead to 
stroke, CAD to myocardial ischemia/infarction, 
renovascular disease to hypertension, and aor- 
toiliac and infrainguinal arterial disease to IC or 
limb-threatening ischemia. The mainstay of the 
medical management of vascular disease is to 
understand that it is a systemic disorder and 
must be managed as such. Thus the medical 
management of PAD is a complex area that 
includes strategies for vascular risk reduction, 
lifestyle advice, and direct pharmacotherapy 
for the vascular disease. Its implementation 
requires a multidisciplinary team, which in- 
cludes not only vascular physicians, vascular 
surgeons, and interventional radiologists but 
also many of the professions allied to medicine 
and, importantly, our primary care colleagues. 
Areas of focus for such a vascular team are out- 
lined in Table 6.1. 



Vascular Risk Factor 
Modification in Peripheral 
Arterial Disease 

The medical management of vascular disease 
must focus on modification of the following 
specific risk factors that promote the progres- 
sion of the disease: (1) platelet aggregation, (2) 
smoking, (3) obesity, (4) diabetes, (5) dyslipi- 



53 




> 



54 



VASCULAR SURGERY 





100 


t 


80 


-1 
< 


60 


H 




(T 




O 


4U 



20 



COLO 
RECTAL) 



BREAST 



PROSTATE 



PAD 



LUNG 



Figure 6.1 . Mortality of peripheral arterial disease (PAD). The 5- 
year mortality of PAD is relatively equivalent to numerous 
cancers (prostate, breast, colorectal, and lung). 



demia, (6) hypertension, (7) sedentary lifestyle, 
and (8) type A personality or stress. 

The literature suggests that cigarette smok- 
ing, hypertension, dyslipidemia, and diabetes 
mellitus are important factors in the develop- 
ment of PAD, and these will be addressed in turn 
(Table 6.2). A key risk factor for PAD is platelet 
aggregation along with other hemorheological 
factors such as increased plasma fibrinogen and 
decreased fibrinolysis. Currently an area of 
interest that is being explored is that of the con- 
tribution of inflammation to PAD. The high 
white blood count (WBC) contributes vascular 
risk to the patient with PAD (Belch et al, 1999) 
as does increased oxidative stress. Of the above, 
however, platelet activation and release in the 
patient with PAD has been well documented 
and has led to the evidence-based use of 
antiplatelet agents in PAD. 



Platelet Aggregation 



Platelet aggregation is increased in patients with 
PAD, and the role of such aggregates in arterial 
thrombosis has been well documented. Platelet 
release products also contribute to the underly- 
ing pathology. (3-Thromboglobulin is one such 
product that, once released from platelets, con- 
tributes to neutrophil activation. Platelet factor 
4 neutralizes heparin, and platelet-derived 
growth factor stimulates proliferation of vas- 
cular smooth muscle cells. Some of the release 



products are vasoactive, and these include 
thromboxane A 2 and serotonin, both potent 
vasoconstrictors. 



Antiplatelet Agents 

Current interest in antiplatelet agents relates to 
their use as prophylactic treatments against 
arterial events in other beds and atherosclerosis 
disease progression. 



Aspirin in Peripheral Arterial Disease 

Aspirin is the most commonly used antiplatelet 
agent, reflecting the fact that it is currently 
the cheapest agent available and one of the 
best studied agents in clinical trials. Its action 
includes the irreversible inhibition of the cyclo- 



Table 6.1 . Medical strategies for the management of 
peripheral arterial disease 

Address vascular risk 

Antiplatelet therapy 

Cigarette smoking 

Dyslipidemia 

Hypertension 

Diabetes 

Obesity 

Thrombophilia 
Specific therapy for intermittent claudication 

Exercise 

?Drug therapy 
Vasculitides as a cause of symptoms 

Connective tissue disease 

Raynaud's phenomenon 

Vasculitis 

Antiphospholipid syndrome 
Embolism as a cause of symptoms 

Detect and treat arrhythmia and/or cardiac 
thrombus 

Exclude aortic aneurism 
Chronic limb symptoms 

Control edema 

Control infection 

Improve cardiac output 
Acute critical limb ischemia (CLI) 

Thrombolysis 

Anticoagulation 
Deep vein thrombosis (DVT) 

Prophylaxis 

Treatment 



55 



MEDICAL MANAGEMENT OF PERIPHERAL ARTERIAL DISEASE 




Table 6.2. Risk factors for intermittent claudication 
Risk 




Other cardiovascular events, e.g., Ml, stroke 


• Antiplatelet agent 




Aspirin, clopidogrel, dipyridamole 


Smoking 


• Cessation program 




Counseling 




Nicotine replacement, e.g., patch, gum, spray 


Dyslipidemia 


• Lipid-lowering therapy 




Diet + drug 




Target: LDL cholesterol <3.0 mmol/L 




HDL >20% 




Triglycerides <1. 8 mmol/L 


Hypertension 


• Antihypertensive therapy 




Select: ACE inhibitor 




Calcium channel blockade 




Diuretic 




Avoid beta-blockade (unless vasodilatory) 




Target BP <140/85 (in diabetes <140/80) 




(Note: 25% will have renal artery stenosis; watch renal function) 


Diabetes 


• Check for hyperglycemia 




Fasting glucose > 7.8 mmol/L 




If borderline glucose (random or fasting), glucose tolerance test with 




fasting and 2-hour sample 




2-hour sample: <7.8 mmol/L normal 




>7.8-1 1.0 impaired 




>1 1.1 diabetic 


Obesity 


• Check for obesity 




Body mass index >30 




Low-fat, high-fiber diet, healthy eating 




Calorie control 


Thrombophilia 


• Consider referral for thrombophilia screen (inherited or acquired) 




Suspect if: 




Young (<50 years of age) 




Connective tissue disease, e.g., SLE 




Additional history of unexplained venous thrombosis 




Previous failed vascular reconstruction 




Strong family history of thrombosis 



SLE, systemic lupus erythematosus. 



oxygenase enzyme. Platelet aggregation is there- 
fore decreased. 



Primary Prevention of Peripheral 
Arterial Disease 

As yet there is no published evidence to suggest 
that aspirin prevents the primary development 
of PAD in a normal population. Such a study 
would require a huge population base. One 
method of decreasing the numbers required 
to be studied would be to increase the risk of 
the population enrolled in the trial. Two such 



studies are currently under way. In both these 
studies aspirin is being evaluated in subjects 
with decreased ABIs but who are currently 
asymptomatic in terms of PAD. One of the 
studies is a population-based study and the 
other is a study of patients with diabetes melli- 
tus. This latter study, Prevention of Progression 
of Arterial Disease in Diabetes (POPADAD) has 
recruited 1250 patients with decreased ABI, and 
aspirin versus placebo is one of the arms of this 
study, which has a composite end point of vas- 
cular events and mortality. This is a Scotland- 
wide study and results should be available soon. 




> 



56 



VASCULAR SURGERY 



Primary Prevention of Vascular Surgery 

Aspirin has been evaluated in the primary pre- 
vention of requirement for peripheral vascular 
surgery in the U.S. Physicians' Health Study. In 
the aspirin group the risk of undergoing surgery 
for PAD was decreased by 46% (p = .03). Aspirin 
did not, however, affect the likelihood of devel- 
oping claudication de novo during the trial 
period. 

Aspirin in the Prevention of Coronary and 
Cerebrovascular Events in Peripheral 
Arterial Disease 

The Antiplatelet Trialists Collaboration, now 
called the Antithrombotic Trialists Collabora- 
tion, has provided the most convincing data 
supporting the use of aspirin in PAD. In a meta- 
analysis of 174 randomized trials of various 
antiplatelet agents (mainly aspirin), a decrease 
in nonfatal MI, nonfatal stroke, and vascular 
death in patients treated by antiplatelet therapy 
was detected. Subgroup analysis of high-risk 
patients was carried out. This included patients 
with PAD, and the percentage of risk reduction 
versus placebo were as follows: 46% for non- 
fatal stroke, 32% for the risk of vascular disease, 
MI, or stroke, and for nonfatal MI, and 20% for 
death from vascular causes. The most frequently 
used dosages of aspirin were between 75 and 
325 mg per day. Subsequent work has confirmed 
that there is no evidence that the higher doses 
are more effective than the lower ones (i.e., 
>75mg per day), although bleeding risk is dose 
dependent. 



Clopidogrel 

Clopidogrel irreversibly blocks the binding of 
adenosine diphosphate (ADP) and thus its 
activation of platelets. The Clopidogrel versus 
Aspirin in Patients at Risk of Ischemic Events 
(CAPRIE) study evaluated the risk of vascular 
death, MI, and stroke in patients with vascular 
disease receiving either clopidogrel or aspirin. 
Clopidogrel reduced the event rate by 5.32% per 
annum versus 5.83% with aspirin (p = .043). 
These figures reflect a relative rate reduction of 
8.7% in favor of clopidogrel. No major differ- 
ences in safety issues between the two drugs 
was detected. An ad hoc subgroup analysis in 



patients with PAD suggested greater benefit to 
these patients from clopidogrel than in other 
patient populations. An event rate of 3.1 per 
annum compared to 4.86 per annum in the 
aspirin group gave a relative risk reduction of 
23.8% [95% confidence interval (CI) 8.9-36.2] in 
favor of clopidogrel (p = .0028). A reasonable 
strategy, therefore, in the 30% of all patients who 
developed gastrointestinal (GI) side effects from 
aspirin is to combine the aspirin initially with 
the gastric protectant, and if this fails, change to 
clopidogrel. This drug provides an improve- 
ment to our therapeutic resources in terms of 
tolerated antiplatelet drugs. 



Dipyridamole 

Dipyridamole is an antiplatelet agent that is 
thought to work through a number of mecha- 
nisms including increasing the effect of prosta- 
cyclin (PGI 2 ), and inhibiting the cellular uptake 
of adenosine and platelet phosphodiesterase, 
thus enhancing further the effects of PGI 2 . The 
use of dipyridamole itself or in combination 
with aspirin has produced much controversy. 
Three decades ago there was excitement about 
the potential benefits of combining aspirin and 
dipyridamole, but by the 1980s aspirin alone 
became the favorite choice. Evidence that the 
combination treatment is effective has been 
forthcoming in one of the large stroke studies 
(Diener et al., 1996), and certainly the combina- 
tion has been found to be more effective in the 
prevention of peripheral graft failure in one 
study. It is our current practice to use both 
aspirin and dipyridamole in patients with 
stroke or transient ischemic attack (TIA) but to 
use aspirin or clopidogrel alone in patients with 
PAD. In stable PAD we usually reserve dipyri- 
damole for the aspirin-intolerant patient, al- 
though clopidogrel is now our first choice for 
this indication. 



Conclusion 

There is convincing evidence that antiplatelet 
agents such as aspirin and clopidogrel are effec- 
tive in preventing cardiac and stroke events in 
patients with PAD. It is therefore recommended 
that an antiplatelet drug should be prescribed 
for these patients unless there is a clear con- 
traindication to such therapy. 



57 



MEDICAL MANAGEMENT OF PERIPHERAL ARTERIAL DISEASE 




Smoking 

Both the risk of PAD and its progression is 
significantly increased by the smoking of 
tobacco. Patients who smoke have a significantly 
increased risk of atherosclerosis and of another 
tobacco-related peripheral vascular disease, 
thromboangiitis obliterans (Buerger's disease). 
Of all the risk factors discussed here, the use of 
tobacco contributes most to the development of 
PAD. A number of studies have linked tobacco 
smoking and the development of IC. In the 
Framingham study smokers were twice as likely 
to develop IC. The progression to critical limb 
ischemia is more likely in smokers, as is ampu- 
tation. The effect of cigarette smoking on vas- 
cular graft patency is well recognized. In one 
study the 5-year cumulative patency rate for 
grafts was between 80% and 90% for nonsmok- 
ers, with the corresponding patency rates being 
between 30% and 45% for those who smoked 
more than five cigarettes a day. 

Smoking cessation slows the progression 
of peripheral arterial disease to critical limb 
ischemia and lowers the risk of MI and death. 
The nicotine present in tobacco products is 
highly addictive; hence, smoking cessation is 
a very difficult process for patients, and the 
recidivism rate is high. The key feature in the 
approach to smoking cessation is for the physi- 
cian to positively encourage the patient to stop 
smoking. Without a clear message from the 
physician that smoking is an underlying cause 
of atherosclerosis and promotes the progression 
of atherosclerosis, patients will continue to 
smoke. Thus, the first step is to clearly tell 
patients that it is medically indicated that they 
stop smoking. 

Although the majority of smokers would like 
to quit, 90% are physically addicted to nicotine 
and at least 75% have tried to stop smoking 
more than once. Three quarters of those who 
do stop restart within 3 months, and it is clear 
that the issue of nicotine dependence must be 
addressed. During the early stages of quitting, 
smokers experience both behavioral and physi- 
cal withdrawal symptoms. It is crucial to explain 
to the patient that the chemical withdrawal 
symptoms are short lived as they can exert 
strong pressure on the smoker's will to stop. 
This also underlines the fact that we must 
provide support to these patients. After re- 
ceiving only medical advice, only 5% of 



PAD patients stop smoking. Improved cessation 
rates, however, can be achieved through increas- 
ing support through counseling or the provision 
of judicious nicotine replacement therapy. 
There are numerous smoking cessation pro- 
grams and pharmacological aids available for 
patients. Smoking cessation aids such as nico- 
tine patches and nicotine gum work best in the 
setting of a specific smoking cessation program. 
Nicotine chewing gum was the first type of nico- 
tine replacement therapy (NRT) to become 
widely available, with subsequent development 
of transdermal patches, intranasal sprays, and 
inhalers. These latter forms of therapy may 
attenuate some problems with the gum such as 
transfer of dependency. A meta-analysis of 53 
trials of NRT (42 gum, nine patch, one spray, and 
one inhaler) (Silagy et al., 1994) showed that 
NRT increased the odds ratio for abstinence: 
1.61 for gum, 2.07 for patch, 2.92 for nasal spray, 
and 3.05 for inhaled nicotine. Nicotine replace- 
ment therapy can be an effective aid to smok- 
ing cessation. However, it is contraindicated in 
acute MI, unstable angina, and in patients with 
cardiac arrhythmias. Thus, it is important to 
work with patients to engage them in a specific 
program. 

Finally, because the recidivism rate is high, it 
is important to be encouraging at follow-up 
visits once a patient has stopped smoking. 
Physician encouragement throughout the 
process of smoking cessation and remaining 
tobacco free is essential. 



Conclusion 

Smoking causes PAD. Its cessation is difficult 
without support. Nicotine replacement and 
other aids to cessation should be made available 
to PAD patients. 

Obesity 

Obesity has reached epidemic proportions in 
developed countries. Obesity contributes to 
numerous medical problems including athero- 
sclerosis. It is important to recognize if patients 
are overweight and to obtain the appropri- 
ate dietary consultation to assist patients with 
weight loss. Most weight-loss programs are 
not successful unless they include lifestyle 
modification and a specific exercise program. 




> 



58 



VASCULAR SURGERY 



Conclusion 

Obesity contributes to the development of type 
2 diabetes, hypertension, and dyslipidemia, all 
known to be associated with PAD. Appropriate 
weight-reduction programs should be recom- 
mended. It should be noted that smoking cessa- 
tion promotes weight gain, and that in a patient 
with PAD smoking cessation is the most impor- 
tant element in risk reduction. A level of weight 
gain therefore should be tolerated, at least in the 
short term. 



Diabetes Mellitus 

Peripheral arterial disease and diabetes mellitus 
frequently occur together. Probably half of all 
patients with diabetes mellitus have evidence of 
PAD 10 to 15 years after diabetes onset. Fur- 
thermore, glucose intolerance correlates with 
angiographic disease extent. Diabetic patients 
account for one third of below-knee amputa- 
tions and 50% of amputations when distal 
amputations are included. The pathology of the 
diabetic limb is multifactorial, including contri- 
butions from both micro- and macrovascular 
disease. Asymptomatic PAD occurs in 20% of all 
diabetic patients with no other evidence of 
vascular disease (POPADAD screening of 8000 
patients). Thus the occurrence of overt diabetes 
and PAD is well documented. We are less expert 
at detecting early diabetes in our patients with 
PAD, however. Distal disease on angiography or 
increased ABI (due to vessel stiffening) can lead 
to the retrospective diagnosis of diabetes melli- 
tus, but this still underestimates the figure. Of 
100 consecutive patients presenting at our vas- 
cular surgery clinic, 40% had abnormal glucose 
tolerance tests. The majority of these patients 
had normal random or fasting sugars and were 
diagnosed only on the basis of this glucose tol- 
erance test. Currently we do not give a glucose 
tolerance test to all PAD patients but do use this 
form of diagnosis for patients who have raised 
ABI in the presence of symptomatic IC, in those 
in whom the distal distribution of the PAD 
would arouse a suspicion of diabetes, and in 
those with a "diabetic" lipid profile where both 
cholesterol and fasting triglyceride levels are 
elevated and high-density lipoprotein (HDL) 
levels are low. Detection of asymptomatic dia- 
betes mellitus in the PAD patient is a major part 
of the medical management of these patients. It 



is consistently underdiagnosed in PAD and this 
can have serious consequences. 

Diabetes contributes to atherosclerosis, and 
studies have indicated that tight glucose control 
limits the progression of end-organ damage due 
to diabetes, including atherosclerosis. Physi- 
cians involved in the care of diabetic patients 
with atherosclerosis must work closely, as a 
team, with other caregivers involved in the man- 
agement of diabetes to ensure tight glucose 
control. 

Conclusion 

It should be remembered that all risk factors 
are synergistic in terms of vascular disease, not 
merely additive, and the failure to diagnose 
underlying diabetes in a PAD patient who 
smokes will have serious consequences for that 
patient. 

Dyslipidemia 

Abnormal lipid profiles are well recognized in 
patients with atherosclerosis. The relationship 
between cholesterol level and CAD risk is con- 
tinuous, with no obvious "safe" cut-off point. 
Patients with PAD are likely to be identical. A 
number of studies investigated cholesterol in 
PAD and found it to be a significant, though 
weak, risk factor for claudication, and above the 
age of 55 years to correlate with ABI. Reduced 
HDL is associated with increased PAD severity, 6 
with strong inverse relationships between HDL 
cholesterol and PAD that persist after adjust- 
ment for other risk factors. Furthermore, ele- 
vated serum triglyceride levels have been 
reported in both cross section and longitudinal 
studies. It has been suggested that the link 
between increased triglyceride level in patients 
with PAD and the development of the disease 
might be explained by the increase in low- 
density lipoprotein (LDL) providing the 
enhanced vascular risk. 

Despite the links between abnormal lipid 
profiles and PAD, there have been no clinical 
studies with the PAD patient as primary end 
point. The situation has now been overtaken by 
the Heart Protection Study (2002) and various 
guidelines with recommendations for lipid- 
lowering therapy if there is a >3% chance of a 
vascular event per annum. Such a risk clearly 
occurs in patients with PAD. 



59 



MEDICAL MANAGEMENT OF PERIPHERAL ARTERIAL DISEASE 




All of the major trials in the field of lipid 
lowering that have been reported over the 
past decade have showed variously a decrease 
in CAD mortality, major coronary events, 
and the need for coronary revascularization. 
Together the data suggest that four out of nine 
deaths occurring in a group of subjects with 
cholesterol >5.5mmol/L will be prevented by 
treatment. 

More recently, the Heart Protection Study 
(HPS) demonstrated for the first time a benefit 
from statin therapy for prevention of vascular 
events in patients with PAD (HPS Collaborative 
Group, 2002). This largest-ever trial of choles- 
terol-lowering therapy enrolled more than 
20,000 high-risk patients, of whom 2700 had 
symptomatic PAD but no prior CAD. Treatment 
with simvastatin 40mg/day reduced major vas- 
cular events (coronary events, stroke, and revas- 
cularization) by 20% in patients with PAD, a 
similar reduction to that observed in the study 
overall. Moreover, the benefits observed in the 
HPS were not influenced by baseline levels of 
blood lipids. The findings of the HPS, therefore, 
strongly suggest that statin therapy should be 
considered on the basis of high risk rather than 
high cholesterol, a category into which patients 
with PAD undoubtedly fall. 

Conclusion 

Elevated cholesterol and triglycerides contri- 
bute to the development and progression of 
atherosclerosis. Thus, it is important to obtain a 
lipid profile on any patient with atherosclerosis. 
Management of dyslipidemia initially involves 
dietary change followed by specific medical 
management if necessary. The goal of lipid- 
lowering therapy is to achieve a serum LDL 
cholesterol concentration less than lOOmg/dL 
or 3mmol/L and a serum triglyceride less than 
150mg/dL. 

The first choice for pharmacological therapy 
is a statin. 

Hypertension 

Although it has been suggested that raised sys- 
tolic and raised diastolic blood pressure (BP) 
are linked to PAD development, results from 
prospective studies are less convincing. Target 
levels of blood pressure have been clearly 
defined in a number of guidelines, For example 



the Scottish Intercollegiate Guidelines Network 
(SIGN) guideline on the management of hyper- 
tension and the British Hypertension Society 
guidelines have given clear guidance in terms of 
acceptable BP levels. Therapy should be started 
in all PAD patients with sustained systolic BP 
elevations recorded above 140mmHg or dias- 
tolic BP above 90mmHg. The optimum target 
blood pressure is a systolic BP of <135mmHg 
and a diastolic of <85mmHg. A more stringent 
target is required for those patients with 
diabetes and this is important, as many PAD 
patients have diabetes nielli tus where a target 
of <130/80mmHg is recommended. Three 
long-term double-blind studies (Materson et al., 
1993) have compared all the major classes of 
antihypertensive drug therapy and overall 
showed no consistent or important differences 
in terms of efficacy of BP control, side effects 
from the drugs, or quality of life. It has been 
suggested that hypertensive patients whose BP 
is controlled by thiazides or beta-blockers may 
continue to experience the excess risk of coro- 
nary death. These two drug classes change 
glucose intolerance and lipid profiles in a dose- 
dependent fashion, and it may be that the lower 
doses of thiazides currently employed do not 
show this effect. Although (3-adrenergic antago- 
nist drugs have been previously reported to 
enhance PAD symptoms through their vasocon- 
strictor effects, more recently it has been sug- 
gested that this is not so. It is of interest that the 
new vasodilating beta-blockers do not even 
have this theoretical contraindication and might 
be used safely in patients with PAD. The use 
of angiotensin-converting enzyme inhibitors 
in patients with PAD may confer protection 
against future atherosclerotic events. However 
care must be taken in the presence of renal 
artery stenosis, another common finding in 
patients with PAD. 

Care must be taken when treating hyperten- 
sion in the patient with CLI, as limb perfusion 
might only be maintained through the elevated 
BP. Too profound or rapid a decrease in BP may 
worsen the symptoms, and care must be taken 
with CLI in the same way as with patients with 
severe carotid disease. 

Conclusion 

Hypertension has been implicated in the 
etiology of PAD and contributes to its vascular 




> 



60 



VASCULAR SURGERY 



comorbidity. Blood pressure should be con- 
trolled to levels consistent with local guidelines. 
Polypharmacy is likely to be required. Caution 
is advised when high BP is diagnosed in a 
patient with CLI, as too rapid a decrease in pres- 
sure may worsen the limb ischemia. 

Sedentary Lifestyle/ 
Exercise Therapy 

Intermittent claudication is a symptom of lower 
limb peripheral arterial disease. It arises when 
the blood flow is insufficient to meet the meta- 
bolic demands of the leg muscles in ambulating 
patients. Intermittent claudication is a "lever- 
aged" disability, as pain increases with walking, 
patients walk shorter distances, muscle strength 
erodes, and walking distances continue to 
decrease. This leads to other negative conse- 
quences such as weight gain, hypertension, and 
diabetes. Overall, patients with claudication 
have a 60% lower functional capacity than 
age-matched individuals without the disease 
(Eldridge and Hossack, 1987). 

Intermittent claudication is a symptom of 
systemic atherosclerosis. At least 60% of pa- 
tients with claudication have significant disease 
of the cerebral or cardiac circulation. Mortality 
rates for patients with claudication are very 
high, with 30% to 50% of patients dying from 
cardiovascular causes within 5 years of the 
initial diagnosis. A meta-analysis of 21 studies 
on the effects of exercise on patients with clau- 
dication suggested that the average improve- 
ment in walking distance was 122% (Gardner 
and Poehlman, 1995) and the benefits have 
been shown to be as high as 180% (Fig. 6.2). 
The programs with the greatest benefit were 
those in which patients exercised for 30 minutes 
at least three times a week for 6 months. How- 
ever, supervised exercise programs are not 
currently covered by most medical insurance 
policies. 

There are numerous salutary effects of 
exercise that contribute to the reduction of 
cardiovascular events. Exercise is associated 
with beneficial changes in body fat percentage, 
lipoprotein profile, carbohydrate tolerance and 
insulin sensitivity, neurohormonal release, and 
blood pressure. There is substantial evidence 
that regular aerobic exercise can even alter 





















hJ 






o 








N 

< 








H 






u 


t/3 




- 


Pi 


o 


O 




w 


uJ 


m 


- 


X 

w 




1 — 1 

o 


< 

H 


w 
u 

< 

PL, 






Oh 


























1 — 1 



W200 

i h 150 

|l 10 ° 

is son 

u < 
** 



Figure 6.2. Walking distance.The improvement in walking dis- 
tance is much greater for a graded exercise program compared 
to the best medical management (cilostazol), percutaneous 
transluminal angioplasty (PTA),or placebo. 



vessel structure. The progression of coronary 
lesions can be inhibited in those patients who 
modify risk factors and engage in regular exer- 
cise. Numerous studies have demonstrated a 
significant correlation between exercise and 
an increase in vessel diameter. Exercise may 
also induce changes in the lumen diameter in 
patients after coronary angioplasty. Patients 
randomized to a 12-week intervention program 
consisting of daily exercise after balloon 
angioplasty of the coronary vasculature had 
a significantly lower rate of re-stenosis than 
patients in the control group. However, the 
major benefit of exercise is likely due to the 
training response. 

Thrombophilia 

Patients under the age of 50 with manifestations 
of vascular disease have an increased risk of a 
defined hypercoagulable disorder. Thus, some 
believe that these patients should be screened 
for hypercoagulable disorders. In contrast, most 
hematology societies indicate that screening for 
thrombophilia in arterial disease does not yet 
have an evidence base. 

There are two general categories used to 
describe the hypercoagulable or prothrombotic 
state: hereditary and acquired. The former is 
often referred to as inherited thrombophilia 
disease. Acquired thrombophilia is a term re- 
served for well-defined syndromes such as the 
antiphospholipid syndrome (APS). This was 



61 



MEDICAL MANAGEMENT OF PERIPHERAL ARTERIAL DISEASE 




previously termed the lupus anticoagulant. 
However, the name was changed for two very 
good reasons. First, the anticoagulant referred 
to in this latter term reflects the behavior of 
the blood in the test tube, and in fact a pro- 
thrombotic effect is observed in vivo. Second, 
although it can be associated with the connec- 
tive tissue disease systemic lupus erythemato- 
sus, this is by no means necessary and we 
are increasingly recognizing APS occurring in 
isolation. 

Although the inherited thrombophilic disor- 
ders are historically linked to venous thrombo- 
sis, there is some evidence suggesting that they 
may contribute to arterial thrombosis and in 
particular in patients with PAD. It has been 
recorded that inherited thrombophilias link to 
failure of vascular grafting following surgery. In 
a prospective study hypercoagulability occurs 
in between 10% and 25% of patients, increasing 
to about 50% in the presence of clinical markers 
for thrombophilia. Preoperative identification 
of such patients is important, as the short- 
and medium-term failure rates of the graft are 
reported to be approximately 50% in such 
patients. Furthermore, such patients are at 
increased risk of deep vein thrombosis during 
the operative procedure (see Chapter 5 for the 
evaluation of these patients). 



to 40% of patients presenting with premature 
PAD have been found to have heterozygous 
homocystinemia. Pyridoxine supplementation 
reduces the thrombotic events in homozygous 
patients. However, it is not known whether this 
or other vitamins such as folate affect the course 
of the premature atherosclerosis in the het- 
erozygous sufferers. 



Vascular Risk Factor 
Management in Patients with 
Peripheral Arterial Disease 

The presentation of a patient with PAD to a 
provider of medical care presents an ideal 
opportunity for the critical assessment of vas- 
cular risk factors. Intervention at this time both 
decreases the risk of coronary and cerebral 
events and is likely to limit progression of the 
arterial disease in the periphery, although the 
latter is theoretical rather than evidence-based 
as yet. Thus the modification of vascular risk is 
of crucial importance in the patient with PAD 
and is likely to become an even more important 
area in the future for clinicians involved with 
the care of these patients. 



Conclusion 

As the inherited hypercoagulable states such as 
protein C, protein S, antithrombin II deficien- 
cies, and factor V Leiden occur only infrequently 
in this patient group, it is not possible to make 
a formal recommendation for screening of PAD 
patients. Probably a selective approach is war- 
ranted, reserving screening to those patients 
with bypass failure, a history of thrombotic 
events, or atherosclerosis at an early age. 



Homocystinemia 

The clinical features of homocystinemia include 
the development of premature atherosclerosis, 
along with manifestations of arterial and 
venous thrombosis. Disease severity leads to 
early diagnosis in homozygous patients, but 
patients with heterozygous disease present 
merely with premature atherosclerosis; 20% 



Drug Treatment of 
Intermittent Claudication 

It is a poor reflection on us, as clinicians 
involved in the care of the PAD patient, that 
angina of the legs is addressed far less aggres- 
sively than symptoms of claudication in the 
heart! Part of the problem reflects the concern 
about the effectiveness of the drug treatment for 
the symptoms of intermittent claudication and 
the consequent unease over the use of financial 
resources to purchase these compounds. Guide- 
lines have been developed for prescribing. There 
are four oral drug therapies that have a license 
for use as a treatment for IC in the United 
Kingdom and two in the United States: 
Naftidrofuryl (Praxilene, not available in the 
U.S.), oxpentifylline (Trental), cilostazol 
(Pletal), and inositol nicotinate (Hexopal). 
These drugs have been evaluated in clinical 




> 



62 



VASCULAR SURGERY 



trials in terms of their effectiveness for alleviat- 
ing the symptoms of leg pain associated with 
walking (IC). 

Naftidrofuryl 

Naftidrofuryl is thought to mediate its benefits 
through vasoactivity (vasodilatation) and via a 
local anesthetic action. Studies have docu- 
mented increased tissue oxygenation, increased 
ADP levels, and reduced lactic acid. The recom- 
mended maximum dose is 200 mg three times a 
day. 

A number of double-blind, placebo- 
controlled studies in this area tend to show a 
significant placebo response in walking dis- 
tance approximately 25% improvement in 
walking distance with placebo. With Naftidro- 
furyl a further 30% improvement can be 
expected. These estimates are supported by two 
meta-analyses. 



Pentoxifylline/Oxpentifylline 

This is a rheological agent that has been 
approved for the treatment of intermittent 
claudication. Only two of the double-blind, 
placebo-controlled studies of oxpentifylline that 
measured walking distance using treadmills 
showed any statistical improvement in such 
walking distance by patients on oxpentify- 
line. Furthermore, one of these was a retrospec- 
tive subanalysis of short-distance claudicants, 
patients who could only walk short distances 
before claudication ocurred already included in 
another study. A meta-analysis of ten random- 
ized, double-blind, controlled studies concluded 
that the limited amount and quality of data for 
this drug precluded an overall reliable estimate 
of its efficacy. We recommend, in the absence of 
any consistent clinical trial evidence, that oxpen- 
tifylline should not be prescribed for use in this 
indication. 



Cilostazol 

Cilostazol has recently been approved for the 
treatment of intermittent claudication. It is a 
phosphodiesterase inhibitor that has been 
shown in randomized placebo-controlled trials 
to improve walking distance by approximately 



50%. However, the improvement remains 
modest compared to exercise programs (180% 
to 200% improvement). Cilostazol is contraindi- 
cated in patients with a history of congestive 
heart failure. 

Inositol Nicotinate (Hexopal) 

Inositol nicotinate is licensed for use in the 
United Kingdom for patients with intermittent 
claudication. However, the evidence base for this 
compound is weak. Of the four double-blind, 
randomized, placebo-controlled trials, three 
were primary care based and used subjective 
or questionable objective criteria for assessment 
of IC without treadmill use. None showed clear 
evidence of improvement in symptoms with 
drug use. We suggest, therefore, that the drug 
may not be of value for patients with IC. 

Conclusion 

Some patients with IC merit drug treatment 
due to the severity of their symptoms. It is rea- 
sonable to consider Naftidrofuryl or cilostazol 
for the symptomatic relief of moderate disease. 
The patients, however, should be reviewed 6 to 
12 months after drug commencement to assess 
its efficacy and the need for continuation. 
Neither oxpentifylline nor inositol nicotinate 
can be recommended for the treatment of 
symptomatic IC. 



Summary 



Peripheral arterial disease is a marker of sys- 
temic atherosclerosis. Patients with PAD are at 
high risk for MI and stroke. Thus, an integrative 
approach to risk factor modification and the 
prevention of the sequelae of atherosclerosis 
is the mainstay of therapy. Patients must be 
advised to "stop smoking and keep walking." In 
addition, however, antiplatelet therapy (e.g., 
aspirin) is indicated in all patients with periph- 
eral arterial disease in whom there is no 
contraindication. Hypertension must be appro- 
priately treated and diabetes mellitus detected 
and managed optimally. The medical manage- 
ment of the symptoms of intermittent claudica- 
tion should also be addressed if these are 
significantly impairing the patient's lifestyle. 



63 



MEDICAL MANAGEMENT OF PERIPHERAL ARTERIAL DISEASE 




The key feature, however, in managing these 
patients is to assess their overall vascular risk 
and treat accordingly. 

References 



Belch JJ, Sohngen M, Robb R, Voleske R Sohngen W. (1999) 
Int Angiol 18:140-4. 



Diener HC, Cunha L, Forbes C, Sivenius J, Smets P, 
Lowenthal A. (1996) J Neurol Sci 143:1-13. 

Eldridge JE, Hossack KF. (1987) Cardiology 74:236-40. 

Gardner AW, Poehlman ET. (1995) JAMA 274:975-80. 

Materson BJ, Reda DJ, Cushman WC, et al. (1993) N Engl J 
Med 328:914-21. 

Silagy C, Mant D, Fowler G, Lodge M. (1994) Lancet 343: 
139-42. 



7 



Anesthesia for Vascular Surgery 

Jamal J. Hoballah and Farid Moulla 




Surgery of the peripheral vascular system 
requires technical precision and perioperative 
vigilance. The outcome of vascular procedures 
depends on various factors. These factors 
include patient selection, the procedure per- 
formed, the surgeon's skills, and the periopera- 
tive care. The importance of the perioperative 
care cannot be underestimated. Patients pre- 
senting with a vascular pathology often have 
comorbidities. The incidence of coronary artery 
disease (CAD) in patients with carotid disease 
is estimated at 50%. 

Patients with operative lower extremity 
peripheral vascular disease (PVD) have an 
even higher incidence of CAD. Hypertension, 
diabetes, and renal insufficiency are all more 
frequent in patients with PVD compared to 
the general population. Following aortic 
surgery, carotid endarterectomy, and lower 
extremity revascularization procedures, the 
most common major complication is a cardiac 
event. Anesthesia is a risk factor contributing 
to the perioperative morbidity and mortality 
of the vascular patient. The preoperative 
preparation and the intraoperative manage- 
ment can predict and influence the postopera- 
tive course. This chapter provides an overview 
of the various anesthetic techniques that can 
be used in patients presenting for vascular 
procedures. 



Anesthesia Risks 

Depending on the complexity of the vascular 
pathology, a vascular procedure can be per- 
formed under local, regional, or general anes- 
thesia, or a combination of these techniques. 
Minimally invasive approaches to vascular 
reconstructions are continuing to be developed, 
allowing for more localized anesthesia. Often it 
is assumed that performing a procedure under 
local or regional anesthesia results in lower 
mortality and morbidity and allows earlier 
recovery. Certain trends are apparent in the 
literature, although not without controversy, 
comparing these anesthetic techniques and can 
be outlined as follows: 

• While laboratory and monitoring tech- 
niques are improving, historically many 
perioperative cardiac events are "silent" 
(Steen et al., 1978; Von Knorring and 
Lepantalo et al., 1986). 

• There does not appear to be a discernible 
difference between regional and general 
anesthesia with regard to cardiac risk or 
mortality within the field of vascular sur- 
gery. There is a large amount of recent con- 
troversy, practice variability, and research 
related to this point (Bode et al., 1996; 



65 




» 



66 



VASCULAR SURGERY 



Christopherson et al, 1993; Rigg et al.,2002; 
Rivers et al, 1991; Rodgers et al.,2000). 

• Regional and general anesthesia offer dif- 
ferent advantages and disadvantages, but 
both are safe and appropriate for carotid 
endarterectomy (Rockman et al., 1996). 

• Combinations of anesthetic techniques 
may offer advantages over single-mode 
therapy. 

• There is a trend toward less need for 
(immediate postoperative) reoperation for 
thrombosis when regional anesthesia is 
employed in lower extremity revascular- 
izations (Christopherson et al., 1993). 

• Minimally invasive techniques warrant 
equally minimally insulting anesthetic 
techniques, contributing to improved out- 
comes over standard surgical intervention. 

Because of the relative equity of major anes- 
thetic risk between regional and general anes- 
thesia, the site and probable duration of the 
procedure itself are often the main determi- 
nants of anesthetic technique. Lower extremity 
revascularizations typically last from 2 to 6 
hours with significant variability. A single 
administration of spinal anesthesia offers no 
ability to redose if the initial infiltration wears 
off. Epidural anesthesia can be continuously 
infused both for long procedures and for post- 
operative pain management. However, it does 
little to treat the musculoskeletal complaints of 
an awake patient prostrate for several hours. 
Conversely, general anesthesia does not allow 
for patient interaction and requires prophylac- 
tic measures to avoid the complications of 
pressure and immobility. It also has the dis- 
advantage of requiring mechanical ventilation 
with its potential respiratory complications and 
possible hemodynamic stresses with induction 
or extubation. In most cases, regional and 
general anesthesias are both acceptable alterna- 
tives for the common vascular surgeries. Some 
issues to remember when deciding on anes- 
thetic techniques are as follows: 

• Open abdominal vascular surgery such as 
for an abdominal aortic aneurysm (AAA) 
is theoretically possible with a celiac axis 
block or regional anesthesia, but in prac- 
tice, warrants general anesthesia. 

• Epidural anesthesia for supplementation 
of general anesthesia, as well as for post- 
operative pain management in major 



abdominal surgery, is becoming the stan- 
dard of care as it gains a wealth of sup- 
porting literature. 

• The need for postoperative full anticoagu- 
lation with (or the use of even prophylactic 
subcutaneous doses of) low-molecular- 
weight heparin usually precludes epidural 
catheter use for fear of developing an 
epidural hematoma upon the removal of 
the catheter. 

• Both prophylactic and therapeutic un- 
fractionated heparin therapy have to be 
interrupted to allow for normalization of 
coagulation prior to removing the epidural 
catheter to avoid a potentially devastating 
complication. 

• Upper extremity vascular procedures can 
be approached with general, regional, 
local, or epidural anesthesia. Axillofemoral 
bypass is a relative contraindication to 
regional anesthesia as is harvesting of arm 
veins, although on occasions the vein may 
be harvested using infiltration of local 
anesthetics. 

• The most common vascular surgery, 
carotid endarterectomy, has been studied 
extensively, with ongoing debate regarding 
the effectiveness of intraoperative moni- 
toring techniques and regional versus 
general anesthesia. 

The following sections review the various 
anesthetic techniques available for commonly 
performed vascular procedures. 

Anesthesia for Endovascular 
Interventions 

Endovascular procedures vary in complexity 
and include diagnostic angiograms, balloon 
angioplasty and stenting, endovascular aortic 
aneurysm treatment with endografts, and vari- 
cose vein treatment by radiofrequency or laser 
ablation. There is a paucity of studies on the 
anesthetic requirements of vascular patients 
treated by endovascular techniques. In general, 
percutaneous angiography, balloon angioplasty, 
and stenting are typically performed under 
local anesthesia. Infiltration of the skin with 1% 
lidocaine or bupivacaine can provide ample 
control of pain at the puncture site. It is prefer- 
able to premedicate the patient prior to the pro- 



67 



ANESTHESIA FOR VASCULAR SURGERY 




cedure and to augment sedation at the onset 
of the intervention. Premedication with 25 to 
50 mg of diphenhydramine and 5mg of 
diazepam given orally 1 hour prior to the pro- 
cedure relieves many patients of the anxiety and 
anticipation of the intervention. Once the 
patient is on the catheterization table, sedation 
with 1 to 2 mg of lorazepam in addition to 50 [ig 
of fentanyl provides further sedation and excel- 
lent pain control. The skin at the puncture site 
is infiltrated with the chosen local anesthetic 
and the potential track of the puncture needle 
is also infiltrated. If during the arterial puncture 
the patient complains of pain, additional local 
anesthetic can be infiltrated in the area directly 
through the Seldinger needle prior to arterial 
puncture. 

Endovascular Aortic 
Aneurysm Repair 

With respect to aortic aneurysm endovascular 
treatment, most of the procedures were origi- 
nally done under general anesthesia. As the 
comfort level of the surgeons with the proce- 
dure increased, epidural and local anesthesia 
started to be used and were noted to be a rea- 
sonable alternative to general anesthesia. The 
idea of being able to treat an abdominal aortic 
aneurysm under local anesthesia is very excit- 
ing and appealing. The question of need for 
general anesthesia to treat AAA with endovas- 
cular technology has been examined (Henretta 
et al., 1999). In 47 patients treated with local 
anesthesia and intravenous sedation without 
intubation, only one required conversion 
to general anesthesia. The conversion was 
needed to repair an injury to the external iliac 
artery. In the remaining 46, there were lower 
rates of cardiac complications (zero) and 
shorter hospital stay when compared to those 
reported with the open technique. Although 
this study did not compare the results of 
endovascular AAA treatment under general 
anesthesia versus local or epidural anesthesia, it 
clearly proved the feasibility and efficacy of 
local anesthesia with sedation as an anesthetic 
technique for endovascular AAA treatment. At 
the University of Iowa, epidural anesthesia is 
our preferred technique for endovascular AAA 
treatment. However, patients' wishes and prefer- 
ences often play a decisive role in the selection 
of the anesthetic technique used. The require- 



ments for anesthesia are likely to change as 
rapidly as the endovascular techniques and 
devices employed. 

Lower Extremity Vein Therapy 

Radiofrequency or laser ablation of the greater 
saphenous vein (GSV) is gaining popularity as 
an alternative, less invasive method for GSV 
stripping in the treatment of varicose veins. 
This transforms varicose vein treatment into an 
office practice and limits the anesthesia needs to 
infiltration of local anesthetics. The skin is typ- 
ically infiltrated at the site of insertion of the 
sheath through which the laser or radioablation 
catheter is introduced. Because of the heat 
generated with the venous ablation, additional 
anesthesia is needed along the course of the 
GSV. Tumescent anesthesia is used for this 
purpose. Tumescent anesthesia is prepared by 
the following concentration: 

500 cc of normal saline in IV bag 
16 cc of 8.4% sodium bicarbonate solution 
50 cc of 1% lidocaine with epinephrine 
1:100,000 

It is infiltrated along the course of the GSV using 
a long spinal needle. In addition to providing 
anesthesia to the infiltrated area, tumescent 
anesthesia provides a protective layer between 
the vein and the dermis to avoid thermal skin 
injury during the laser or radiofrequency abla- 
tion. This tumescent anesthesia is also used 
during stab avulsion of branch varicosities. In 
addition to providing pain control, it is useful in 
minimizing subcutaneous bleeding from the 
ends of the avulsed veins. 

Anesthesia for Open 
Aortic Surgery 

Physiological and Mechanical 
Considerations 

Open aortic procedures are typically performed 
under general anesthesia. The anesthesia issues 
in open aortic surgery depend on the level of 
aortic clamping and the extent of the surgical 
incision. These issues include the need for 
cardiopulmonary arrest, spinal cord protection, 
renal protection, and management of the hemo- 
dynamic changes associated with clamping and 




> 



68 



VASCULAR SURGERY 



unclamping of the aorta. Other issues include 
the need for single-lung ventilation, intraopera- 
tive hemodynamic monitoring, and the use of 
combined epidural and general anesthesia. 
Although aortic valve, ascending aortic, and 
arch vessel surgery often require cardiopul- 
monary bypass, debate still exists regarding 
descending thoracic and suprarenal aortic 
surgery. For descending thoracic and suprarenal 
aortic surgery, the approaches include the clamp 
and sew technique versus the use of an adjunc- 
tive atriofemoral bypass, aortofemoral bypass, 
or temporary axillofemoral bypass. 

In patients requiring a thoracoabdominal 
incision, the placement of a double-lumen or 
balloon excluder endotracheal tube is essential 
to deflate the left lung when needed and main- 
tain the patient on single-lung ventilation. Such 
a maneuver facilitates better exposure to the 
thoracic aorta. With respect to spinal cord pro- 
tection, maintaining distal and pelvic perfu- 
sion, reimplantation of intercostals vessels, 
and control of spinal fluid pressure have been 
described as essential to minimizing spinal cord 
ischemia during thoracoabdominal aortic 
surgery. Although debatable, a spinal catheter is 
usually inserted prior to induction of general 
anesthesia, and the spinal pressure is monitored 
intraoperative ly and for 2 to 3 days after the pro- 
cedure. Spinal fluid is drained to keep the spinal 
pressure below 10 mm Hg or 14 cm H 2 0. Rever- 
sal of spinal cord ischemia has been observed in 
situations where neurological symptoms have 
developed in the postoperative period in associ- 
ation with elevated spinal fluid pressure. Debate 
still exists with respect to hemodynamic goals 
and monitoring, visceral organ protection, and 
control of cardiac responses to clamp stresses. 

Aortic cross-clamping leads to an increase in 
cardiac afterload. This acute increase in after- 
load is both measurable and manipulable, 
directly and indirectly. Uncontrolled hyperten- 
sion, although tolerated by the healthy heart, 
can lead to systolic or diastolic dysfunction and 
cardiac decompensation. Blood pressure is 
often viewed as dependent on circulating blood 
volume, cardiac preload and afterload, and 
cardiac function. All of these factors are 
influenced by, and essentially precluded by, the 
ability of the heart to withstand the stress of any 
acute physiologic change. The effects of aortic 
cross-clamping are initially mechanical due to 
abrupt change of afterload. The rough percent- 



age of cardiac output blocked by clamping at 
various levels is as follows: infrarenal, 10% to 
15%; suprarenal, 15% to 25%; and supraceliac, 
55%. The precise percentage varies with certain 
disease states, such as aortic occlusive disease 
and collateralization. Typically, the surgeon 
works in concert with the anesthesiologist to 
decrease the afterload (lower the blood pres- 
sure) prior to applying a clamp on the aorta. 
In thoracoabdominal procedures where an 
aortofemoral or axillofemoral bypass is being 
used, the increase in afterload with cross- 
clamping is less pronounced. The use of the car- 
diopulmonary pump in these procedures 
facilitates easier manipulation and removal of 
circulating blood volume to treat hypertension. 
The importance of continuous communication 
between the surgeon and the anesthesia team 
cannot be overemphasized. Cross-clamping or 
unclamping of the aorta should not be per- 
formed before the anesthesia team has made all 
the necessary interventions needed to address 
the hemodynamic changes expected with the 
aortic clamp application or removal. 



Pharmacological Considerations 

Although mechanical intervention related to 
circulating blood volumes are undertaken rou- 
tinely, so too is pharmacological control of the 
vascular system. Nitroprusside and nitrogly- 
cerine titrations are often employed to decrease 
afterload and preload, respectively. The advan- 
tages of nitroprusside include its effectiveness 
in peripheral arteriolodilatation and treatabil- 
ity Furthermore, the vasodilatation of the vas- 
cular beds causes decreased oxygen extraction 
ratio and decreased cardiac work. Nitroprusside 
effects cardiac preload to a lesser extent, but it 
can possibly decrease the perfusion pressure 
to infra-clamp tissues provided by collateral 
arterial supply due to arteriolar dilatation. A 
common side effect is pulmonary vasoconstric- 
tion, arteriovenous shunting, and resultant 
desaturation of blood. Furthermore, prolonged 
use of nitroprusside can result in systemic 
cyanide toxicity. Nitroglycerine decreases 
cardiac preload and is titratable. It vasodilates 
coronary circulation and indirectly decreases 
cardiac work. It does not effect infra-clamp 
organ perfusion and has less systemic toxicity, 
seen as methemoglobinemia; however, it is not 



69 



ANESTHESIA FOR VASCULAR SURGERY 




as potent as nitroprusside. Other factors for 
visceral protection include the use of mannitol 
12.5 g intravenously a few minutes prior to 
suprarenal cross-clamping in an attempt to 
decrease renal reperfusion injury. 



Anesthesia in 
Carotid Surgery 

General anesthesia is the traditional anesthetic 
technique for carotid endarterectomy (CEA). In 
1962 Spencer and Eiseman described a local 
anesthetic technique for CEA. A "cervical block" 
is regional anesthesia consisting of infiltration 
of the paravertebral nerve roots at C2-C4 with 
one or a mix of local agents. General anesthesia 
and regional anesthesia are equally acceptable 
approaches to anesthesia for a patient undergo- 
ing a carotid endarterectomy and depend on the 
experience of the operative team. 

Pros and Cons of Regional 
Anesthesia and General Anesthesia 
for Carotid Endarterectomy 

The advantage of general anesthesia goes 
beyond keeping the patient motionless. The 
brain's metabolic demand is usually reduced 
with general anesthesia, and thus acts as a 
protective mechanism against ischemia. The 
disadvantages of general anesthesia in carotid 
surgery include the need to routinely place a 
shunt or the use of EEG or stump pressure mon- 
itoring to selectively shunt. The decision to 
shunt is sometimes determined preoperatively 
in cases of contralateral occlusion, history of 
ipsilateral stroke, or known lack of collateral 
circulation (from cerebral angiogram). In these 
cases, many surgeons would routinely shunt 
during general anesthesia. 

The advantages of regional anesthesia are 
more often seen in a different patient popula- 
tion than with that of general anesthesia. 
Published selective shunt rates in regional 
anesthesia are in the range of 15%, which is a 
lower rate of shunting than techniques that 
involve monitoring. Regional anesthesia has 
been shown to invoke less postoperative hyper- 
tension and hypotension and reduced posto- 
perative intensive care unit and hospital stay. 



"Cerebral protection" in CEA refers to pro- 
tection of the brain from ischemia induced by 
blood flow changes at the time of CEA. Cerebral 
protection can be obtained from a shunt, or 
monitoring for its need. The placement of a 
shunt from the common carotid artery to the 
internal carotid artery is the standard way 
to maintain intracerebral blood flow during 
CEA. The disadvantages of shunting include 
potential injury to the proximal and distal ends 
of the shunted vessel (intimal flaps), increased 
blood loss, the rare potential for embolism 
through the shunt, and the technical aspects of 
performing the endarterectomy around the 
shunt device. It is for these reasons that selective 
shunting is advocated by some. Monitoring for 
significant cerebral effects of carotid cross- 
clamping has been shown to decrease the need 
for shunting, and thus allows for use of a selec- 
tive shunt only if brain tissue is thought to 
be compromised. Electroencephalography, tran- 
scranial Doppler, and ipsilateral stump pres- 
sure measurement are techniques most often 
employed. 

Cerebral perfusion pressure (CPP) is equal to 
the mean arterial pressure (MAP) minus the 
intracranial pressure (ICP). Thus, an additional 
approach to cerebral protection is to maintain 
an elevated MAP during CEA. One approach is 
to maintain the patient's blood pressure at the 
same level or within 20 mm Hg higher than 
the patient's baseline blood pressure. Again, 
this requires a coordinated effort between the 
surgeon and anesthesiologist. 

Regional Anesthesia Techniques 
for Carotid Endarterectomy 

The dermatomal distribution of the neck inner- 
vation is in cervical nerves 2 through 4. Regional 
block anesthesia is targeted at these nerves in a 
paravertebral location. The cervical plexus is 
comprised of the anterior divisions of cervical 
nerves 1 through 4. The plexus sits anterior to 
the median scalene and levator scapulae, and 
behind the sternocleidomastoid muscles. The 
cervical plexus divides into superficial and deep 
branches. It is the deep branches that innervate 
the deep structures of the neck including the 
muscles via an internal and an external series of 
nerves. The internal series consists of the fol- 
lowing nerves: communicating branches, mus- 




> 



70 



VASCULAR SURGERY 



cular branches, communicantes cervicales, and 
phrenic. The external series includes the com- 
municating and the muscular nerves. The 
superficial branches of the cervical plexus 
become the following sensory nerves: smaller 
occipital, great auricular, cutaneous cervical, 
and supraclaviculars. A deep cervical plexus 
block is a paravertebral block targeted to C2 
through C4. Due to its proximal location, an 
effective block involves the superficial and deep 
branches. A superficial cervical block has the 
advantage of not involving any nerves that 
innervate muscle including the phrenic, and 
the disadvantage of not involving the deeper 
sensory nerves that the deep branches of the 
plexus innervate. 

Deep Cervical Plexus Block: Selected 
Division Blockade Technique 

After placing the patient in a neutral supine 
position, and appropriately prepping the skin 
for aseptic technique, a 4- or 5-cm 22-gauge 
needle with a short bevel is used to instill local 
anesthetic. An example of local anesthetic that 
could be used is a half-and-half mix of 1% 
lidocaine and 0.5% bupivacaine. A total of about 
3 or 4cc of local anesthetic should be used at 
each level. To find the proper position for needle 
placement at all levels, an imaginary line can be 
drawn between the tip of the mastoid process 
and Chassaignac's tubercle of C6, which is pal- 
pable at the level of, and posterior to, the cricoid 
cartilage. The C2 injection is performed about 
1 to 1.5cm below the mastoid process on this 
line, just posterior to the sternocleidomastoid. 
Moving about 1.5 cm caudad from the C2 site 
along the same line can place injections at C3 
and C4. A horizontal line from the ramus of the 
mandible posterior can be a guide to the level of 
the C4 injection site as well. A slight caudal 
angle of the needle can prevent the needle from 
tracking to the intervertebral space, and thus 
avoiding peridural or spinal anesthesia. If the 
transverse process is hit with the tip of the 
needle, paresthesias result. This indicates that 
the needle tip is in the proper vicinity and the 
injection can be performed. 

The complications of this approach to deep 
cervical block include intravertebral artery 
injection causing convulsions, unconsciousness, 
or temporary blindness. The nerve block can 



also spread to prevertebral (superficial) fascia 
and the cervical sympathetic chain, causing 
Horner's syndrome or hoarseness secondary to 
recurrent laryngeal nerve involvement. 

Deep Cervical Plexus Block: 
Interscalene Technique 

This technique utilizes the proximity of the cer- 
vical plexus branches' proximity to each other 
to administer a single dose of anesthetic. Using 
the same needle as the selective approach but a 
larger syringe to inject 10 yo 12 cc of an equiva- 
lent anesthetic, a single bolus of local is injected 
in an "interscalene" location as follows: Palpate 
the interscalene groove separating the anterior 
and middle scalenes at a level of the cricoid again 
using Chassaignac's tubercle of C6. At this loca- 
tion, enter using a caudal and slightly posterior 
angle. A C5-6 dermatomal paresthesia is elicited, 
and 10 to 40 cc of local is injected while hold- 
ing caudal pressure. A 40-cc injection without 
caudal pressure also involves the brachial plexus. 
Regardless of the amount or pressure used, the 
ipsilateral phrenic nerve is blocked by this proce- 
dure, an effect that is usually well tolerated. 

The complications of this approach of deep 
cervical blockage include phrenic nerve involve- 
ment and the same complications as for the 
selective technique; however, there is a lower 
chance of inadvertently puncturing the verte- 
bral artery. 

Superficial Cervical Plexus Block 

Using a 22-gauge, 4-cm needle, local is injected 
just posterior to the midpoint of the posterior 
border of the sternocleidomastoid muscle. The 
injection should be angled to attempt infiltra- 
tion of the posterior and medial aspects of the 
muscle. As it only affects the cutaneous nerves, 
the complications of this approach are minimal. 



Anesthesia for Vascular 
Surgery of the Lower 
Extremity 

Transfemoral thromboembolectomy, focal vein 
bypass revisions, and even femorofemoral 
bypasses can be effectively performed under 



71 



ANESTHESIA FOR VASCULAR SURGERY 




local anesthesia. In most other procedures 
where long skin incisions, long segments of vein 
harvesting, deep exposures, and tunneling are 
required, the options include regional or general 
anesthesia. Regional anesthesia appears to avoid 
the hypercoagulability noted with general anes- 
thesia and appears to provide an early patency 
advantage with infrainguinal bypass procedures 
(Christopherson et al., 1993). However, in pro- 
longed procedures, the patient may become 
restless and uncomfortable due to prolonged 
immobility. 

Conditions that suggests avoiding regional 
anesthesia include tremor at rest, inability to 
follow commands (the level of cooperation of 
the patient), peripheral neuropathy limiting the 
sensation of pain of the distal lower extremities, 
and a history of lower back surgery or pain. In 
the presence of severe heart failure or diastolic 
dysfunction, general and regional anesthesia 
have been shown to be similar in overall 
outcome. In these high-risk patients, local anes- 
thesia with monitored anesthesia care is the best 
option if the surgical procedure is appropriate 
for such anesthesia. 

In the presence of significant pulmonary 
disease [forced expiratory volume in 1 second 
(FEV]) <1.0], the risk of prolonged intubation 
or pulmonary infection after endotracheal intu- 
bation would indicate regional anesthesia. 
However, some patients with significant pul- 
monary disease are unable to maintain a supine 
position for prolonged periods. 

Significant coagulopathy precludes spinal 
and epidural anesthesia due to the risk of 
epidural hematoma and subsequent paralysis. 
Furthermore, an infection either in the region of 
placement of a regional anesthetic or causing 
systemic sepsis usually precludes regional anes- 
thesia due to the risk of seeding the paraspinal 
space. 

During procedures involving the foot such 
as toe or forefoot amputations, an ankle 
block can be an ideal regional anesthesia. The 
technique for ankle block involves infiltrating 
the ankle circumferentially with a local anes- 
thetic just at the level of the medial malleolus. 
Further infiltration of the posterior tibial nerve 
is usually needed to achieve adequate anesthe- 
sia. The neuropathy associated with diabetes 
can lead to a nearly anesthetic foot, and pro- 
cedures can be performed with minimal 
anesthesia. 



Anesthesia for Vascular 
Surgery of the Upper 
Extremity 

In general, most vascular procedures in the 
upper extremity can be managed by local or 
regional anesthesia. In spite of the lack of evi- 
dence of the benefit of regional anesthesia 
versus general anesthesia in randomized studies 
in terms of morbidity and mortality, there is 
enough proven or anecdotal benefit in terms of 
pain relief, cost-efficiency, and time to dis- 
charge. These benefits of regional anesthesia are 
particularly valued in outpatient surgery and 
should therefore encourage its use. 

Regional Anesthesia Techniques 
for the Upper Extremity 

The brachial plexus supplies all of the motor, 
and most of the sensory, innervation to the 
upper extremity. It is important to remember 
that the medial aspect of the upper arm to near 
the elbow is enervated by the medial cutaneous 
and intercostobrachial nerves and not the 
brachial plexus. Similarly, the skin on the shoul- 
der is supplied by the "caudal" branches of the 
cervical plexus supply. The common regional 
anesthesia techniques of the upper extremity 
are categorized based on location of surgical 
dissection. 



Axillary Artery /Vein Dissection 

Axillary artery and vein exposure including 
axillofemoral bypass proximal dissection can be 
performed under local anesthesia, or with an 
intrascalene approach to block the brachial and 
cervical plexus. Applying inferior pressure on 
the interscalene groove can guide the infiltra- 
tion of local anesthesia superiorly along the 
scalene muscles to effect a predominantly cer- 
vical plexus block. Using less pressure inferiorly 
can result in a predominantly brachial plexus 
block. 



Brachial Artery /Vein 

This is the "gray zone" of brachial plexus blocks, 
as the skin overlying the brachial vessels is often 




> 



72 



VASCULAR SURGERY 



at least partially innervated by the medial cuta- 
neous and intercostobrachial nerves. However, if 
local anesthesia is used in combination with the 
brachial plexus block, adequate and safe anes- 
thesia can result. 

Radial, Ulnar,and Antecubital 
Regional Anesthesia 

The region from the elbow to the wrist is ideal 
for a brachial plexus block. The techniques 
include an intrascalene approach, as mentioned 
above, and a subclavian approach. The intrasca- 
lene approach, as mentioned in the discussion 
of cervical plexus block, above, often involves 
the ipsilateral phrenic nerve and possibly some 
cranial nerves. The subclavian approach has less 
phrenic nerve involvement (50%) and requires 
smaller volumes of anesthetic, but can lead to 
pneumothorax. 

Distal Ulnar and Radial Artery,and 
Hand Surgery 

Local anesthesia is the best option in the com- 
pliant patient. Axillary blockade is another 
option for distal arm/hand surgery and is rela- 
tively easy and safe. However, axillary blockade 
effects only the forearm, requires arm abduc- 
tion, and can lead to hematoma. 

Conclusion 

Numerous approaches to vascular anesthesia 
are available. The approach must be tailored to 
the patient and the procedure. It is important 



that the operative team, surgeons and anes- 
thetists, work in concert throughout any vascu- 
lar procedure. 

References 



Bode Jr RH, Lewis KP, Zarich SW, et al. (1996) Cardiac 
outcome after peripheral vascular surgery: comparison 
of general and regional anesthesia. Anesthesiology 
84:3-13. 

Christopherson R, Beattie C, Frank SM, et al. (1993) Periop- 
erative morbidity in patients randomized to epidural or 
general anesthesia for lower extremity vascular surgery. 
Anesthesiology 79:422-34. 

Henretta JP, Hodgson KJ, Mattos MA, et al. (1999) Feasibil- 
ity of endovascular repair of abdominal aortic 
aneurysms with local anesthesia with intravenous 
sedation. J Vase Surg. 

Rigg JRA, Jamrozik K, Myles PS, et al. (2002) Epidural anaes- 
thesia and analgesia and outcome of major surgery: a 
randomized trial. Lancet 359:1276-82. 

Rivers SP, Scher LA, Sheehan E, Veith FJ. (1991) Epidural 
versus general anesthesia for infrainguinal arterial 
reconstruction. J Vase Surg 14:765-70. 

Rockman CB, Riles TS, Gold M, Lamparello PJ, Giangola G, 
Adelman MA. (1996) A comparison of regional and 
general anesthesia in patients undergoing carotid 
endarterectomy. J Vase Surg 24:946-56. 

Rodgers A, Walker N, Schug S, et al. (2000) Reduction of 
postoperative mortality and morbidity with epidural or 
spinal anaesthesia: results from overview of random- 
ized trials. BMJ 321:1493-7. 

Steen PA, Tinker JH, Tarhan S. (1978) Myocardial reinfarc- 
tion after anesthesia and surgery. JAMA 239:2566. 

Von Knorring J, Lepantalo M. (1986) Reduction of periop- 
erative cardiac complications by electrocardiographic 
monitoring during treadmill exercise testing before 
peripheral vascular surgery. Surgery 99:610. 



8 



Nonatherosclerotic Vascular Disease 

Jonathan R.B. Hutt and Alun H. Davies 




Vasospastic Disorders 

Raynaud's Phenomenon 

In 1862 Maurice Raynaud first described the 
problem of an asphyxia and symmetrical gan- 
grene of the extremities. Raynaud's phenome- 
non is now defined as episodic vasospasm of 
arterioles in the extremities. It is classified as 
either an idiopathic form, known as primary 
Raynaud's or Raynaud's disease, or as secondary 
Raynaud's or Raynaud's syndrome, where it 
occurs in the presence of an underlying cause 
(Table 8.1). 

Raynaud's has an incidence of up to 5%. It is 
found worldwide, but due to the common pre- 
cipitant factor of cold extremities, it appears to 
have a higher incidence in countries with a low 
ambient temperature. It affects women far more 
than men at a ratio of 9: l.In the case of primary 
Raynaud's, there is evidence of a genetic predis- 
position, as 25% of sufferers have a first-degree 
relative who is affected. 

The underlying pathophysiology of Ray- 
naud's presents a complicated picture. The 
overall effect is due to vasospasm of small mus- 
cular arteries and arterioles. Many factors and 
mechanisms have been implicated in this 
process, and it is occasionally unclear whether 
they are causal as part of the primary pathology 
or consequential as a result of it. That there are 
many causes of secondary Raynaud's is testa- 
ment to the fact that there may be subtly differ- 



ent underlying mechanisms with a similar final 
end point. In a similar vein, although some 
factors may not be primary instigators of the 
problem, their activation and subsequent action 
may contribute to the severity of the pathology 
and thus provide therapeutic targets in certain 
situations. The increasing number of implicated 
mechanisms may also indicate the future poten- 
tial for further delineation of the spectrum of 
disorders associated with Raynaud's into more 
distinct pathological mechanisms. 

Broadly speaking, the etiological factors that 
have so far been implicated in the development 
of Raynaud's can be divided into local factors 
present at the immediate neurovascular level, 
and more general humoral factors (Fig. 8.1). 

The Local Microvascular Unit 

Neurogenic Mechanisms 

Vasoconstriction of the cutaneous vessels when 
induced by cold is regulated by a 2 receptors. 
This mechanism is open to derangement in the 
pathology of Raynaud's. It has been shown that 
sufferers have a reduced basal blood flow, which 
would obviously be further decreased in the 
setting of sympathetic-induced vasoconstric- 
tion. Notably, it also appears that baseline 
cutaneous blood flow is decreased in healthy 
women compared with men, which may provide 
an insight into the condition's preference for 
the female sex. Although there is no increase 
in background catecholamine circulation 
detected in affected patients, dermal arteriolar 



73 




74 



VASCULAR SURGERY 



Table 8.1 . Secondary causes of Raynaud's phenomenon 

Mechanical 

Frostbite 

Vibration 

Rheumatological disease 
Scleroderma 

I Systemic lupus erythematosus 
Dermatomyositis 
Polymyositis 
Rheumatoid arthritis 
Sjogren's syndrome 
Takayasu's arteritis 
Giant cell arteritis 



Arterial disease 

Brachiocephalic atherosclerosis 
Buerger's disease 
Thoracic outlet syndrome 

Vasospastic disorders 
Migraine 
Prinzmetal's angina 

Endocrine disorders 
Carcinoid syndrome 
Hypothyroidism 

Blood dyscrasias 
Cryoglobulinemia 
Paraproteinemia 
Polycythemia 

Infective causes 
Parvovirus B19 
Helicobacter pylori 

Drugs 
Vinblastine 
Bleomycin 
Methysergide 
Ergot alkaloids 
Beta blockers 






samples from Raynaud's patients show an exag- 
gerated response to a 2 -stimulating agents. The 
use of sympathectomy as a therapy and the 
documented benefits from the a 2 -blocking 
agent Prazosin also support a place for 
abnormal sympathetic activity in Raynaud's 
pathophysiology. 

Another neurological factor is calcitonin 
gene-related peptide, a powerful vasodilator 
found in cutaneous nerve endings. Response 
to this mediator is unaffected in diseased 
tissue, but immunocytochemistry demonstrates 
a decrease in calcitonin gene-related peptide 
levels in the local digital neurons of people with 
primary and secondary Raynaud's. Intravenous 
infusion of calcitonin gene-related peptide has 
also been shown to increase blood flow in the 
hands and may also help digital ulceration in 
more severe cases. 

Endothelial Mechanisms 

At the level of the endothelium, there are a 
number of mechanisms which may have been 
implicated in the pathophysiology of Raynaud's. 
Endothelin-1 is a vasoconstrictor derived from 
vascular endothelial cells. Increased levels of 
this mediator have been described in patients 
presenting with Raynaud's. Similarly, nitric 
oxide, an endogenous vasodilator, appears to be 
present at reduced levels. It appears, however, 
that these findings may be more consequential 
than causal. Reports on patients with primary 
Raynaud's have not shown increased levels of 
endothelin-1, and it is accepted that endothelial 
damage is more a feature of secondary disease. 
Moreover, endothelin-1 levels correlate with 
severity of disease and are also shown to 
decrease in patients with systemic sclerosis as 
digital ulcers undergo healing. Administration 
of L-arginine, a precursor of nitric oxide 
synthesis, has no benefit, and samples from 






' HUMORAL 






FACTORS 






^r^ 




Cellu 


br ^ 


r ' 

1 Mod- Cellular 


Erythrocytes 
Neutrophils 

Platelets 

v. > 




Hormones 



Figure 8.1. Etiological mechanisms in 
Raynaud's phenomenon. Both local and 
humoral factors have been implicated. 
CGPR, calcitonin gene-related peptide. 



75 



NONATHEROSCLEROTIC VASCULAR DISEASE 




nonaffected arterioles from patients with sys- 
temic sclerosis show normal endothelium- 
mediated vasodilatory responses. This suggests 
that this mechanism is unlikely to be a primary 
causative one. 



Erythrocytes 

Abnormalities of erythrocyte aggregation and 
deformability have been described in Raynaud's 
patients. 



Humoral Factors 

Cellular 

Platelets 

The potential for platelets to act as a significant 
pathological factor is high. Activated platelets 
have the ability to aggregate and cause mechan- 
ical blockage, or release mediators that are both 
prothrombotic and stimulate vasoconstriction. 
Similar to the effects seen on endothelial cells, 
increased expression of a 2 receptors on platelets 
in Raynaud's patients may provide a lower 
threshold for activation in the presence of sym- 
pathetic stimulation. Platelets from Raynaud's 
patients also appear to be hypersensitive to 5- 
hydroxytryptamine (5-HT) (serotonin) stimu- 
lation. An increase in activation of platelets is 
seen in Raynaud's, with augmented release of 
the mediators thromboxane A 2 and serotonin. 
Similar to endothelin-1, thromboxane A 2 is 
related to disease severity, primarily in second- 
ary disease. Administration of antagonists to 
this substance, however, does not appear to 
have a marked effect on disease expression or 
progression. 

The potential for platelets to be primary 
etiological agents is also called into question 
with the interesting observation that sufferers 
of Glanzmann's thrombasthenia, who display 
a complete lack of platelet aggregation, may still 
suffer attacks of Raynaud's syndrome. 



Neutrophils 

Although they are unlikely to be causative 
factors, circulating neutrophils may certainly 
have a propensity for causing damage. Trapping 
and activation in the setting of occluded 
vessels followed by ischemia and reperfusion 
lead to an escalation of the reactive response in 
the local vicinity of the arterioles. However, in 
primary Raynaud's, endothelial damage is not 
really a feature. In light of this fact, this mecha- 
nism may simply represent an augmenting 
effect. 



Hormonal 

Estrogen 

Certainly the large predilection for the female 
sex that Raynaud's displays is cause enough to 
consider an underlying hormonal basis. Differ- 
ent phases of the menstrual cycle are associated 
with varying responses of digital blood flow to 
cold stimuli, which certainly suggests that 
female sex hormones may have an etiological 
role. 



Clinical Factors 

Raynaud's is characterized classically by the 
following symptoms: 

• Initial pallor due to vasoconstriction 

• Cyanosis due to sluggish blood flow 

• Redness due to hyperemia 

This gives the classical picture of white, blue, 
and then red discoloration described by suf- 
ferers. This picture usually occurs in the pres- 
ence of cold or emotional stress. It may be 
accompanied by numbness and burning pain, 
which may be severe as the blood flow returns. 
The most common site to be affected is the 
fingers, with the toes involved to a lesser extent. 
More rarely, other extremities such as the nose, 
ears, tongue, or nipple may be affected. The 
attacks range from mild, with an asymptomatic 
picture when seen by the clinician, to more 
severe effects with necrosis and gangrene of 
peripheral tissues. In extreme cases, this may 
progress from superficial to deep structures and 
require amputation. 

In the setting of the presentation of 
Raynaud's as a primary complaint rather than 
on the background of known disease, investiga- 
tion into underlying causes is required. Up to 
5% of patients presenting with primary 
Raynaud's ultimately develop a connective 
tissue disorder. It does appear that there are 
notable differences in the demographics and 
presentation between primary and secondary 
disease, as outlined in Table 8.2. 




> 



76 



VASCULAR SURGERY 



Table 8.2. Clinical differences between primary and 
secondary Raynaud's 



Feature 


Primary 


Secondary 


Age at onset 


<30 


>30 


Digital gangrene 


Rare, superficial 


Common 


Nail fold capillaries 


Normal 


Large and 
tortuous 


Auto antibodies 


Negative or low 


Frequent 




titer 





History and examination should be directed 
at the exclusion of secondary causes of 
Raynaud's phenomenon. History should include 
questioning on symptoms of connective tissue 
diseases as well as details of drugs and exposure 
to toxic agents. A history of vibrating tool use, 
trauma, or positional triggering consistent with 
thoracic outlet syndrome is also helpful. 
Routine examination should not omit careful 
examination of the peripheral pulses and 
blood pressure (BP) in both arms, along with 
neck examination for the presence of a cervical 
rib. 

A further test that can be performed at the 
consultation is capillaroscopy of the nail fold, 
where capillary loops lie horizontal to the 
surface (Box 8.1). Other techniques for the 
measurement of peripheral blood flow such as 
analysis of digital systolic pressure during 
cooling or red blood cell velocity in nail fold 
capillaries have yet to find their way into clini- 
cal practice. 

A chest radiograph shows a cervical rib if 
it is present, and may also show changes con- 
sistent with connective tissue disease. Blood 
testing for autoantibodies or prothrombotic 
states can provide good indicators for the pres- 



Box 8.1. Capillaroscopy 

The nail-fold capillaries can be visualized 
through immersion oil placed on the finger. 
Although best seen with a microscope, they 
can also be viewed using an ophthalmoscope 
set at a diopter of 10-40. In the presence of 
connective tissue disease, the nail-fold 
vessels are tortuous and dilated. 



ence of underlying disease. Further tests such as 
Doppler imaging or angiography of vessels as 
well as further blood tests, should be instigated 
on the basis of previous indications. A potential 
pathway for diagnosis in Raynaud's is shown in 
Figure 8.2. 

As an idiopathic cause, primary Raynaud's 
should be considered a diagnosis of exclusion, 
to coincide with the following criteria: 

• Symmetrical vasospastic attacks 

• Absence of necrosis or gangrene 

• History and physical findings not indica- 
tive of secondary cause 

• Normal capillaroscopy of nail bed 

• Normal erythrocyte sedimentation rate 
(ESR) and negative serology for 
autoantibodies 

A follow-up period of 2 years is likely to be 
sufficient to pick up any underlying pathology 
of what initially appears to be Raynaud's 
disease. The most likely culprits by far that may 
reveal themselves are the connective tissue 
diseases. 



Treatment 

Acute Ischemia 

In the setting of acute tissue-threatening 
ischemia, instigation of vasodilatation is the key 
treatment. Intravenous prostaglandin analogues 
such as iloprost are effective. In the absence of 
the availability of such therapy, a short-acting 
calcium channel blocker such as nifedipine 
can be given, along with aspirin as antiplatelet 
therapy. A digital or wrist block with local anes- 
thesia without adrenaline may be beneficial 
both for pain relief and for the underlying 
pathology, as it effectively provides a localized 
chemical sympathectomy. 

In persistent cases, a prolonged infusion of 
IV heparin may be required, and in intract- 
able cases, surgical intervention with localized 
digital sympathectomy is indicated. 

In the absence of tissue-threatening compli- 
cations, disease therapy is best approached in a 
stepwise manner. Increasing levels of inter- 
vention can then be introduced if the disease 
proves refractory to a particular level of therapy. 
A potential pathway for the treatment of 
Raynaud's disease is shown in Figure 8.3. 



11 



NONATHEROSCLEROTIC VASCULAR DISEASE 




Figure 8.2. Algorithm for the 
diagnosis of Raynaud's 
phenomenon. 



History suggestive of 
Raynaud's phenomenon 



Exclusion of causative or exacerbating factors 

• Drugs, Toxins 

• Environmental agents. Occupation 

• Trauma 



ROUTINE TESTS 

Examination 
Capillaroscopy 

Blood tests: FBC, U+Es, Autoantibodies 
Chest radiograph 




In the presence of: 

• Abnormal capillaroscopy or CXR 

• Autoantibodies 



In the presence of: 

• Asymmetrical disease 

• Abnormal vascular examination 



Rheumatic disease likely 



May require 

• Doppler studies/angiography 



If all tests normal 



Low suspicion of 2 Raynaud's 

• No digitat lesions/gangrene 



High suspicion of 2 Raynaud's 

• Severe symptoms 

• Evidence of tissue damage/gangrene 



> These patients can be considered to have primary Raynaud's 
1 Infrequent follow-up for two years may be necessary to rule 
out developing CTD 



Further tests 

• Thyroid function 

• Cryoglobulins 

• Serum protein electrophoresis 




Nonpharmacological Therapy 

Initial treatment is based on the avoidance of 
precipitants, such as cold and emotional stress. 
Prevention of exposure to cold is best based on 
a total body approach, rather than just localized 
to fingers and toes, and may include specifically 
designed clothing such as heated gloves. The 
cessation of contributing factors is also involved 
at this step. This includes lifestyle modifications 
regarding smoking and caffeine intake, plus 
other vasoconstricting substances such as 
cocaine and amphetamines. Any medical thera- 
pies that may be contributing to the disease 
should be stopped or modified to other, less 



harmful agents. Use of vibration-proof impact 
tools or in certain cases a change of job may 
prevent the progression of secondary Raynaud's 
associated with hand-arm vibration syndrome. 

Pharmacological Therapy 

There are a multitude of putative pharmacolog- 
ical therapies for Raynaud's, some of which have 
a more solid evidence base than others: 

Ca + channel blockers: These have been the 
mainstay of treatment for some time. The 
best agents are those based on dihydropy- 
ridines, due to their decreased action on 




> 



78 



VASCULAR SURGERY 




Figure 8.3. Algorithm for the 
treatment of Raynaud's disease. 



Sympathectomy 
Proximal sympathectomy 
Digital artery sympathectomy 



cardiac muscle and relative selectivity 
for vascular smooth muscle. Classically, 
nifedipine is the drug of choice, but newer 
dihydropyridines such as amlodipine and 
felodipine are also effective. These are 
usually started at a low dose with gradual 
escalation as required. 

Angiotensin II receptor antagonists: Trials 
with losartan show a good efficacy com- 
pared with nifedipine in reduction of 
quantity and severity of attacks (Dziadzio 
et al., 1999). 

Serotonin modulation: The serotonin antago- 
nist Ketanserin has been used in treatment, 
particularly in the setting of scleroderma, 
although its evidence base may be ques- 
tionable. There is also some evidence that 
selective serotonin reuptake inhibitors 
(SSRIs) such as fluoxetine may have a 
beneficial effect. 



Nitroglycerines: The use of topical nitrates 
may aid in symptomatic relief and in the 
reduction of the number of attacks, 
although side effects may limit their 
usefulness. 

Prostaglandins: There is good evidence that 
an intravenous infusion of iloprost, a syn- 
thetic prostacyclin analogue, is efficacious 
in Raynaud's, certainly in severe disease. 
The key to its widespread use in less acute 
settings lies in the production of an effec- 
tive oral preparation. However, attempts to 
date to produce a similar effect to that seen 
with intravenous administration have 
proved equivocal and have been troubled 
by a high incidence of side effects. 

Future possibilities: Further medical treat- 
ments are likely to continue to appear, in 
the light of the large quantity of mooted 
causative factors. Initial trials with calci- 



79 



NONATHEROSCLEROTIC VASCULAR DISEASE 




tonin gene-related peptide certainly indi- 
cate potential for development. Further 
delineation of the genomics behind 
primary Raynaud's may show the road to 
more interventional possibilities in the 
setting of gene therapy. 

Surgical Therapy 

Due to its invasive nature as well as its inherent 
complications, surgery for Raynaud's is usually 
reserved for cases that are refractory to medical 
therapy. The main surgical option is sympa- 
thectomy, which may be performed as a proxi- 
mal procedure, or as a microsurgical localized 
operation: 

Upper limb: This involves destruction of the 
second and third thoracic ganglia and their 
interconnections, which maybe performed 
through a number of surgical approaches. 
Results are disappointing in terms of 
relapse rate, although it may still have a 
role in severe cases. 

Lower Limb: This involves destruction of 
the second, third and sometimes fourth 
lumbar sympathetic ganglia plus intercon- 
nections. It appears to be a more effective 
therapy than the similar procedure in the 
upper limb. 

Another option, this one microsurgical, is 
digital artery sympathectomy, a more effective 
procedure than proximal sympathectomy. 
Although effective, its place probably lies in 
treatment of tissue-threatening disease or in 
cases refractory to medical therapy. 



Other Vasospastic Disorders 

Erythromelalgia 

This is a rare condition characterized by vasodi- 
lation associated with erythema and increased 
temperature of the extremities along with a 
burning pain. Similarly to Raynaud's phenome- 
non, it may occur in an idiopathic form or 
secondary to other disease. Associated pathol- 
ogy includes hypertension, myeloproliferative 
disorders, diabetes, rheumatic disease such as 
rheumatoid arthritis or systemic lupus erythe- 
matosus (SLE), gout, spinal cord disease, and 
multiple sclerosis. 



The clinical picture varies in severity. Therapy 
with aspirin and vasoconstrictive agents can be 
helpful, and treatment of the underlying disease 
may aid symptoms in secondary disease. 

Acrocyanosis 

This presents as a symmetric cyanosis of the 
hands or less commonly of the feet, which is 
both persistent and painless. Affected body 
parts show decreased temperature and blue dis- 
coloration, and they may be swollen and sweaty. 
Examination of peripheral pulses is normal. 
Generally, it is not a harbinger of underlying or 
threatening pathology, and supportive treat- 
ment with reassurance and advice about 
avoiding the cold is usually sufficient. 

Vasculitis 

Vasculitis is broadly defined as inflammation of 
the vessel wall. It covers a wide range of clinical 
and pathological entities, which have both 
confusing similarities and yet important dis- 
tinctions, most notably in their treatment and 
prognosis. 

Classification of these diseases is compli- 
cated, as there is considerable overlap both in 
clinical presentation and histological appear- 
ance. The most common approach used is to 
classify them by consideration of the size of 
the vessels that the disease affects (Table 8.3). 
Vasculitis that affects the smaller arteries can be 
further classified according to whether or not 
antineutrophil cytoplasmic antibodies (ANCAs) 
are found. Such a distinction is possible at a his- 
tological level, as only those associated with 
ANCAs affect the small arteries, with the others 
affecting arterioles, venules, and capillaries. 

The underlying pathophysiology of many of 
the noninfective vasculitides appears to have an 
immunological basis. This is clear not only from 
the temporal associations of some of the con- 
ditions with certain viral infection, but also 
from the fact that the majority are treated with 
various combinations of immunosuppressants. 

Their clinical presentation arises from the 
consequences of vessel wall inflammation. 
These include thrombosis and subsequent 
ischemia or infarction, aneurysm formation, 
and hemorrhage. The extremely broad range of 
clinical phenomena and affected target vessels 




80 



VASCULAR SURGERY 



Table 8.3. Classification of the vasculitides 

Large-vessel vasculitis 
Giant cell arteritis 
Takayasu's arteritis 

Medium-vessel vasculitis 
Kawasaki's disease 
Polyarteritis nodosa 

Small-vessel vasculitis 
ANCA associated 
Wegener's granulomatosis 
Churg-Strauss syndrome 
Microscopic polyangiitis 

Not ANCA associated 
Henoch-Schbnlein purpura 
Cryoglobulinemic vasculitis 
Cutaneous leukocytoclastic angiitis 

ANCA,antineutrophil cytoplasmic antibody. 



means that the vasculitides present to a myriad 
of clinicians: dermatologists, rheumatologists, 
and gastrointestinal(GI) and vascular surgeons. 

Large-Vessel Vasculitis 

Giant Cell Arteritis 

This is a granulomatous inflammatory disease 
of the aorta and its branches. It has a tendency 
to affect the extracranial branches of the carotid 
artery, hence its other name of temporal arteri- 
tis. It was first described by Sir Jonathan 
Hutchinson in 1890, who noticed it in a retired 
hospital porter who was unable to wear his hat 
due to tender and inflamed temporal arteries. It 
is a disease that is predominantly seen in Euro- 
peans over 50 years old, and has a slightly higher 
incidence in women, with a male to female ratio 
of 1 :2. Incidence can be up to 25/100,000 in the 
older age groups. Although the actual cause 
remains a mystery, it has a strong association 
with polymyalgia rheumatica, which is present 
in up to 50% of cases. Both these conditions 
share genetic risk factors and geographical 
populations. 

At a microscopic level, there is intimal thick- 
ening and edema. A chronic inflammatory 
picture is seen with giant cell and granuloma 
formation. 

The usual clinical presentation is with severe 
unilateral occipital or temporal headaches. 
There is scalp tenderness, which classically 
becomes evident on brushing or combing hair. 



Features of vascular insufficiency may also be 
present, such as jaw claudication, or, more 
rarely, tongue or limb claudication. These symp- 
toms may be accompanied by systemic symp- 
toms of fever, malaise, fatigue, and a sore throat. 

The most feared complication is involve- 
ment of the ophthalmic artery. This leads to 
ischemia of the optic nerve and sudden painless 
visual loss, which can be temporary or may be 
permanent. 

On examination, there may be swelling and 
tenderness of affected superficial arteries, which 
will be pulseless. The most notable finding on 
simple blood tests is a significantly raised ESR 
and C-reactive protein (CRP). 

The key to diagnosis of this condition is 
obtaining histology from an arterial biopsy, 
which is usually taken from the temporal arter- 
ies. Due to the nature of the disease, which 
occurs as skip lesions, it is important to take a 
multiple sections, each of an amount of at least 
3 to 5 cm. 

The most effective treatment is corticos- 
teroids, and if a diagnosis of giant cell arteritis 
(GCA) is suspected, immediate initiation of 
steroid therapy is required to prevent the onset 
of blindness. A prompt biopsy should then be 
organized, preferably within 7 days of starting 
the steroid course. This reduces the possibility 
of a negative biopsy in an affected patient, 
avoiding the clinical conundrum that follows 
of a patient on long-term steroids without a 
definitive diagnosis. 

In 75% of patients, the disease settles within 
a period of 3 years, but in others it proves refrac- 
tory to treatment. As in all diseases treated with 
long-term steroids, the side effects of the treat- 
ment itself can lead to difficulties. Currently, 
however, it appears that no steroid-sparing 
agent has emerged as a suitable alternative or 
adjunct. 

Takayasu's Arteritis 

In 1908 Mikito Takayasu, a Japanese ophthal- 
mologist, described the association of retinal 
arteriovenous anastomoses and absent upper 
extremity pulses. Takayasu's arteritis is a rare 
granulomatous arteritis of the aorta and its 
major branches. It has a predilection for 
females, and tends to affect people in their 
second and third decades, although it has been 
reported in patients as young as 6 months and 



81 



NONATHEROSCLEROTIC VASCULAR DISEASE 




in adults at every age. Although it is encoun- 
tered worldwide, it is especially prevalent in 
people of Asian descent. 

Pathologically, Takayasu's arteritis somewhat 
resembles giant cell arteritis, displaying severe 
necrotizing inflammation that leads to marked 
intimal thickening. This causes eventual nar- 
rowing and occlusion of the lumen. It can also 
weaken the vessel wall, providing a predisposi- 
tion to aneurysm formation. 

Clinically, patients suffer from systemic 
symptoms such as fever, malaise, weight loss, 
arthralgia or myalgia, and night sweats. More 
specific symptoms are related to the affected 
arterial system and related end-organ ischemia, 
(Table 8.4). Clinical presentation can vary from 
incidental discovery in the asymptomatic pa- 
tient to a catastrophic event. 

Physical examination may reveal bruits or 
decreased pulses (Takayasu's is also known as 
pulseless arteritis). Blood pressure may be ele- 
vated, and also asymmetrical due to subclavian 
stenosis. There may be valvular regurgitation 
and arterial tenderness. This latter sign may be 
especially noticeable over the carotids. 

Investigation is primarily by arteriography. 
Neither the level of symptoms nor other indica- 
tors of disease such as the ESR provide a clear 
indication of disease extent or severity. As a rule, 
these indicators are thus not a good guide to 
management. 



Treatment 

Some patients demonstrate a self-limiting 
disease that requires minimal intervention, 
whereas others require more intensive therapy. 

Medical 

In the first instance, treatment is with glucocor- 
ticoids. If the disease proves refractory to 
steroid therapy, or if the patient is unable to be 
weaned off their long-term use, further treat- 
ment with cytotoxic agents is indicated. 
Adjunct therapy with methotrexate is effective 
in increasing progression to remission as well a 
having steroid-sparing effects. Cyclophos- 
phamide has similar properties, but due to its 
significant toxicity is probably best used as a 
third-line treatment in those who are intolerant 
of or nonresponsive to methotrexate. 

Surgical 

There is a place for open vascular surgery and 
endovascular options in the cure of fixed vascu- 
lar lesions producing significant ischemia. Indi- 
cations include cerebral hypoperfusion, 
hypertension due to renovascular disease, and 
aneurysms or valvular insufficiency. Clinically 
significant improvement of symptoms is usually 
possible with surgery, although restenosis is 
common. 



Table 8.4. Site-dependent presentation of Takayasu's arteritis Prognosis 



Clinical presentation 


and severity depend on: 


Site 




Degree of stenosis 




Availability of collateral blood supply 


Subclavian: 


Arm claudication, Raynaud's 


Carotids: 


Visual changes, syncopeJIAs/ 




stroke 


Vertebrals: 


Visual changes, dizziness 


Aortic arch: 


Aortic insufficiency, CHF 


Aorta 




(includes main enteric branches): 




Abdominal pain, nausea, 




vomiting 


Renal arteries: 


Hypertension/renal failure 


Iliac arteries: 


Leg claudication 


Pulmonary arteries: 


Atypical chest pain, dyspnea 


Coronary arteries: 


Chest pain/Ml 



CHF, chronic heart failure; Ml, myocardial infarction; TIA, transient 
ischemic attack. 



Takayasu's is a chronic disease with a 45% 
relapse rate, although 23% of patients never 
reach a state of remission. Following diagnosis, 
survival rates are greater than 80%. However, 
the disease still carries a significant morbidity. 

Cogan's Syndrome 

This rare disorder of young adults of either sex 
warrants a mention at this point primarily due 
to the vessel it affects. It is an inflammatory 
process that leads to interstitial keratitis and 
may lead to permanent hearing damage or 
effects on the vestibulo auditory system, 
although these effects appear unrelated to the 
vasculitic phenomena. From a vascular point 
of view, patients may suffer aortitis or other 
large-vessel inflammation. The primary source 




> 



82 



VASCULAR SURGERY 



of morbidity is cardiovascular involvement and 
auditory problems. 

Treatment is with prednisolone, and is indi- 
cated for severe ocular or auditory disease and 
vasculitis. Prompt treatment at the first sign of 
hearing damage improves the prognosis for 
overall hearing loss and return of function. 

Medium-Vessel Vasculitis 

Kawasaki's Disease 

Kawasaki's disease was first identified in 1967 by 
Tomisahu Kawasaki, a Japanese pediatrician, 
and is also called mucocutaneous lymph node 
syndrome. Although it is seen in adults, this is 
primarily a disease of childhood, and the major 
cause of acquired heart disease in children 
in the United States and Japan. Despite the 
original demographical description, it is a 
disease that is found worldwide. In the United 
Kingdom the incidence is less than 5/100,000 in 
children less than 5 years old; 80% of sufferers 
are in this age bracket, and males are affected 
with slightly greater frequency than females at 
a ratio of 2 : 1. 

Clinical Factors 

The condition begins with an acute febrile 
illness. This is followed by a polymorphic rash 
that may affect any part of the body, congestion 
of the conjunctivae, dryness and erythema of 
the oral mucosa, cervical lymphadenopathy, and 
erythematous edema of the palms and soles. 
These features are the criteria for the diagnosis 
of Kawasaki disease (Table 8.5). Other clinical 
features are outlined in Table 8.6. 

The major cause of morbidity and mortality 
in the disease arises from the cardiovascular 
complications, including pancarditis, but more 
specifically the vasculitic changes in the coro- 
nary arteries may lead to the formation of 



Table 8.5. Diagnostic criteria for Kawasaki's disease 

Fever plus: 
Rash 

Conjunctival injection 
Oral mucosal changes 
Brawny induration of extremities 
Cervical lymphadenopathy 



Table 8.6. Other clinical features of Kawasaki's disease 
System Feature 

Cardiovascular Pancarditis 

Gastrointestinal Diarrhea 

Genitourinary Albuminuria 

Neurological Aseptic meningitis 

Joints Arthralgia 

Blood Leukocytosis, thrombocytosis, raised 
C-reactive protein 



aneurysms. These occur 1 to 4 weeks after the 
onset of fever, and are seen in up to 25% of 
untreated cases. Thus, monitoring of the disease 
is usually structured with an electrocardiogram 
(ECG) performed in the first week of illness, 
with an echocardiogram both initially at diag- 
nosis and repeated 2 to 4 weeks later. 

Treatment is twofold. High-dose IV 
immunoglobulin is given as a single infusion. 
This prevents the formation of coronary 
aneurysms but also decreases fever and 
inflammation in the myocardium. Patients are 
also given aspirin as an antithrombotic 
measure. In certain cases, long-term follow-up 
and anticoagulation maybe required. 

Polyarteritis Nodosa 

Polyarteritis nodosa (PAN) was the name even- 
tually given to the disease process in the first 
definitive report of a patient with necrotizing 
arteritis, which was written in 1866. For some 
time, before it was clear that there were fur- 
ther underlying pathological mechanisms, all 
patients presenting with necrotizing arteritis 
were given a diagnosis of PAN. In truth, it is an 
uncommon disorder. Unlike other vasculitic 
disorders, PAN is most frequently observed in 
middle-aged men. It often leads to severe sys- 
temic complications affecting many of the organ 
systems. The underlying pathology of PAN is 
one of florid acute inflammatory changes seen 
in a pattern of fibrinoid necrosis. This is similar 
to what is seen in models of immune complex 
vasculitis, and there is some evidence that 
certain viruses have associations with the devel- 
opment of PAN. These include the hepatitis B 
and C viruses and HIV. Clinically, the patient 
may present with a number of complaints, due 
to the widespread nature of the disease (Table 
8.7). 



83 



NONATHEROSCLEROTIC VASCULAR DISEASE 




Table 8.7. Clinical manifestations of polyarteritis nodosa 





System 


Manifestation 




Cardiovascular 


Coronary arteritis may lead to Ml or 
HF 




Respiratory 


Asthmatic symptoms + hemoptysis, 
(rare) 




Gastrointestinal 


Hemorrhage and mucosal ulceration 
Presentation may mimic the acute 
abdomen 




Genitourinary 


Hematuria 




Neurological 


Mononeuritis multiplex 
(involvement of vasa nervorum) 




Joints 


Subcutaneous nodules,* 

hemorrhage, and gangrene 
Livedo reticularis, (seen if chronic) 




Blood 


Leukocytosis, thrombocytosis, raised 
CRP 



* These occur due to the involvement of subcutaneous arteries and, 
although uncommon, they are part of the reason why the disease 
was originally named. 

CRP, C-reactive protein; HF, heart failure; Ml, myocardial infarction; 
TIA, transient ischemic attack. 



Blood tests in PAN reveal anemia, leukocyto- 
sis, and a raised ESR; ANCA is rarely positive 
in classic PAN. Diagnosis is primarily by 
two methods. Histologically, biopsied lesions 
show a classical pattern of necrotizing arteritis, 
whereas angiography reveals microaneurysms, 
which may be found in hepatic, intestinal, or 
renal vessels. 

Treatment of the condition is primarily with 
immunosuppressants. Therapy is initially with 
corticosteroids, but further agents such as 
azathioprine or cyclophosphamide maybe indi- 
cated in life-threatening disease. 

In cases where the vasculitis occurs in a 
picture of viral infection, control of the 
inflammation with immunosuppression may be 
necessary before effective antiviral therapy can 
be considered. However, combination antiviral 
therapy may prevent the problems of conven- 
tional treatment, which may enhance viral repli- 
cation, increasing the chance of progression to 
chronic infection. In viral-associated vasculitis, 
plasma exchanges may also be considered, as 
they are effective in reducing the amount of 
circulating immune complexes. 

The prognosis, perhaps unsurprisingly, is 
worse in those patients who suffer multisystem 



involvement. Five-year survival with treatment 
is 80%, and it is a disease that relapses only 
infrequently. 

Small-Vessel Vasculitis 

Estimates of incidence of the small-vessel vas- 
culitides vary, most likely due to variations in 
the diagnosis. They share similar nonspecific 
systemic features such as fever, malaise, weight 
loss, arthralgia, and myalgia. Investigation of 
these patients should include screening for anti- 
neutrophil cytoplasmic antibodies (Box 8.2). 

Broadly speaking, the vasculitides of the 
small vessels can be divided into those in which 
the ANCAs are positive or negative (Table 8.8). 

Wegener's Granulomatosis 

In Wegener's (named for Friedrich Wegener, 
a German pathologist, 1907-1990), the small 
arteries are predominantly affected above all 
others. It may occur at any age, and appears to 
show no predilection for either sex (Hoffman 
et al, 1992). 

Pathologically, it has a pattern of necrotizing 
granulomatous vasculitis and is typically char- 
acterized by lesions that involve three main 



Box 8.2. Antineutrophil 
cytoplasmic antibodies 






Antineutrophil cytoplasmic antibodies 
(ANCA) 

There are two primary types of ANCA, 
differentiated on the basis of their pattern 
of immunofluorescence on ethanol-fixed 
neutrophils. 

cANCA 

Antibodies directed against serine pro- 
tease proteinase 3 (PR3). 

These give a cytoplasmic pattern. 

pANCA 

Antibodies directed against the enzyme 
myeloperoxidase (MPO). 

These give a perinuclear pattern. 

The ANCA typing can be combined with 
antigen-specific testing for PR3 and MPO to 
increase specificity. 




> 



84 



VASCULAR SURGERY 



Table 8.8. Distribution of ANCA among the small vessel 
vasculitides 




ANCA, antineutrophil cytoplasmic antibody. 



organ systems: the upper respiratory tract, the 
lungs, and the kidneys. Greater than 90% of 
patients seek help for symptoms related to the 
respiratory tract. 

Patients suffer initially from severe rhinor- 
rhea. This is followed by nasal and sinus 
inflammation, which eventually leads to carti- 
laginous ischemia, the sequelae of which are 
perforation of the nasal septum and saddlenose 
deformity of the nasal bridge. Tracheal 
inflammation and sclerosis lead to stridor and 
potentially dangerous airway stenosis. This par- 
ticular complication is more common in chil- 
dren who are affected. Disease activity in the 
lower respiratory tract leads to cough, hemopt- 
ysis, and pleuritic pain. Renal tract disease man- 
ifests as glomerulonephritis. This is present in 
only 20% of patients at the time of diagnosis, 
but appears overall in 80% eventually. 

A chest radiograph may show nodular 
masses, which can be multiple. There may also 
be ground-glass infiltration and cavitation. Over 
time, the lesions show a migratory pattern, dis- 
appearing in one area only for new lesions to 
appear in others. 

The typical histology of Wegener's is best 
seen in the kidney. A renal biopsy demonstrates 
necrotizing microvascular glomerulonephritis. 
However, diagnosis is often by nonrenal biopsy, 
which demonstrates the granulomatous necro- 
tizing inflammation with neutrophil aggrega- 
tion and vasculitis. The site of biopsy is 
important. An upper airway biopsy, although 
easy to perform, demonstrates diagnostic 
changes in only 20% of cases. A more invasive 
biopsy of the lung parenchyma shows the diag- 
nosis in 90%. 

Before treatment became available, patients 
suffering from Wegener's granulomatosis would 
have a mean survival time of only 5 months 



before succumbing to pulmonary or renal 
failure. Even with treatment, relapse is seen in 
around 50% of cases. The presence of lung or 
kidney involvement at diagnosis is a poor 
prognostic indicator. Wegener's also has an 
especially mutilating form, known as midline 
granuloma, which carries a particularly poor 
outlook. 

Therapy is with a combination of glucocorti- 
coids and other immunosuppressants. Treat- 
ment with prednisolone alone is ineffective, 
and the decision on which further agent to use 
is based on a number of factors. In active 
Wegener's where the disease is life threatening, 
treatment with prednisolone is combined with 
cyclophosphamide. This leads to a 90% 
improvement rate, achieving complete remis- 
sion in around 75% and giving a survival rate 
of 80%. The main drawback with this regimen 
is the significant toxicity associated with 
cyclophosphamide use. To avoid it, a progressive 
approach may be needed, using cyclophos- 
phamide until the disease is in remission, then 
changing to another agent such as methotrexate 
or azathioprine, which can then be tapered out. 
In active disease that is not life threatening, 
combination treatment with prednisolone and 
methotrexate is equally as good at maintain- 
ing remission compared with a cyclophos- 
phamide, and a similar regimen may also be 
used in those patients who are intolerant of 
cyclophosphamide. 

Churg-Strauss Syndrome 

Churg-Strauss syndrome, named for Jakob 
Churg, a Polish-American pathologist (1910- 
1966) and Lotte Strauss, an American pathol- 
ogist (1913-1985), has an incidence of about 
3 per million. The typical patient is usually 
male and in the fifth decade of life. Classically 
the syndrome is a triad of rhinitis and asthma, 
eosinophilia, and systemic vasculitis. Because 
of its makeup, there are a number of theories 
about the underlying pathophysiology. It may 
be a progression of an allergic phenomenon, 
or a primary vasculitis that has an asthmatic 
component due to the involvement of 
eosinophils. 

Pathologically, the picture is one of 
eosinophilic infiltration, which typically affects 
the lung, peripheral nerves, and the skin. Histo- 
logically, a necrotizing vasculitis of the small 



85 



NONATHEROSCLEROTIC VASCULAR DISEASE 




arteries is seen along with extravascular, 
allergic granulomas. 

Although initially presenting as above, the 
syndrome has a progressive nature, which is 
eventually vasculitic and involves the lungs, 
nerves, heart, and the GI and renal tracts. A 
complete blood count shows a high eosinophil 
count. 

Treatment is primarily with corticosteroids 
alone, although in life-threatening cases 
cyclophosphamide can be added. Due to its 
association, it may be difficult to taper pred- 
nisolone treatment when the vasculitic parts of 
the disease are in remission, as this may lead to 
asthmatic exacerbations. 



Microscopic Polyangiitis 

Another vasculitis that primarily affects the 
small vessels, microscopic polyangiitis has only 
recently been distinguished from PAN, and for 
this reason there is a relative dearth of data 
about the disease. Pathological distinction from 
PAN is accepted to be on the basis that micro- 
scopic polyangiitis leads to vasculitis in vessels 
smaller than arteries, whereas PAN can be diag- 
nosed if these vessels are not involved. Histo- 
logically, it represents a necrotizing vasculitis, 
with few immune deposits and no granuloma 
formation. 

Clinically, microscopic polyangiitis mainly 
affects the kidney, presenting with glomeru- 
lonephritis and hemoptysis. It may also be asso- 
ciated with mononeuritis multiplex and fever. 
Diagnosis is usually based on clinical features, 
testing for ANCAs, and renal biopsy. It can be 
severe, and survival rates at 5 years may be as 
low as 74%. In disease that is potentially life 
threatening, treatment is with prednisolone and 
cyclophosphamide. 

Non-Antineutrophil Cytoplasmic 
Antibody-Associated Small-Vessel 
Vasculitis 

Henoch-Schonlein Purpura 

This is the most common systemic vasculitis in 
children. It is named for Edouard Henoch, a 
German pediatrician (1820-1910), and Johann 
Schonlein, a German physician (1793-1864). 
The peak incidence is at 5 years. Pathologically, 



it is characterized by the deposition in vascular 
structures of immunoglobulin A (IgA)-contain- 
ing immune complexes. It has a preference for 
venules, capillaries, and arterioles. The common 
clinical presentation is usually with arthralgia, 
colicky abdominal pain, and a purpuric rash; 
50% of sufferers have hematuria or proteinuria. 
However, this compromises renal function in 
only 15%. 

It is important to distinguish Henoch- 
Schonlein as a distinct pathological entity from 
other small-vessel vasculitides. It has an excel- 
lent prognosis, and supportive care is usually 
sufficient for most patients, in contrast to the 
life-threatening disease that can be caused by 
the other inflammatory disorders. End-stage 
renal failure occurs in less than 5% of patients. 

Cutaneous Leukocytoclastic Angiitis 

This is the most common cutaneous vasculitic 
lesion. It is an acute purpuric skin lesion, under- 
lying which is an inflammation of the dermal 
postcapillary venules. Although primarily 
affecting the skin, it may also be associated with 
an arthralgia or glomerulonephritis. Etiologi- 
cally, there is some indication that viral agents 
such as hepatitis C virus may play a part. It 
can also result from drug therapy with certain 
agents such as sulfonamides or penicillin. 

Most patients suffer a single episode, with 
resolution occurring over a few months. About 
10% suffer recurrent disease at varying inter- 
vals. Symptomatic relief from cutaneous irrita- 
tion and associated arthralgia and myalgia is 
possible with antihistamines and nonsteroidal 
antiinflammatory drugs. Treatment of severe 
cutaneous disease is with corticosteroids. 

Cryoglobulinemic Vasculitis 

Cryoglobulins are cold-precipitable immuno- 
globulins that maybe monoclonal or polyclonal. 
They are encountered in myriad disease pic- 
tures including lymphoid and plasma cell neo- 
plasms and chronic infective and inflammatory 
processes. Essential mixed cryoglobulinemia 
describes the presence of cryoglobulins in the 
absence of any precipitating underlying pathol- 
ogy. Although this has been described his- 
torically, it appears that most of these cases 
are related to infection with hepatitis C virus 
(HCV). 




> 



86 



VASCULAR SURGERY 



Vasculitis occurring in association with cryo- 
globulins affects people at about 50 years of age. 
A clinical picture of palpable purpuric lesions, 
arthritis, weakness, neuropathy, and glomeru- 
lonephritis is seen. The latter complication has 
an association with a poor prognosis, if not 
always a progression to end-stage renal failure. 
A useful diagnostic test is to look at complement 
levels. A distinctive picture of low levels of C4 
with normal or slightly low C3 is seen. Renal 
biopsy shows an endothelial membranoprolifer- 
ative glomerulonephritis with intraglomerular 
deposits. 

Mild disease can be approached symptomati- 
cally Antiviral therapy that produces a sus- 
tained response appears to be the most effective 
way of treating HCV-associated cryoglobuline- 
mic vasculitis. In severe disease, the use of 
immunosuppressants has been advocated, 
although as expected, these agents may lead to 
an increase in HCV viremia. 



Miscellaneous Disorders 

Buerger's Disease 

This eponymous disease, named for Leo 
Buerger, an Austrian-born American physician 
and urologist (1879-1943), is also known by the 
more descriptive term thromboangiitis obliter- 
ans. It is an occlusive disease of the small and 
medium-size arteries of the extremities, which 
it affects in a segmental fashion. It primarily 
affects young people, with a median age of 34 
years. Initial descriptions by Buerger noted a 
strong predilection of the disease for men, 
affecting 2 of 500 cases that he reported. More 
recent studies report a male to female ratio of 
up to 5:1. The disease also has a somewhat 
unusual geographical distribution. It has a low 
incidence in people of European descent, but is 
high in India, Japan, Korea, and the Middle East, 
especially among Jews of Ashkenazi descent. In 
fact, in this latter micropopulation, Buerger's 
disease accounts for up to 80% of cases of 
peripheral vascular disease. 

The primary causative factor associated with 
Buerger's disease is tobacco usage. Although 
this usually manifests as smoking, it has also 
been observed in patients who chew tobacco. 
Tobacco use holds the key not only to causation 
but also to persistence, progression, and recur- 



rence. This much is clear from observations of 
the patient base, although interestingly the 
actual underlying mechanism has proved 
elusive. The indications appear to be that of an 
immune-mediated pathology, and a high preva- 
lence has been associated with certain tobacco 
types in India and Bangladesh. Histologically, 
the disease manifests as an intense endarteritis 
with cellular infiltration, giant cell foci, and the 
presence of cell rich thrombi, which may show 
recanalization. Other immunological markers 
have also been demonstrated within the intima 
of the vessels. The internal elastic lamina and 
overall vascular wall architecture are well 
preserved. 

Clinically, there is a picture of ischemia of the 
distal extremities with rest pain, ulceration, and 
gangrene (Ohta and Shionoya, 1988). Both 
upper limb and lower limb disease is seen in 
40% of cases. In 10% there is only upper limb 
involvement, and in 50% only the lower limb is 
affected. Claudication is a rare symptom due to 
the distal nature of the diseases, and if present 
occurs most notably on the instep. 

Diagnosis is based both on typical clinical 
features as well as the exclusion of other causes, 
as outlined in Table 8.9. 

The investigations are thus directed initially 
at ruling out other conditions. The next step is 
to image the affected vessels. Arteriograms of 
the upper limb often show occluded radial and 
ulnar arteries that have become tortuous where 
they have recanalized. There may also be 
"pruning" of the digital arteries. In the lower 
limb, the vessels are often normal down to the 
popliteal trifurcation. Distal to this, segmental 
occlusive disease is seen. This frequently 
affects the tibial branches more than the 
popliteal. None of these changes, however, is 
pathognomonic. 

Although the treatment of infected lesions 
with antibiotics and debridement where neces- 
sary is similar to that of other ischemic disease, 
the mainstay of therapy in Buerger's disease is 
the cessation of smoking. Recurrence is almost 
always associated with resumption of tobacco 
usage, and new ischemic lesions are rare 
without reexposure. Conversely, persistent 
abstinence from tobacco results in quiescence of 
the disease process. As the lesions in Buerger's 
disease appear to have healing potential, this 
intervention can have a significant clinical 
effect. 



87 



NONATHEROSCLEROTIC VASCULAR DISEASE 




Table 8.9. Features of Buerger's disease 

1. Distal extremity ischaemic symptoms 

2. Exclusion of other causes 

Proximal embolic source 
Localised lesion (popliteal entrapment, cystic 
disease 
° Vasculitis 
Drugs — ergot 
Hypercoagulable states 
Trauma 

3. Onset at <45yrs 

4. Tobacco usage 

5. Supporting features 

Migratory superficial phlebitis 
Raynaud's phenomenon 
Upper limb involvement 

Distal extremity ischemic symptoms 
Exclusion of other causes 

Proximal embolic source 

Localized lesion (popliteal entrapment, cystic 
disease) 

Vasculitis 

Drugs: ergot 

Hypercoagulable states 

Trauma 

Onset at <45 years of age 
Tobacco usage 
Supporting features 

Migratory superficial phlebitis 

Raynaud's phenomenon 

Upper limb involvement 



Other options are used in refractory progres- 
sive disease. Sympathectomy has been advo- 
cated, and surgical bypass may occasionally be 
attempted. This latter treatment, however, has 
a poor rate of success. The results are affected 
not only due to the segmental, distal nature of 
the disease, but also because the associated 
phlebitis makes the veins poor conduits. 

A number of new treatments are being eval- 
uated, including implantation of spinal cord 
stimulators and intramuscular injections of 
vascular endothelial growth factor. The use of 
iloprost has also been advocated, although a 
significant clinical effect seems to be absent. 

The final surgical option is amputation. It 
should be noted that healing of surgical ampu- 
tation is much improved when patients are free 
from tobacco usage. 

Compared with other necrotizing arteritides, 
Buerger's disease has a worse prognosis with 



regard to limb loss. Some form of amputation, 
ranging from digital to major lower limb 
removal, is required in up to 20% of patients. 
The extreme importance of smoking is again 
evident here. In one study, 94% of patients who 
quit smoking remained amputation free, 
whereas 43% of those who continued eventually 
required at least one amputation. Interestingly, 
and perhaps due to the distal nature of the 
vasculature affected, the long-term survival of 
patients with Buerger's disease is affected only 
very slightly, with life expectancies approaching 
that of the normal population. 

Fibromuscular Dysplasia 

This is a noninflammatory, nonatherosclerotic 
vascular disease affecting arteries of small and 
medium size. It appears in a variety of histol- 
ogical manifestations depending on the arterial 
wall layer that is most significantly involved 
(Table 8.10). Although different types show a 
slight variation epidemiologically, the common- 
est form has a predilection for females in their 
second to fourth decades. Disease has been 
noted in patients as young as 2 months old. 

The etiology of fibromuscular dysplasia 
(FMD) is still unclear, but it appears to be mul- 
tifactorial. Suggested factors include localized 
ischemia due to poor perfusion from the vasa 
vasorum, sequelae of mechanical damage, and 
there is evidence for a familial link. There are 
also many documented associations, both with 
diseases such as pheochromocytoma, neurofi- 
bromatosis, Ehlers-Danlos, Alport's, Rubella 
syndrome, and heterozygous OCj-antitrypsin 
deficiency as well as other factors such as 
ergotamine and methysergide. 

Symptomatic disease from FMD may result 
from vessel stenosis, dissection, embolization, 
thrombosis, or rupture of associated ane- 
urysms. Disease and thus symptoms have been 
recorded across the whole spectrum of the 



Table 8.10. Histological classification of fibromuscular 
dysplasia 



Classification 

Intimal fibroplasia 
Medial fibroplasias 
Medial hyperplasia 
Perimedial fibroplasia 



Frequency (%) 

5 

1-2 
80-90 
10-15 




> 



88 



VASCULAR SURGERY 



vascular bed, and it is likely that there will be 
subclinical disease present in other areas in 
most patients. The most common manifesta- 
tions are hypertension from renovascular 
disease and stroke from carotid disease. In 
terms of the kidney, a significant proportion of 
renovascular disease arises on a background of 
FMD, the primary result of which is hyperten- 
sion. However, as renovascular disease is a 
relatively uncommon cause of hypertension in 
itself, FMD is not a major contributing factor to 
adult hypertension. When it affects the cere- 
brovascular system, it is predominantly seen in 
the internal carotid artery, and frequently is 
seen bilaterally. Vertebral artery involvement 
also occurs, although rarely in isolation from 
carotid disease. There is also a reported 
association between cerebrovascular FMD and 
intracranial "berry" aneurysms of an order of 
around 7%. The symptoms of cerebrovascular 
disease depend on the location and severity of 
the lesions. 

Where visceral vessels are affected, symptoms 
are usually due to ischemia. Infarction is rare 
due to the collateral arterial supply. In the 
peripheral vascular tree, lesions at all levels have 
been identified. The commonest sites are the 
subclavian artery in the upper limb and the 
external iliac artery in the lower limb. 

Investigations are directed at ruling out other 
causes of disease, but ultimately are based on 
imaging of the affected arterial system. As a 
noninflammatory process, FMD is not associ- 
ated with any of the acute-phase changes seen 
in the vasculitides, which can be detected by 
simple blood tests. 

Imaging can be either by noninvasive 
methods such as duplex scanning or magnetic 
resonance angiography (MRA), or by angio- 
graphy. The angiographic appearance of the 
most common histological type of FMD is of a 
string of beads. If duplex scanning is used to 
image the carotid system, it is important to 
image as distally as possible in order not to miss 
FMD, which in contrast to atherosclerotic 
lesions is not commonly seen around the 
carotid bifurcation. 

Therapeutic intervention in FMD should be 
considered on an individual basis and depend 
on the individual as well as the location and 
severity of the disease. In renovascular disease, 
initial medical therapy for hypertension is 
usually indicated. It should be remembered 



that as with other reno -occlusive disease 
angiotensin-converting enzyme (ACE) inhi- 
bitors should be avoided. If pharmacological 
treatment fails, there are a number of options. 
Percutaneous transluminal angioplasty (PTA) is 
an effective intervention, and although it is 
associated with up to a one in five re-stenosis 
rate, the rate of reappearance of hypertension is 
lower. Given the success in actually curing 
hypertension, PTA should probably be consid- 
ered earlier in younger patients to avoid unnec- 
essarily long courses of medical therapy along 
with progressive deterioration of renal func- 
tion. There are also a number of surgical 
options, which carry a higher morbidity and 
mortality. However, if considered in the appro- 
priate setting both in terms of patient suitabil- 
ity and available expertise, they can be very 
effective. 

Management of cerebrovascular FMD 
depends on the presence or absence of symp- 
toms to a certain extent. However, there is not a 
great deal of information on the progression of 
the disease with which to make this judgment. 
In the asymptomatic setting, medical therapy is 
appropriate. In the presence of symptoms, intra- 
operative or percutaneous dilatation of lesions 
can be considered. 

Where the visceral vessels are affected, surgi- 
cal treatment may be indicated when medical 
therapy fails. Treatment of peripheral disease 
can broadly be considered as medical therapy, 
dilation therapy, either percutaneously or by 
open technique, and surgical bypass or 
reconstruction. 

Cystic Adventitial Disease 

This is a rare, nonatherosclerotic cause of limb 
claudication. Since being first reported in 1947, 
there have been less than 350 documented cases. 
It affects people mostly in the fourth and fifth 
decades, with a male to female ratio of 5: 1. 

Pathologically, it is defined by synovial- 
like cysts in the adventitial layer of the arterial 
wall. These contain a mucinous gel that 
contains various proportions of mucoproteins, 
mucopolysaccharides, hyaluronic acid, and 
hydroxyproline. Although the exact underlying 
etiology is unclear, a number of mechanisms 
have been proposed. It has been suggested 
that the fluid may come from the reactivation 
of mesenchymal cells trapped in developing 



89 



NONATHEROSCLEROTIC VASCULAR DISEASE 




vessels during embryological vessels. Another, 
more widely supported hypothesis is that the 
cysts develop from a herniation of adjacent 
synovium. This latter theory is backed up by 
imaging results and by the late age of onset of 
disease. 

From the reported cases, cystic adventitial 
disease has a predilection for the popliteal 
artery above all others, but has also been 
described in the external iliac, axillary, and 
distal brachial, radial, and ulnar arteries. In the 
acute setting the picture is of sudden onset and 
progressive claudication. Physical examination 
may reveal a mass. Indeed, some of the cases are 
discovered in patients referred for investiga- 
tions of a soft tissue mass, before the emergence 
of significant vascular symptoms. 

The mainstay of investigation is imaging. 
Although the main accepted method is angiog- 
raphy, good diagnostic results can also be 
obtained from less invasive techniques. Duplex 
ultrasonography can reveal the cystic disease 
itself, which appears as anechoic or hypoechoic 
masses in the external wall. It also has the 
advantage of providing information as to the 
degree of vascular impairment. Magnetic reso- 
nance imaging also produces characteristic 



images, demonstrating multiple intramural 
cystic masses oriented along the long axis of the 
vessel. 

There are numerous therapeutic options. 
Spontaneous resolution of symptoms with 
conservative management has been docu- 
mented. The cysts can also be drained percuta- 
neously under computed tomography or 
ultrasound guidance. Surgical intervention con- 
sists of either excision of the cysts and diseased 
wall with preservation of the artery or use of an 
interposition graft. Both of these techniques 
have good initial results, although long-term 
follow-up data are lacking. 

Given the paucity of cases on which to base 
interventional judgment, the decision is pro- 
bably best made by considering the clinical 
urgency and available expertise. 



References 



Dziadzio M, Denton CP, Smith R, et al. (1999) Arthritis 

Rheum 42:2646-55. 
Hoffman GS, Kerr GS, Leavitt RY, et al. (1992) Ann Intern 

Med 116:488-98. 
Ohta T, Shionoya S. (1988) Br J Surg 75:259-62. 



9 



Lower Limb Ischemia 

Rajabrata Sarkar and Alun H. Davies 




Lower limb ischemia is an increasingly preva- 
lent disorder that has a wide range of clinical 
presentations and variable consequences for 
the patient. Although atherosclerosis is by far 
the most common cause of lower extremity 
ischemia, a variety of other conditions can cause 
either acute or chronic lower extremity 
ischemia. Three major factors are contributing 
to an increase in both the prevalence and inci- 
dence of lower extremity ischemia. The first 
is the general aging of the population in devel- 
oped countries, with its attendant increase in 
the prevalence of atherosclerosis, peripheral 
aneurysms, and other vascular lesions associ- 
ated with advanced age. The second factor is the 
alarming increase in the incidence of diabetes, 
particularly among adolescents and younger 
adults. As diabetes accelerates the progression of 
atherosclerosis and lower extremity ischemia, 
we can anticipate further increases in the 
number of patients presenting at a younger age 
with lower extremity ischemia. The third factor 
is the increasing numbers of patients who have 
undergone prior peripheral arterial bypass 
surgery and are potentially at risk for either 
graft occlusion or progression of disease. At 
many major medical centers the majority of 
patients presenting with acute limb ischemia 
are those with thrombosis of a prior lower 
extremity arterial reconstruction. This chapter 
reviews the causes, clinical presentations, diag- 
nostic approach, treatment options, and out- 
comes of chronic and acute lower extremity 
ischemia. 



Etiology and Presentation 

Peripheral arterial occlusive disease due to ath- 
erosclerosis is the most common cause of lower 
extremity ischemia in developed countries, with 
3% to 6% of the population over the age of 65 
suffering from symptomatic disease. The clini- 
cal presentation of long-standing ischemia can 
be variable, with symptoms ranging from inter- 
mittent claudication to rest pain, arterial ulcers, 
and frank gangrene. The classic progression of 
symptoms in atherosclerotic lower extremity 
ischemia is (1) decreased pulses without any 
symptoms, (2) intermittent claudication, (3) rest 
pain, and (4) arterial ulceration or gangrene 
(Fig. 9.1). Patients with limited ambulation due 
to other causes (e.g., stroke, musculoskeletal dis- 
orders) or diabetic neuropathy may present ini- 
tially with evidence of advanced ischemia such 
as arterial ulceration or frank gangrene. Limb 
ischemia should always be considered in the 
evaluation of the older patient who presents 
with a nonhealing ulcer of the lower extremities, 
or with an extensive or persistent skin or soft 
tissue infection of the foot. 

Other causes of limb ischemia include 
embolic or thrombotic sequelae of aortic or 
peripheral aneurysms, embolization from the 
heart or proximal arterial sources, and arterial 
dissection (usually aortic). More unusual causes 
include popliteal entrapment syndrome, ad- 
ventitial cystic disease, and Buerger's disease 
(thromboarteritis obliterans). Some of these eti- 
ologies such as embolization or thrombosis of 



91 




> 



92 



VASCULAR SURGERY 



Loss of pulses 

Claudication 

Rest pain 

Gangrene/Ulceration 




Figure 9.1 . Chronic limb ischemia, progression of disease. Atherosclerosis leads to arterial occlusion, resulting in loss of pulses fol- 
lowed by claudication. In patients with mild peripheral arterial disease, the best treatment option is risk factor modification and exer- 
cise. In more severe cases there is rest pain, which usually occurs at night followed by gangrene (photo insert) or ulceration. Rest pain 
and gangrene/ulceration are considered limb threatening ischemia and the surgical options are amputation or revascularization. 
Aggressive attempts at revascularization are usually undertaken in ambulatory patients. 



an aneurysm present as acute limb ischemia, 
which is characterized by the 5 P's: pulseless- 
ness, paralysis, paresthesia, poikilothermia 
(coolness), and pallor. 



Chronic Ischemia 

Patient History 

Patients are often referred for vascular evalua- 
tion if they have reproducible pain in the lower 
extremities with walking. Although many disor- 
ders can cause these symptoms, several basic 
questions can be asked to ascertain a vascular 
etiology. Patients with vascular claudication 
always have pain when they walk a relatively 
constant distance on level ground; they do not 
have variable days when they can walk for con- 
siderably greater distances without pain. Often 
patients know exactly how far or for how long 
they can walk before the symptoms occur. This 
is in contrast to patients with neurogenic clau- 
dication or musculoskeletal causes of lower 
extremity pain, where the symptoms occasion- 
ally occur at rest or at with highly variable 
walking distances. Stopping results in resolution 
of vascular claudication within a few minutes, 



and this resolution occurs if the patient simply 
stops and stands in place. Patients with neuro- 
genic claudication usually have to sit down to 
relieve their pain. Neurogenic claudication and 
musculoskeletal pain are often induced by 
standing in one place for prolonged periods 
(waiting in line at the bank or washing dishes). 
This is not the case with vascular claudication, 
where lower extremity muscular oxygen 
demands are not as greatly increased by 
standing as they are by prolonged walking. 
Neurogenic claudication is relieved by leaning 
forward, so patients with this disorder often 
note that they can lean forward onto a grocery 
cart or lawn mower and go substantially further 
than they can walk unaided. Similarly, the 
patient with neurogenic claudication often can 
walk further on an incline, whereas vascular 
claudication is marked worsened if the patient 
is on an incline. Patients with musculoskeletal 
disorders often have pain that is present at rest, 
or worsened by standing or sitting in certain 
positions. The pain in neurogenic claudication 
often is described as originating in the thigh 
and then extending down the leg, which is quite 
different from the focal posterior calf pain 
usually noted in vascular claudication. Together, 
these aspects of the history of the patient's 



93 



LOWER LIMB ISCHEMIA 




symptoms help differentiate vascular from neu- 
rogenic claudication. 

More pronounced ischemia results in pain at 
rest, which also has specific features that distin- 
guish it from the many other causes of lower 
extremity pain. Ischemic rest pain occurs when 
the blood flow to the foot is decreased to the 
point where ischemia of the sensory nerves 
occurs, hence the burning causalgia-like quality 
of the pain. Cardiac output decreases with sleep, 
and most patients describe symptoms that are 
initially present only at night. As the ischemia 
becomes advanced, the pain is present con- 
stantly. The more distal aspects of the lower 
limb are the most ischemic, and rest pain is 
most commonly described as occurring across 
the metatarsal heads of the affected foot. 
Ischemic pain awakens the patient from sleep 
and is relieved by dangling the affected limb 
over the edge of the bed, which patients quickly 
learn will allow uninterrupted sleep. Alterna- 
tively patients awakened by the pain find that 
rubbing the foot or walking to stimulate circu- 
lation relieves the pain. Dangling (or standing) 
causes the perfusion pressure of the foot to be 
augmented by the hydraulic pressure due to the 
gravity component of the height of the calf. 
This is approximately 40 cm of water pressure 
(length of the calf), which equals a 29mmHg 
augmentation of foot perfusion pressure. 
This increase is enough to overcome the critical 
closing pressure (CCP) of the precapillary 
sphincter in the vascular bed and restore flow to 
the ischemic regions of the foot. With progres- 
sion of disease and more pronounced ischemia, 
this maneuver no longer provides relief as the 
net pressure falls below the CCP and capillary 
perfusion ceases. Several other disorders cause 
lower extremity pain at night and can be 
confused with ischemic rest pain. Diabetic leg 
cramps are quite common and occur at night, 
but the site of pain is variable and the pain often 
migrates up or down the leg. Musculoskeletal 
pain rarely occurs in the midfoot at night, and 
is usually localized to the joint in question 
(commonly the ankle or knee). Musculoskeletal 
causes of foot pain are usually exacerbated by 
walking or standing and relieved by rest, in con- 
trast to ischemic pain. Infections in the foot, 
particularly osteomyelitis, can cause constant 
pain at rest but are often easily recognized due 
to other signs and symptoms. 



History-taking in the patient with lower 
extremity ischemia should also focus on 
symptoms of atherosclerosis in other vascular 
beds, particularly the cerebral and coronary 
circulation. Patients with symptomatic lower 
extremity ischemia have a 20% to 60% in- 
cidence of significant coronary artery disease, 
and the coexistence of cerebrovascular and 
lower extremity arterial occlusive disease is also 
well established. Symptoms of angina, conges- 
tive heart failure, and transient ischemic attacks 
or strokes should be diligently investigated as 
many patients may ascribe these symptoms to a 
nonvascular cause and may not volunteer this 
important information. The history should also 
include any prior events, such as blue or painful 
toes, which may be suggestive of an embolic 
cause of the ischemia. 

Physical Examination 

The physical examination should be complete 
and focused on the detection of occlusive and 
aneurysmal disease throughout the peripheral 
circulation. The presence (or absence) of carotid 
bruits, cardiac arrhythmias, peripheral pulses, 
and bruits should be documented, and any prior 
scars consistent with arterial bypass surgery 
or vein harvest should be noted. This is of par- 
ticular importance when planning reoperative 
infrainguinal bypass surgery, which may involve 
harvesting autogenous vein from multiple 
sites and limbs. The stigmata of chronic occlu- 
sive or embolic disease should be diligently 
sought, including muscle atrophy, loss of sec- 
ondary skin structures such as hair and nails, 
dependent rubor, splinter hemorrhages, and 
embolic skin lesions or dusky toes. Nonpalpable 
pulses should be interrogated with a handheld 
Doppler, and a bedside ankle — brachial index 
(ABI) determined with an inflatable blood pres- 
sure cuff placed above the site of the Doppler 
signal and then at the wrist. Peripheral and 
aortic aneurysms may be difficult to detect on 
physical examination, particularly in the obese 
patient. A wide or easily palpable popliteal pulse 
is suspicious for a popliteal aneurysm, and eval- 
uation with ultrasound or computed tomogra- 
phy (CT) scanning is indicated to determine the 
true diameter of the vessel. 

More advanced limb ischemia may be asso- 
ciated with arterial ulcers or frank gangrene. 




> 



94 



VASCULAR SURGERY 



Arterial ulcers are distinguished from venous 
ulcers by their location on the more distal 
aspects of the foot, and their exquisite sensitiv- 
ity to touch. They appear as small, dry, punched- 
out lesions in the skin, and have often been 
present for long periods of time without evi- 
dence of granulation tissue or scar contracture 
at the edges. Larger ulceration located at the 
ankle, particularly if moist or weeping, is more 
characteristic of venous disease, although 
patients with combined arterial and venous 
insufficiency may present with long-standing 
ankle ulceration that fails to heal despite 
aggressive treatment of venous insufficiency. 
All patients with presumed venous ulceration 
should undergo examination of peripheral 
pulses, and if not present, prompt evaluation for 
arterial ischemia and consideration for revascu- 
larization. Even moderate degrees of arterial 
insufficiency in conjunction with venous 
insufficiency may result in failure of a primarily 
venous ulcer to heal. Similarly, mo derate arterial 
occlusive disease (ABI 0.4 to 0.5), which ordi- 
narily does not cause tissue loss, may cause a 
surgical incision in the lower extremity not to 
heal in a timely fashion. This is most commonly 
seen after harvest of the lower aspect of the 
greater saphenous vein for coronary artery 
bypass surgery, but can occur after orthopedic 
or podiatric surgery in the lower limb. 

Gangrene of a toe may be produced by 
advanced ischemia alone, or can be secondary 
to infection, particularly in diabetic patients. If 
associated with infection, the infectious process 
often extends further into the forefoot than the 
extent of cutaneous changes. Less frequently a 
patient presents with isolated toe gangrene or 
pregangrenous changes (blue toe) without evi- 
dence of either infection or advanced ischemia 
of the limb. This scenario, particularly if present 
in more than one toe, and especially if found in 
nonadjacent toes, is suspicious for embolic 
disease, or so-called blue-toe syndrome. If the 
involved toes are on both feet, then an embolic 
source proximal to the aortic bifurcation is the 
cause. All patients with evidence of chronic 
peripheral embolization should undergo 
echocardiography and complete angiography of 
the thoracic and abdominal aorta, ileofemoral 
system, and proximal aspect of the involved 
limbs. Embolic sources can include proximal 
aneurysms, which may not be readily detectable 
by angiography, and ultrasound examination of 



the aorta and femoral and popliteal arteries 
should be performed. 

Diagnostic Studies 

The history and physical examination generally 
facilitate classification of the degree of arterial 
insufficiency. Diagnostic studies are indicated 
when the diagnosis is in question, or in prepa- 
ration for intervention. Noninvasive vascular 
testing is also useful in establishing the degree 
of ischemia when there are other confounding 
factors present, such as venous disease, diabetic 
foot ulcers, or active infection. Usually the ABI 
facilitates accurate determination of the degree 
of limb ischemia; however, several conditions 
exist in which the ABI and segmental pressures 
may be falsely elevated. These include diabetes, 
chronic renal failure, and advanced age (over 80 
years), which can cause calcification of the 
medial layer of the arterial wall, which in turn 
causes incompressibility and subsequent false 
elevation of any cuff-based determination of 
peripheral perfusion pressure. An ABI of greater 
than 0.9 is associated with a readily palpable 
pulse, and the absence of a pulse with such an 
ABI value is evidence of incompressibility. In 
these cases several alternatives can be used to 
establish the diagnosis of arterial ischemia. A 
toe cuff can be used to determine a toe — 
brachial index (TBI), as the medial calcification 
rarely extends into the vessels of the foot. The 
waveform tracings from the pulse volume 
recorder are not altered by vessel calcification, 
and examination of the contour of these wave- 
forms at the various arterial levels can suggest 
the site of the occlusive lesions. Flattened wave- 
forms at the ankle or more distal level or a TBI 
less than 0.6 is an indication of arterial insuffi- 
ciency. More sophisticated diagnostic measures 
such as transcutaneous oxygen measurement 
are sometimes useful to determine perfusion 
in the foot of patients with confounding 
factors such as lymphedema or severe venous 
insufficiency. 

Exercise testing plays an important role in the 
subset of patients with symptoms of early occlu- 
sive disease despite relatively normal perfusion 
at rest. Increasing lower extremity blood flow by 
treadmill testing can accentuate the gradient 
across a moderate stenosis and demonstrate a 
drop in distal perfusion pressures after exercise 
that is not present at rest. This is based on 



95 



LOWER LIMB ISCHEMIA 




Poiseuille's law where the pressure drop across 
a stenosis is directly proportional to the volume 
flow across the lesion. Patients with a normal 
ABI at rest and a decreased ABI after exercise 
testing are uncommon and almost always 
have aortoiliac occlusive disease. Many patients 
cannot complete exercise testing on a treadmill 
because of angina or pulmonary dysfunction; 
however, a normal ABI after exercise testing 
excludes arterial insufficiency as a cause of 
lower extremity pain with walking. 

Imaging studies of the aorta and lower 
extremity arteries are not necessary to deter- 
mine the presence or extent of arterial 
insufficiency and are thus reserved for planning 
interventions to revascularize the lower extrem- 
ity. The most widely used study is contrast 
angiography, although duplex scanning and 
magnetic resonance angiography are less inva- 
sive modalities that can provide images that can 
obviate the need for conventional angiography. 
These are utilized in patients with documented 
adverse reactions to contrast agents, or with 
renal insufficiency that increases their risk of 
contrast-induced nephropathy. In most patients, 
contrast angiography provides the most de- 
tailed information to direct catheter-based or 
surgical limb revascularization. Other imaging 
modalities that play a lesser role in the evalua- 
tion of limb ischemia include CT scans and 
ultrasound studies to determine the presence 
of aortic and peripheral aneurysms (especially 
as sources of emboli), echocardiography to 
evaluate potential cardiac embolic sources, and 
duplex evaluation of veins preoperatively for 
use as bypass conduits. 

Treatment 

Mild to moderate limb ischemia that does 
not warrant invasive revascularization can be 
followed with serial examinations. An ABI 
determination is obtained at the initial visit to 
establish a baseline, as this measure may 
improve with exercise or deteriorate with pro- 
gression of disease. The emphasis in treatment 
of these patients is on risk factor modification 
to prevent progression of disease and con- 
currently increase longevity, and a walking 
program to encourage exercise and increase 
exercise tolerance. The most common causes of 
death in patients with symptoms of lower 
extremity arterial insufficiency are ischemic 



cardiac disease and cerebrovascular disease, 
which are both responsive to reduction of the 
same causative risk factors as lower extremity 
ischemia. An important risk factor reduction is 
cessation of tobacco use, and management of 
dyslipidemia, diabetes, and hypertension also 
plays a role. A critical factor is reassurance to 
patients and their family that disease progres- 
sion to critical limb ischemia and possible 
amputation, which is many patients' greatest 
fear, is unlikely, particularly with cessation of 
further tobacco use. The 10-year risk of limb 
loss with claudication is less than 10%, and 
often simply alleviating this fear is the most 
valuable aspect of evaluation and treatment of 
mild to moderate limb ischemia. 

A wide range of pharmacological agents 
and alternative therapies have been utilized to 
treat the symptoms of mild to moderate limb 
ischemia. Unfortunately, controlled clinical 
trials coupled with careful evaluation of long- 
term improvement have demonstrated a consis- 
tent lack of benefit for the vast majority of 
agents tested. This includes vasodilator drugs, 
pentoxifylline, antiplatelet and antithrombotic 
drugs, chelation therapy, and a variety of herbal 
medications such as gingko. The agents that 
have been shown to be of some value in con- 
trolled clinical trials include cilostazol, a phos- 
phodiesterase inhibitor that cannot be used in 
patients with cardiac dysfunction, the Tibetan 
herbal supplement Padma Basic, and high doses 
of L-arginine, the amino acid precursor of 
the endogenous vasodilator nitric oxide. The 
benefit in walking distance with these agents, 
although statistically significant in clinical 
trials, is often minimal in terms of functional 
improvement for the patient. For example in a 
randomized multicenter trial, the mean walking 
distances after 4 weeks of either cilostazol or 
placebo were 306 versus 267 m (Money et al., 
1998). It is unclear whether these minimal 
increases represent a meaningful improvement 
over a graduated exercise program alone. 
Studies of graded exercise programs have 
demonstrated that motivated patients can 
double their walking distance; however, this 
requires walking to near-maximal pain levels 
for 30 minutes on a regular basis for at least 6 
months (Gardner and Poehlman, 1995). 

Many patients with moderate chronic limb 
ischemia have symptoms that they consider dis- 
abling, and seek revascularization to increase 




> 



96 



VASCULAR SURGERY 



their ability to exercise or perform job-related 
tasks. The role of revascularization, whether 
catheter-based or surgical, in patients with 
claudication remains controversial. Factors that 
should be considered include ongoing cigarette 
use, the patient's commitment to exercise and 
physical activity, and the anatomical level of 
occlusive disease that requires correction. Aor- 
toiliac occlusive disease, as judged by physical 
examination and noninvasive studies, can be 
treated well by angioplasty and stenting with 
minimal risk to the patient. More extensive 
disease, particularly involving either the 
infrarenal aorta or the external iliac arteries, is 
better treated with aortobifemoral bypass graft- 
ing. The long-term results of aortobifemoral 
bypass grafting are excellent, with patencies 
exceeding 90% at 5 years. Although aortoiliac 
endarterectomy was the first procedure devel- 
oped for the treatment of occlusive disease of 
these vessels, it has largely been replaced by 
bypass grafting. Extensive unilateral disease, 
particularly occlusion of the external iliac 
artery, can often be treated with femoral- 
femoral bypass grafting. This is a procedure of 
substantially smaller magnitude than an aorto- 
bifemoral bypass, and is often the procedure of 
choice in patients with coexisting cardiac and 
pulmonary disease. Femoral-femoral bypass 
requires normal flow in the contralateral ile- 
ofemoral system, and mild to moderate disease 
of the contralateral artery can be treated with 
angioplasty and stenting to obtain sufficient 
inflow to support the femoral-femoral bypass 
graft. Although some centers have advocated 
axillofemoral bypass grafts for mild to 
moderate limb ischemia and claudication, our 
policy is to reserve this form of extensive extra- 
anatomical bypass for poor-risk patients with 
limb-threatening ischemia or when revascular- 
ization is required after removal of infected 
aortic grafts. 

Occlusive disease of the distal superficial 
femoral artery, another common site in mild 
to moderate limb ischemia, generally requires 
bypass with either prosthetic or autologous 
grafts (Table 9.1). Femoral-popliteal bypass 
grafting is often performed with excellent 
results in patients with disabling claudication. 
A randomized prospective trial of prosthetic 
versus greater saphenous vein bypass grafts 
in the femoral to above-knee popliteal artery 
did not demonstrate a difference in long-term 



Table 9.1 . Conduits for revascularization 



Axillofemoral, aortofemoral, 


Dacron or ePTFE 


femoral-femoral 




Femoral: above the knee 


Autologous vein, 




Dacron, or PTFE 


Femoral: below the knee 


Autologous vein 



PTFE, polytetrafluoroethylene; ePTFE, expanded PTFE. 



patency (Veith et al., 1986). A randomized mul- 
ticenter trial demonstrated an improved graft 
patency and decreased risk of subsequent 
amputation with heparin-bonded Dacron grafts 
in comparison to polytetrafluoroethylene 
(PTFE) grafts (Devine et al., 2001), and repre- 
sents the first major advance in synthetic vas- 
cular grafts that has been shown to improve 
clinical results relative to standard materials. 
Self-expanding and balloon-expandable stents 
in conjunction with balloon angioplasty are 
being applied to stenoses and occlusions of the 
distal superficial femoral artery with improving 
results, and are extending the ability to provide 
percutaneous revascularization of moderate 
lower limb ischemia. 

Occlusive disease of the popliteal artery or 
more distal vessels, although uncommon in the 
nondiabetic patient with claudication, requires 
bypass with autogenous vein to the distal 
popliteal artery or tibial vessels. These proce- 
dures are generally reserved for more severe 
ischemia where salvage of the extremity is in 
question. If there is calf claudication with exten- 
sive occlusive disease of the proximal aspects of 
the tibial arteries, a femoral-tibial bypass to a 
distal vessel may result in excellent perfusion of 
the foot with minimal relief of claudication. 
This is due to the lack of retrograde perfusion 
to the geniculate arteries that supply the major 
muscles of the upper calf. 

More advanced ischemia of the lower limb 
generally requires a more aggressive approach 
to revascularization if long-term viability of the 
limb is to be preserved. The need for revascu- 
larization needs to be combined with a complete 
evaluation of the medical and functional status 
of the patient. Patients with critical limb 
ischemia, which presents with rest pain, arterial 
ulcers, or frank gangrene, commonly have mul- 
tilevel occlusive disease, which usually requires 



97 



LOWER LIMB ISCHEMIA 




major surgical revascularization to achieve 
long-term limb salvage. They also have corre- 
spondingly more advanced atherosclerosis in 
their coronary and cerebrovascular circulation, 
and their perioperative morbidity and mortal- 
ity is higher than for patients with claudication. 
The incidence of renal insufficiency or chronic 
failure as well as diabetes is higher in patients 
who present with signs of critical limb ischemia, 
and these medical factors have been noted in 
several multivariate analyses to be independent 
predictors of poor outcomes and higher mor- 
tality after arterial reconstruction. 

Revascularization in patients with critical 
limb ischemia often requires correction of 
both aortoiliac and infrainguinal (femoral- 
popliteal-tibial) occlusive disease. In patients 
who present with advanced tissue loss or gan- 
grene and multilevel occlusive disease, restora- 
tion of in-line arterial flow to the foot is 
required to heal the large tissue defects. A com- 
bination of percutaneous treatment of the aor- 
toiliac disease (if anatomically suitable) with 
femoral-popliteal or femoral-tibial bypass can 
be used to rapidly restore foot perfusion in 
these cases. If the presenting ischemic symp- 
toms are rest pain or small arterial ulcers, then 
correction of one level of occlusive disease 
(usually the aortoiliac) with angioplasty/stent- 
ing or surgical bypass relieves the symptoms. 

The conduit of choice for femoral-tibial 
bypass, or femoral-popliteal bypass with poor 
runoff is the greater saphenous vein (Table 9.1). 
Prospective studies comparing reversed versus 
in situ saphenous vein grafts have not demon- 
strated a difference in patency or limb salvage 
rates between the two techniques (Harris et al., 
1987). In many patients with critical limb 
ischemia, the ipsilateral greater saphenous vein 
is not available for use as a conduit due to prior 
harvest or vein stripping. If the contralateral 
greater saphenous vein is not available, then 
secondary sources of autogenous vein such as 
the lesser saphenous vein or arm veins should 
be preoperatively mapped by duplex scanning 
and utilized. The use of spliced segments of 
autologous vein was recently compared to PTFE 
grafts with vein cuffs in a randomized prospec- 
tive trial of patients without an available greater 
saphenous vein (Kreienberg et al., 2002). 
Patency was greater in the spliced arm vein 
group (87% vs. 59% at 2 years) with similar rates 
of limb salvage (94% and 85%). Alternative 



choices for conduit include cryopreserved 
cadaver saphenous vein, umbilical vein grafts, 
conventional prosthetic grafts (PTFE and 
Dacron), and heparin-bonded Dacron. These 
have variable patency rates, all of which are infe- 
rior to saphenous vein or spliced autologous 
veins when utilized for femoral-tibial bypass. 

A useful technique in patients with limited 
amounts of available vein is to originate the 
graft from a more distal site than the common 
femoral artery, thus requiring shorter segment 
of vein conduit. Long-term patency is not com- 
promised by originating the graft from the deep 
femoral artery, the superficial femoral artery, or 
the popliteal artery provided that there is not 
significant occlusive disease above the inflow 
site (Wengerter et al., 1992). Autogenous vein 
grafts should be studied postoperatively along 
their entire length periodically with duplex 
ultrasound to identify potential areas of mid- 
graft stenosis, which should be corrected with 
either balloon angioplasty or surgical repair 
before graft thrombosis occurs. 

Many patients with critical limb ischemia 
are bedridden or do not ambulate because of 
neurological or musculoskeletal problems. In 
these patients, primary amputation is a safer 
and more expeditious means of dealing with 
foot gangrene than surgery for revasculariza- 
tion. The level of amputation, which is always 
of great patient concern even when ambula- 
tion is not an issue, is determined by the ambu- 
lation potential of the patient and the degree 
of perfusion required to heal the amputation. 
Although there are numerous guidelines and 
means of measuring skin perfusion to deter- 
mine the appropriate level of amputation, 
clinical judgment remains the final factor. 
Enthusiasm for various quantitative means of 
measuring limb and skin blood flow has been 
tempered by prospective studies of these 
various techniques, which have failed to identify 
one quantitative test that preoperatively esti- 
mates probability of healing with sufficient 
positive and negative predictive value. Transcu- 
taneous oxygen testing (TcP0 2 ) is the most 
readily available of these sophisticated blood 
flow measurements, which include radiolabeled 
xenon washout, laser Doppler velocimetry, and 
photoplethysmography perfusion studies. A 
TcP0 2 of greater than 40mmHg at the level of 
proposed amputation is predictive of healing, 
and a TcP0 2 of less than 20mmHg is indicative 




> 



98 



VASCULAR SURGERY 



of a high likelihood of failure to heal. Unfortu- 
nately, many patients with lower extremity 
ischemia fall into the range of 20 to 40mmHg, 
where clinical judgment must be used to decide 
on the level of amputation. 

We recommend attempting a below-knee 
amputation in any ambulatory patient with rea- 
sonable rehabilitation potential in whom there 
is detectable popliteal artery Doppler signal. 
This approach is based on the known benefits 
with preservation of the knee joint for ambula- 
tion, and will predictably result in a small 
number of failures that require revision as a cost 
of salvaging as many below-knee amputations 
as possible. In the nonambulatory patient with 
advanced ischemia, a primary above-knee 
operation is performed if perfusion at the level 
of the popliteal artery is poor or nondetectable. 
Although there is some benefit to preservation 
of the knee joint even in the nonambulatory 
patient for aid in transferring from bed, major 
amputations in this population carry at least a 
10% to 15% mortality per procedure and it is 
recommended to perform a single definitive 
above-knee procedure if there is questionable 
perfusion in a nonambulatory patient. 

Outcomes 

There has been a steady improvement in limb 
salvage rates due to refinements in both percu- 
taneous and surgical revascularization for 
chronic limb ischemia. In particular, the wide- 
spread application of femoral-tibial bypass 
has led to revascularization of limbs that until 
recently would have been deemed inoperable. 
Awareness of limb ischemia as a cause of non- 
healing ulcers and patient and practitioner edu- 
cation are responsible for earlier evaluation and 
referral of patients for revascularization. The 
success of aortoiliac angioplasty and stenting 
has led to relief of claudication in patients 
without the morbidity and length of hospital stay 
previously associated with aorto femoral bypass. 
Despite the advances in percutaneous and 
surgical revascularization, a large number of 
patients with critical limb ischemia will eventu- 
ally undergo an amputation (>50,000 cases per 
year in the United States). Many patients are 
not candidates for standard revascularization 
techniques because of anatomical or medical 
factors. The discovery that endogenous peptides 
can induce growth of new blood vessels (angio- 



genesis) has spurred interest in applying angio- 
genic therapy to patients with limb ischemia. 
Preliminary results with growth factors such 
as fibroblast growth factor are encouraging 
(Lederman et al., 2002), and further studies are 
needed to define the optimal growth factor, 
route of delivery, and duration of therapy. Sim- 
ilarly, the finding that circulating bone 
marrow — derived stem cells contribute to the 
angiogenesis and spontaneous revasculariza- 
tion seen in experimental hindlimb ischemia 
has led to trials of stem cell therapy for the treat- 
ment of chronic limb ischemia in humans, with 
promising preliminary results (Tateishi-Yuyama 
et al., 2002). These areas of investigation should 
lead to therapies for critical limb ischemia that 
will complement surgical revascularization and 
extend our ability to provide limb salvage. 



Acute Ischemia 

Patient History 

The most important determination in the eval- 
uation of acute limb ischemia is whether the 
ischemia is due to an arterial embolus or throm- 
bosis of a chronically diseased artery. Two 
aspects of the patient's history contribute to this 
decision. The first is the onset of symptoms. 
Embolic occlusion of a normal arterial bed 
results in sudden onset of symptoms, and the 
patient can often recollect the exact moment of 
onset of the ischemia. In contrast, thrombotic 
occlusion of a diseased native artery occurs 
more gradually, with symptoms often worsen- 
ing over several days. The second important 
aspect of the history is the presence or absence 
of chronic peripheral arterial occlusive disease, 
which is more frequently associated with 
thrombosis rather than embolism. Thus the 
presence of previous symptoms such as claudi- 
cation or rest pain, or a history of prior 
interventions for peripheral arterial occlusive 
disease, strongly suggests that the acute 
ischemia is secondary to thrombosis of a dis- 
eased vessel or bypass graft. Many of these 
patients have an antecedent history of acute 
dehydration from gastrointestinal causes or 
poor perfusion, from acute cardiac dysfunction. 
Conversely a lack of prior claudication coupled 
with the presence of risk factors for arterial 
embolization (atrial fibrillation, a recent 



99 



LOWER LIMB ISCHEMIA 




myocardial infarction, dilated cardiomyopathy, 
or prior embolic event) is more consistent with 
either arterial embolization or another unusual 
nonatherosclerotic cause of acute ischemia such 
as dissection or thrombosis of a peripheral 
aneurysm. Unfortunately, this maxim cannot 
always be relied on, as up to 25% of patients 
with embolism as a cause of acute limb ischemia 
have long-standing signs and symptoms of 
prior peripheral arterial occlusive disease. 

Physical Examination 

Acute limb ischemia is characterized by the five 
P's: pulselessness, paralysis, paresthesia, pain, 
and pallor. The level of arterial occlusion is gen- 
erally one anatomical level higher that the clin- 
ical manifestation of the ischemia. Thus patients 
with an embolus lodged in the proximal superfi- 
cial femoral artery present with an ischemic 
calf. The severity of the ischemia can be deter- 
mined from the physical findings, with pain and 
pallor occurring early and paresthesia and 
paralysis being later findings. Paresthesia is due 
to direct ischemia of the sensory nerves within 
the extremity and is often reversed with prompt 
revascularization. Paralysis can be due to either 
ischemia of the motor neurons or muscle death. 
Muscle death can be determined on examina- 
tion by rigidity of the ischemic muscle to pal- 
pation (rigor) and corresponding difficulty 
moving the joints with passive motion. If skele- 
tal muscle death has occurred, there is little to 
no chance for meaningful limb salvage. 

A major problem with diagnosis of acute 
limb ischemia remains the failure to consider 
ischemia as a cause of acute symptoms in the 
limb, leading to delays in treatment that can lead 
to limb loss. These delays are usually seen in 
patients without known prior peripheral vascu- 
lar disease, such as those with undiagnosed 
popliteal aneurysms, aortic dissection, or 
(most commonly) arterial embolism. The most 
common misdiagnosis is a primary neurologi- 
cal problem, such as spinal cord impairment or 
stroke, to which the paresthesia and paralysis of 
ischemia is attributed. 



Diagnostic Studies 

The history and physical examination in acute 
limb ischemia are focused on the critical issue 



of whether the occlusion is due to embolism 
or thrombosis of a diseased artery, as this dis- 
tinction leads to either immediate surgical 
embolectomy or preoperative angiography to 
delineate the cause of ischemia. The arteries of 
the lower extremity have tremendous capacity 
for collateral flow, and thus thrombosis of a 
native artery occurs only when atherosclerotic 
lesions are very advanced. Such advanced ath- 
erosclerosis is usually symmetrical, and the con- 
tralateral limb does not have a normal pulse 
exam. The presence of normal pulses in the 
contralateral limb is highly suggestive of an 
embolism as a cause of acute ischemia, and 
warrants operative embolectomy without the 
delay associated with preoperative angiography. 
Unfortunately, the converse is not always true, 
as signs and symptoms of prior peripheral 
arterial occlusive disease do not guarantee 
that the acute ischemia is due to thrombosis 
rather than embolism. Thus the presence of 
significant peripheral arterial occlusive disease 
(defined by both prior symptoms or physical 
findings in the contralateral limb) in patients 
with acute limb ischemia is an indication for 
angiography. Angiography definitively identi- 
fies the cause of the acute ischemia and delin- 
eates the proximal and distal arterial anatomy 
should a bypass be required either immediately 
or subsequently to relieve ischemia. Nonem- 
bolic causes of acute ischemia include 
thrombosis of a chronically diseased vessel, 
thrombosis of a previous bypass graft, throm- 
bosis of a peripheral aneurysm (particularly a 
popliteal aneurysm), and proximal arterial dis- 
section (usually aortic). The treatment of these 
conditions is quite varied, and immediate surgi- 
cal exploration without angiography is often 
unsuccessful in restoring flow. In many cases, 
angiography is not only diagnostic but is thera- 
peutic in terms of initiating thrombolytic 
therapy for occlusion of bypass grafts or throm- 
bosed popliteal artery aneurysms. Aside from 
assessment of limb perfusion by Doppler exam- 
ination, immediate angiography is the only 
diagnostic study utilized in the evaluation of 
acute limb ischemia. 

Treatment 

For any form of acute limb ischemia, systemic 
anticoagulation with intravenous unfraction- 
ated heparin is immediately instituted while 




> 



100 



VASCULAR SURGERY 



preparations are made for surgery or angiogra- 
phy. Patients with known sensitivity to heparin 
or a documented history of heparin-induced 
thrombocytopenia can be treated with direct 
thrombin inhibitors. Anticoagulation alone 
usually does not relieve the ischemia, but it 
helps prevent further propagation of the throm- 
bus and preserve flow in collateral vessels 
around the occlusive thrombus. The specific 
treatment of acute limb ischemia rests on pre- 
operative establishment of the cause of the acute 
ischemia. In a patient with a prior history of 
peripheral bypass surgery, the most common 
cause of acute limb ischemia is thrombotic 
occlusion of the bypass graft. In these patients, 
as well as patients with preexisting chronic 
peripheral vascular occlusive disease, preopera- 
tive angiography is usually obtained to deter- 
mine the location of the occlusion and the 
inflow and outflow sites for bypass grafting. Pre- 
operative angiography is not utilized in those 
cases of advanced acute ischemia where the 
location of the problem is clinically obvious, 
such as a patient with a prior aortobifemoral 
bypass graft, an absent femoral pulse, and a pro- 
foundly ischemic limb. The delay associated 
with obtaining preoperative angiography is also 
avoided in patients with suspected arterial 
embolism, which is treated with emergent 
catheter embolectomy. 

Restoration of a functional and viable intact 
limb is extremely rare when skeletal muscle 
death has already occurred. If physical exami- 
nation suggests that limb paralysis is due to 
death of the skeletal muscle, then the status of 
the muscle is determined by operative explo- 
ration. The high likelihood of amputation is dis- 
cussed with the patient and family members 
prior to surgery, and primary amputation is per- 
formed if there is no bleeding from the muscle 
and no muscle contraction with direct electrical 
stimulation. Aortoiliac revascularization maybe 
subsequently required to obtain adequate blood 
flow to allow healing of the definitive amputa- 
tion site. 

Treatment of lower limb ischemia secondary 
to aortic dissection can be either directed at cor- 
recting the dissection or an extra-anatomical 
bypass to relieve the limb ischemia. Correction 
of the underlying dissection may require 
surgical repair of the thoracoabdominal aorta, 
although these procedures are formidable 
undertakings in patients who are often critically 



ill with concurrent renal, mesenteric, or spinal 
cord ischemia. Mortality rates approaching 40% 
have been reported from major centers in 
patients with coexisting cardiac and renal 
disease. Advances in catheter-directed therapy 
for acute dissection are allowing rapid restora- 
tion of perfusion to the involved branches of the 
aorta without the physiological stress of opera- 
tive repair of the thoracoabdominal aorta 
(Slonim et al., 1996). These procedures include 
endovascular stenting of dissections within the 
thoracic aorta and catheter-based percutaneous 
fenestration to restore perfusion to both false 
and true aortic lumina. 

For limb ischemia secondary to suspected 
femoral, iliac, or aortic bifurcation emboli, 
embolectomy is performed via the common 
femoral artery. For bilateral limb ischemia 
where the embolus is lodged in the aortic bifur- 
cation, bilateral transfemoral embolectomy is 
performed. Catheter embolectomy should be 
performed both proximally and distally from 
the common femoral artery. This is usually per- 
formed through a transverse arteriotomy in the 
common femoral artery placed opposite the 
orifice of the deep femoral artery to facilitate 
passage of the catheter into both the superficial 
and deep femoral arteries. The first priority 
should be establishment of adequate inflow to 
the common femoral artery. This is usually 
accomplished easily with catheter embolectomy, 
but occasionally the arterial flow from the exter- 
nal iliac artery may be unsatisfactory even after 
removal of all possible thrombus. This is usually 
due to preexisting chronic occlusive disease 
of the iliac arteries, but can also be caused 
by dissection of a diseased vessel during the 
embolectomy. There are two options to manage 
inadequate inflow after transfemoral em- 
bolectomy. The traditional solution is surgical 
bypass, usually with a femoral-femoral or 
axillofemoral graft. More recently, intraopera- 
tive angioplasty and stenting of the iliac system 
can be used to treat occlusive disease or dissec- 
tion of the ipsilateral iliac arteries. This can be 
performed via either an ipsilateral retrograde 
approach or the contralateral femoral artery. 
The use of angioplasty and stenting in this 
setting can often restore adequate inflow more 
rapidly than constructing an extra-anatomical 
bypass, and is the procedure of choice if 
the appropriate expertise and equipment are 
available. 



101 



LOWER LIMB ISCHEMIA 




Caution must be taken when restoring blood 
flow to the severely ischemic limb, as the stag- 
nant and ischemic venous blood contains toxic 
metabolites from the limb. Sudden return 
into the systemic circulation can be associated 
with serious complications including acido- 
sis, cardiac arrhythmias, or arrest and renal 
damage. Release of the venous return from the 
ischemic limb to the body is done in close 
cooperation with the anesthesiologists, and 
administration of antiarrhythmic agents, 
sodium bicarbonate, and mannitol may be 
required. In patients with prolonged ischemia, 
particularly those with preexisting cardiac dys- 
function, consideration is given to draining the 
initial venous return from the limb to prevent 
flow of these toxic metabolites back to the cir- 
culation of a compromised patient. The com- 
mon femoral vein is encircled at the level of the 
inguinal ligament with a Rummel tourniquet 
and the first 300 to 600 mL of venous blood is 
exsanguinated via a transverse venotomy at the 
time that arterial inflow to the limb is restored. 
Appropriate replacement with banked blood is 
essential to maintain adequate cardiac output 
and oxygen delivery, and should be done con- 
currently with the venous drainage. 

Most emboli of cardiac origin lodge in the 
common femoral or proximal superficial fe- 
moral artery. Smaller emboli may lodge in the 
popliteal artery, and embolic occlusion of indi- 
vidual tibial arteries from a cardiac source is 
unusual. Once adequate inflow to the common 
femoral artery is established as described above, 
the embolectomy catheter is directed distally 
down the superficial femoral artery to retrieve 
distal thromboemboli. Restoration of back- 
bleeding from the superficial femoral artery is 
followed by embolectomy of the profunda 
femoris artery, which is rarely the site of 
embolization. Propagation of a secondary 
thrombus, however, does occur in the proximal 
aspect of this vessel, although more distal seg- 
ments remain patent due to collateral circula- 
tion via the numerous branches. Transfemoral 
embolectomy of the distal circulation may 
sometimes not result in restoration of a satis- 
factory Doppler signal at the level of the ankle. 
There may be subsequent propagation of 
thrombus distally into tibial vessels, or frag- 
ments of more proximal emboli may become 
dislodged into the distal circulation during 
catheter embolectomy. In this case on-table 



angiography is performed to determine the 
location and amount of residual thrombus. If 
thrombus is found in the proximal aspects of the 
individual tibial arteries and blood flow to the 
remains poor, then more distal embolectomy is 
performed to restore blood flow to the foot. This 
is usually performed via the infrageniculate 
popliteal artery, which is exposed via a medial 
incision down distally to the tibioperoneal 
trunk in order to allow the passage of the 
embolectomy catheter into each of the tibial 
vessels. If the infrageniculate popliteal artery is 
small or diseased, a longitudinal arteriotomy is 
closed after embolectomy with a small vein 
patch; otherwise a transverse arteriotomy can 
be primarily closed with interrupted sutures. 

Irrigation of the distal circulation with 
heparinized saline containing papaverine helps 
relieve spasm of the tibial vessels induced by 
passage of the embolectomy catheter. Intra- 
operative instillation of thrombolytic therapy, 
usually urokinase or streptokinase, has been 
described as a means of treating residual 
thrombus that cannot be retrieved by catheter 
embolectomy. Dramatic increases in patency of 
the distal circulation have been noted after 
intraoperative thrombolytic therapy, and this 
technique is particularly useful when there 
appears to be insufficient runoff to maintain 
patency of either a distal bypass graft or the 
native proximal popliteal artery. 

Arterial bypass plays an important role in the 
management of acute limb ischemia, particu- 
larly in patients with ischemia secondary to 
thrombosed popliteal artery aneurysms, arte- 
rial dissection of the ileofemoral arteries, or 
thrombosis of a chronically diseased aortoiliac 
segment. Bypass operations for acute occlusion 
of the aortoiliac segment include aortofemoral 
bypass, femoral-femoral bypass, and axillo- 
femoral bypass. The latter two are less extensive 
procedures that are particularly useful in the 
emergent management of acute aortoiliac 
thrombosis secondary to acute medical illness 
such as cardiogenic shock. The femoral-femoral 
bypass is performed for acute unilateral 
ischemia if embolectomy fails to restore ade- 
quate inflow or if the acute ischemia is due to 
any nonembolic cause. A clinically normal con- 
tralateral femoral pulse is a prerequisite for a 
femoral-femoral bypass graft, and a decreased 
contralateral pulse would favor the placement of 
an axillofemoral bypass or aortofemoral bypass 




> 



102 



VASCULAR SURGERY 



in the patient whose medical condition will 
tolerate it. Emergent axillofemoral bypass and 
aortofemoral bypass are also used to treat bilat- 
eral acute limb ischemia. Axillofemoral bypass 
is applied in the unstable or critically ill patient 
in whom the more extensive incisions, pro- 
longed time to revascularization, and greater 
fluid shifts associated with aortofemoral bypass 
may be detrimental. Emergent aortofemoral 
bypass for acute ischemia has the advantage of 
providing definitive revascularization from the 
most reliable source of inflow, namely the 
nondiseased juxtarenal aorta. The disadvantage 
of urgent or emergent aortofemoral bypass is 
the substantial magnitude and duration of an 
aortic procedure in a potentially ill or medically 
unstable patient with insufficient time for pre- 
operative optimization of associated medical 
conditions. 

Infrainguinal bypass procedures are also 
commonly used in the management of acute 
limb ischemia. Thrombosis of a popliteal artery 
aneurysm or a diseased popliteal artery is a 
typical indication, as are symptomatic acute 
thrombosis of the superficial femoral artery 
and dissections extending more distal to the 
common femoral artery. Acute thrombosis of a 
popliteal artery aneurysm or diseased popliteal 
artery can be successfully treated with popliteal 
or tibial bypass only if a patent target vessel 
below the popliteal artery is identified on pre- 
operative angiography. Although some authors 
have advocated bypass to isolated or "blind" 
segments of the popliteal or tibial arteries, we 
prefer to utilize target vessels that flow across 
the ankle joint to supply the pedal arch, or in the 
case of the peroneal artery, collateralize via the 
anterior or posterior branch at the malleolus to 
provide flow to either the anterior tibial or pos- 
terior tibial artery that subsequently supplies 
the foot. Failure to visualize any target vessel 
in the calf below the level of acute thrombosis 
is an indication for intraarterial thrombolytic 
therapy to improve the outflow and provide a 
suitable target for a subsequent bypass graft. 
Neurological changes secondary to acute 
ischemia are traditionally considered con- 
traindications for thrombolytic therapy due to 
the time required for effective thrombolysis. 
However, the results with thrombolytic therapy 
for moderate to severe acute ischemia (particu- 
larly thrombosed popliteal aneurysms) are 
substantially better than the dismal outcomes 



Table 9.2. Controversies in the management of limb ischemia 

Primary amputation versus complex bypass for limb 

salvage in patients with critical limb ischemia and 

renal failure and diabetes 
Thrombolytic therapy versus surgical thrombectomy 

for aortofemoral graft occlusion. 
Thrombolytic therapy versus surgical therapy for 

primary aortoiliac thrombosis. 



of attempted bypass surgery to a distal vessel 
not angiographically visualized below the level 
of acute thrombosis. Accordingly, we utilize 
thrombolytic therapy even in the presence of 
early neuromuscular changes if there are no 
vessels visualized initially that are suitable 
targets for bypass grafting (Table 9.2). Although 
thrombolytic therapy is usually associated with 
improvement in the acute ischemia, serial exam- 
ination of these patients for worsening ischemia 
is critical to determine if thrombolytic therapy 
should be terminated and surgical bypass 
performed. 

The conduit of choice for infrainguinal 
bypass to treat acute limb ischemia is the 
greater saphenous vein, either from the involved 
limb or if necessary from the contralateral leg. 
There is usually insufficient time for preopera- 
tive vein mapping in such patients, and the suit- 
ability of the saphenous vein is defined by 
operative exploration if there are no signs of 
prior harvest. If there is no available saphenous 
vein, which is often the case in patients with 
acute ischemia secondary to thrombosis of a 
prior saphenous vein graft, then a prosthetic 
(usually PTFE) graft with a vein patch or cuff or 
a cryopreserved cadaver vein can be used. Both 
of these options are associated with substan- 
tially worse long-term patency than autologous 
saphenous vein, particularly when used for 
bypass to the below-knee popliteal artery or 
tibial arteries. 

After revascularization for acute limb 
ischemia, consideration is given to immediate 
fasciotomy if the ischemia was severe and of 
greater than 4 to 6 hours' duration. Immediate 
prophylactic fasciotomy avoids problems with 
identifying compartment syndrome subse- 
quently in patients who are receiving pain med- 
ication or sedation and already have pain in the 
limb from the surgical incisions (particularly if 
the popliteal artery was exposed below the 



103 



LOWER LIMB ISCHEMIA 




knee). The need for fasciotomy is less common 
in patients with preexisting arterial insuffi- 
ciency, as they possess preformed arterial col- 
lateral pathways that decrease the degree of 
acute ischemia induced by the superimposed 
thrombosis or embolus. Nonetheless, either 
immediate fasciotomy or careful observation for 
development of elevated compartment pres- 
sures is mandatory following any prolonged 
period of ischemia of the lower extremity. This 
is particularly true in patients with thrombosis 
of an aortobifemoral bypass limb with poor 
outflow, as profound ischemia is produced when 
the inflow graft occludes. 

The early signs and symptoms of compart- 
ment syndrome are the 3 P's: pink, painful, 
and pulses (present). Before the capillary leak 
induced by the ischemia-reperfusion causes 
intracompartment pressures to eventually ex- 
ceed mean arterial pressure, there is a palpable 
pulse in the involved limb. The skin appears 
pink as the dermal plexus of arterioles main- 
tains perfusion despite ischemia of the underly- 
ing muscle. The pain induced by compartment 
syndrome in the conscious patient is initially 
present only with motion of the muscles in the 
affected compartment. Thus passive stretching 
of the first toe is one of the most sensitive tests 
for compartment syndrome of the anterior 
compartment of the lower leg. As the ischemia 
becomes advanced, pain is present at rest 
and becomes excruciating. Immediate four- 
compartment fasciotomy is performed via 
double incisions if there is any clinical suspi- 
cion or signs of compartment syndrome after 
revascularization. 

In the intubated or otherwise unresponsive 
patient where the diagnosis is unclear, meas- 
urement of intracompartment pressures with 
either a Stryker device or an intravenous infu- 
sion pump [with pressure sensing capability, 
i.e., an intravenous accurate control (IVAC) 
machine] enables the diagnosis of compartment 
syndrome to be made in the absence of the 
characteristic signs and symptoms. Pressures 
greater than 12 to 15mmHg are treated with 
fasciotomy. Fasciotomy of the lower limb is 
usually performed through two incisions, 
although a single incision fasciotomy with 
fibulectomy can enable decompression of all 
four fascial compartments. The skin incisions 
can often be closed at the time of fasciotomy, 
and this allows more rapid healing than the 



weeks associated with healing open wounds by 
secondary intention. If there is a question of 
whether the edema will compromise skin 
closure, we place interrupted nylon horizontal 
mattress sutures in the skin and leave them 
untied at the time of fasciotomy. Once the 
edema subsides (usually 1 to 3 days), delayed 
primary closure is accomplished by tightening 
and tying these sutures at the bedside under 
intravenous sedation. The muscle ischemia in 
compartment syndrome results in myoglobine- 
mia and myoglobinuria, and precautionary 
measures to protect the kidneys from precipita- 
tion of myoglobin within the renal tubules are 
instituted in all patients with compartment 
syndrome. Alkalinization of the urine is 
accomplished by administration of intravenous 
sodium bicarbonate, and close monitoring of 
serum electrolytes as well as serum myoglobin 
and creatine kinase (CK) levels is continued 
until the syndrome subsides. In patients with 
documented myoglobinurea, we routinely 
monitor urine pH to confirm alkalinization of 
the urine. 

Outcomes 

The probability of limb salvage in acute lower 
limb ischemia is dependent on two factors. The 
first and more important is the duration and 
degree of ischemia prior to revascularization. In 
patients with prior peripheral vascular surgery, 
diagnosis and treatment are usually not 
delayed, as both the patient and physician are 
focused on ischemia as a cause of the symptoms 
in the limb. As discussed above, patients with 
arterial embolism or aortic dissection can be 
misdiagnosed as having a primary nonvascular 
cause of their limb symptoms. The associated 
delay in treating the ischemia can often con- 
tribute to ultimate limb loss. The second factor 
in determining the outcome in acute limb 
ischemia is the success of revascularization, 
which is related to the level of disease responsi- 
ble for the acute ischemia. More proximal occlu- 
sive disease, particularly aortoiliac disease or 
occlusion, is readily treated with embolectomy 
or bypass and generally associated with good 
outcomes. Acute limb ischemia due to failure of 
a prior infrainguinal bypass graft placed for 
prior critical limb ischemia, particularly if there 
have been multiple prior bypass procedures 
in the involved limb, is associated with worse 




» 



104 



VASCULAR SURGERY 



outcomes as the quality of the distal target 
vessel and available bypass conduit progres- 
sively decreases. 

Long-term outcomes after arterial embolism 
are determined by the underlying medical con- 
dition of the patient, particularly the cardiac 
status. Despite long-term anticoagulation, up to 
20% of patients with arterial embolism suffer 
another embolic event. Ischemic neuropathy of 
either the sensory or motor nerves can persist 
after revascularization for acute limb ischemia, 
and can be a source of frustration to both the 
patient and physician. Newer nonnarcotic 
modalities for chronic pain and dysesthesia are 
emerging, including electrical nerve stimula- 
tion, use of tricyclic drugs, and other options to 
treat chronic neuropathy. 

Patients who present with acute ischemia sec- 
ondary to thrombosis of a popliteal aneurysm 
have a high rate (40% to 50%) of limb loss 
despite modern advances in thrombolytic 
therapy and peripheral bypass surgery. These 
results reinforce the need to aggressively diag- 
nose and treat popliteal aneurysms prior to 
thrombosis, and the finding that 15% of patients 



with aortic aneurysms have evidence of an 
aneurysm in the lower limb when prospectively 
screened (Diwan et al., 2000) provides a basis for 
such a strategy. 



References 



Devine C, Hons B, McCollum C. (2001) J Vase Surg 33: 

533-9. 
Diwan A, Sarkar R, Stanley JC, Zelenock GB, Wakefield TW. 

(2000) J Vase Surg 31:863-9. 
Gardner AW, Poehlman ET. (1995) JAMA 274:975-80. 
Harris PL, How TV, Jones DR. (1987) Br J Surg 74:252-5. 
Kreienberg PB, Darling RC 3rd, Chang BB, et al. (2002) J Vase 

Surg 35:299-306. 
Lederman RJ, Mendelsohn FO, Anderson RD, et al. (2002) 

Lancet 359:2053-8. 
Money SR, Herd JA, Isaacsohn JL, et al. (1998) J Vase Surg 

27:267-74; discussion 274-5. 
Slonim SM, Nyman U, Semba CP, Miller DC, Mitchell RS, 

Dake MD. (1996) J Vase Surg 23:241-51; discussion 

251-3. 
Tateishi-Yuyama E, Matsubara H, Murohara T, et al. (2002) 

Lancet 360:427-35. 
Veith FJ, Gupta SK, Ascer E, et al. (1986) J Vase Surg 3:104-14. 
Wengerter KR, Yang PM, Veith FJ, Gupta SK, Panetta TF. 

(1992) J Vase Surg 15:143-9; discussion 150-1. 



10 



Chronic Venous Insufficiency, 
Varicose Veins, Lymphedema, and 
Arteriovenous Fistulas 

Andrew W. Bradbury and Peter J. Pappas 




Chronic Venous Insufficiency 

Chronic venous insufficiency (CVI) may be 
denned as symptom or signs of ambulatory 
venous hypertension. In developed countries, 
CVI affects up to half of the adult population. 
Furthermore, the treatment of CVI consumes 
up to 2% of total health spending and is a major 
cause of lost economic productivity. These 
startling data, coupled with the ineffectiveness 
of current treatment modalities in many of the 
most severely affected patients, underscore the 
need for more research. 

Classification 

Chronic venous insufficiency has proved 
difficult to classify for the purposes of scientific 
reporting. This has obfuscated attempts to 
directly compare the findings of different epi- 
demiological, pathophysiological, and clinical 
studies. The clinical, etiological, anatomical, and 
pathophysiological (CEAP) classification, pro- 
posed in 1994 by the American Venous Forum, 
is now the most widely accepted system 
(Table 10.1). 

Epidemiology 

In industrialized countries the lifetime risks of 
developing varicose veins (VVs), skin changes 
(corona phlebectatica, lipodermatosclerosis, 
varicose eczema, atrophie blanche), and chronic 



venous ulceration (CVU) are 30% to 50%, 5% to 
10%, and 1% to 2%, respectively. The bulk of 
advanced disease affects the elderly, with up to 
5% of women over the age of 65 years having a 
history of CVU. However, up to 50% of affected 
patients, especially men, develop their ulcer 
before their 50th birthday. Women often relate 
the development of VVs to pregnancy and child- 
birth. The increase in female sex hormones and 
blood volume during the first trimester may be 
responsible. However, there is little evidence of 
an association with (multi)parity, and men and 
women appear to be affected almost equally by 
CVI. The excess of women observed in clinical 
practice is mainly due to their longevity and the 
reluctance of men to seek medical attention. 
There is no clear evidence that low socioeco- 
nomic class predisposes to CVI, although CVU 
healing and recurrence rates may be worse. 
Clinical experience suggests that occupations 
involving prolonged standing are associated 
with an increased prevalence and severity of 
CVI, poor ulcer healing, and increased recur- 
rence rates. Data on the relationships between 
physical activity and CVI are conflicting, but it 
seems reasonable to assume that an individual 
with a well-developed calf muscle pump is less 
likely to develop CVI. Although a consistent 
relationship between weight and height is 
lacking, VVs appear to be commoner in tall men 
and CVU in obese women. Similarly, there is 
growing evidence of a hereditary predisposition 
to CVI. For example, patients whose parents 
both have VVs have a 90% chance of developing 



105 




106 



VASCULAR SURGERY 



Table 10.1. Clinical, etiological, anatomic, and 
pathophysiological (CEAP) classification 



Clinical 1 




Class 


No visible or palpable signs of venous 




disease 


Class 1 


Telangiectasia 2 or reticular veins 3 


Class 2 


Varicose veins 4 


Class 3 


Edema 


Class 4 


Skin changes (lipodermatosclerosis, 




atrophie blanche, eczema) 


Class 5 


Healed ulceration 


Class 6 


Active ulceration 


Etiological 




Ec 


Congenital (may be present at birth or 




recognized later) 


Ep 


Primary (with undetermined cause) 


Es 


Secondary (with known cause): 




postthrombotic, posttraumatic, 




other 


Anatomical 




A S 


Superficial veins (numbered 1 to 5) 5 


A D 


Deep veins (numbered 6 to 16) 6 


A P 


Perforating veins (numbered 17 and 




18) 


Pathophysiological 


P 


Reflux 


Po 


Obstruction 


Pr,o 


Both 



1 Supplemented with (A) for asymptomatic or (5) for symptomatic, 
e.g,C M . 

2 Intradermal venules up to 1 mm in diameter. 

3 Subdermal, nonpalpable venules up to 4mm. 

4 Palpable subdermal veins usually larger than 4 mm. 

5 Telangiectasia/reticular veins (1); greater (long) saphenous vein 
above (2) below (3) knee; lesser (short) saphenous vein (4); non- 
saphenous (5). 

6 Inferior vena cava (6); common (7), internal (8), external (9) iliac; 
pelvic (10); common (11), deep (12), superficial (13) femoral; 
popliteal (14); crural (15); muscular (16). 



VVs, and CVU patients have a higher prevalence 
of inherited thrombophilia (TP). The influence 
of race and ethnicity is unclear, as there are few 
reliable data from nonwhite populations. 

Normal Venous Function 

Venous blood from the lower limbs returns to 
the right heart against gravity through the deep 
and superficial venous systems. The deep veins 
follow the named arteries and are often paired. 
The superficial system comprises the long 



saphenous vein (LSV) and short saphenous 
vein (SSV) and their tributaries. As there are 
numerous communications between the long 
and short saphenous systems, and between the 
superficial and deep systems through junctional 
and nonjunctional perforators, these three ele- 
ments are highly interdependent, both anatom- 
ically and functionally, in health and in disease. 
Most of the blood draining into the superficial 
veins from skin and subcutaneous tissues 
immediately enters the deep venous system via 
perforators in the foot, calf, and thigh. In healthy 
subjects, less that 10% of the total venous return 
from the lower limb passes through the LSV and 
SSV to the saphenofemoral junction (SFJ) and 
saphenopopliteal junction (SPJ), respectively. 
Blood is forced back up the leg during leg 
muscle systole, and prevented from flowing 
back down the leg under the influence of gravity 
during diastole, through the actions of the 
muscle pumps and closure of venous valves, 
respectively. The act of walking sequentially 
compresses venous sinuses in the sole of the 
foot, the calf (soleus, gastrocnemius), and to a 
lesser extent the thigh and buttock. During 
relaxation these sinuses fill from the deep and 
superficial venous systems and valves close in 
the superficial and axial veins to prevent reverse 
flow (reflux). In both the superficial and deep 
systems, the density of valves is greatest in the 
calf and gradually diminishes in the thigh. The 
iliac veins and inferior vena cava are frequently 
devoid of valves. 

When standing completely motionless, with 
all the leg muscles relaxed, the venous valve 
leaflets come to lie in a neutral midposition. As 
a result, the venous pressure in the dorsal foot 
veins comes to represent the hydrostatic pres- 
sure exerted by the unbroken column of venous 
blood stretching up from the foot to the right 
atrium (approximately 90 to lOOmmHg in a 
person of average height). Contraction of the leg 
muscles immediately leads to the compression 
of deep veins and sinuses and to the movement 
of venous blood cranially Retrograde blood 
flow is terminated by valve closure, and perfo- 
rators that allow unidirectional flow from the 
superficial to the deep venous system only. 
Conventionally, this has also been ascribed to 
the closure of valves within the perforators. 
However, several studies have shown that many 
perforators are devoid of valves. Instead, 
outward flow through perforators may be 



107 



CHRONIC VENOUS INSUFFICIENCY, VARICOSE VEINS, LYMPHEDEMA, AND ARTERIOVENOUS FISTULAS 




limited by external compression from contract- 
ing muscle and a pinch-cock mechanism involv- 
ing the deep fascia. The importance of these 
mechanisms is that the very high pressures 
(up to 200mmHg) generated within the calf 
muscle pump are used exclusively to propel 
blood back up the leg against gravity, and are 
not transmitted to the superficial or distal deep 
systems. When the muscle pump relaxes, the 
previously expelled venous blood tends to flow 
caudally under gravity but is prevented from 
doing so by valve closure. This has the effect of 
dividing a single long (and heavy) column of 
blood into a series of shorter columns lying 
between closed valves. The pressure within each 
of these segments is low and the ambulatory 
venous pressure (AVP) in the dorsal foot veins 
falls typically to <25mmHg. During muscle 
pump diastole, blood in the superficial system 
flows in to the deep system along a pressure 
gradient. 

Pathophysiology 

There are three basic mechanisms that lead to 
raised AVP and the symptoms and signs of CVT: 
(1) muscle pump dysfunction, (2) valvular 
reflux, and (3) venous obstruction. 

Aging, general debility, and a wide range 
of musculoskeletal or neurological lower limb 
pathologies can impair calf muscle pump func- 
tion. The "fixed" ankle secondary to arthritis or 
trauma is a common example. Muscle bulk and 
tone are also important factors in the mainte- 
nance of perforator competence (see above). 
Failure of perforator competence leads to calf 
pump inefficiency (akin to mitral regurgita- 
tion), as well as the transmission of high pres- 
sures directly to the skin of the gaiter area. 

Reflux is present in more than 90% of patients 
with CVI; 5% to 10% have isolated deep, 30% to 
50% isolated superficial, and 50% to 60% com- 
bined. In general, superficial reflux has a better 
prognosis than deep reflux (especially when the 
latter is postphlebitic), and proximal reflux has 
a better prognosis than distal reflux. Valvular 
reflux can arise in two ways that are not mutu- 
ally exclusive in any one patient: 

Primary valvular incompetence (PVI): A loss 
of elastin and collagen in the vein wall 
around the valve commissures leads to 
dilatation, separation of the valve leaflets, 



and reflux. As the vein dilates, the tension 
in the wall increases according to the law of 
Laplace, which leads to further dilatation. 
The end result is an incompetent, elongated 
and tortuous varicose vein. Primary valvu- 
lar incompetence may also affect the deep 
venous system. 
Postthrombotic syndrome (PTS): Approxi- 
mately 25% of CVU patients have a clear 
history of deep venous thrombosis (DVT), 
and many more have probably suffered a 
subclinical or undiagnosed thrombosis. 
Deep venous thrombosis leads to endothe- 
lial hypoxia, valvular destruction, and 
mural inflammation. Even though most 
DVTs recanalize, the end result is a thick- 
ened, valveless tube that permits gross 
reflux and poses an anatomical (narrow- 
ing, fibrous webs) and functional (lack 
of compliance) obstruction to venous 
outflow. Obstruction leads to the formation 
of collateral pathways. For example, blood 
may be forced out of the calf via perfora- 
tors into the superficial venous system and 
thence up the leg with the formation of 
secondary VVs. Removal of such VVs 
increases AVP. Most patients with severe 
and intractable CVU have PTS. 

Clinical Assessment 

History 

Inquiry should be made as to the duration of the 
present ulcer as well as the duration of ulcer 
disease, the number of episodes, and any pre- 
cipitating factors (Table 10.2). Previous treat- 
ment history and contact allergies are recorded. 
Peripheral artery disease (20%), diabetes melli- 
tus (5%), and rheumatoid arthritis (8%) often 
coexist. Malignancy must not be overlooked. 
Many patients with lower limb symptoms, and 
who coincidentally have VVs, have other pathol- 
ogy to explain their symptoms. Orthopedic, 
neurological, and arterial causes of leg symp- 
toms must be excluded. Particular attention 
must be paid to a history of "white leg" of preg- 
nancy, prolonged immobilization, phlebitis, and 
major lower limb fracture, any of which may 
suggest previous the PTS. A family history of 
venous disease, particularly early-onset, recur- 
rent, or unusual thrombotic events, should be 
sought (Cornu-Thenard, 1994). 




> 



108 



VASCULAR SURGERY 



Table 10.2. Distinguishing 


features of arterial and venous ulcers 




Clinical features 


Arterial ulcer 


Venous ulcer 


Gender 




Men > women 


Women > men 


Age 




Usually presents >60 years 


Typically develops at 40-60 years but patient may 
not present for medical attention until much 
older; multiple recurrences are the norm 


Risk factors 




Smoking, diabetes, hyperlipidemia and 
hypertension 


Previous DVT, thrombophilia, varicose veins 


Past medical 


history 


Most have a clear history peripheral, 


More than 20% have clear history of DVT, many 






coronary, and cerebrovascular disease 


more have a history suggestive of occult DVT, 
e.g., leg swelling after childbirth, hip/knee 
replacement or long bone fracture 


Symptoms 




Severe pain is present unless there is 


About a third have pain but it is not usually 






(diabetic) neuropathy, pain may be 


severe and may be relieved on elevation 






relieved by dependency 




Site 




Normal and abnormal (diabetics) 


Medial (70%), lateral (20%) or both malleoli and 






pressure areas (malleoli, heel, 


gaiter area 






metatarsal heads, fifth metatarsal 








base) 




Edge 




Regular, punched-out, indolent 


Irregular, with neoepithelium (whiter than mature 
skin) 


Base 




Deep, green (sloughy) or black (necrotic) 


Pink and granulating but may be covered in 






with no granulation tissue, may 


yellow-green slough 






comprise major tendon, bone and joint 




Surrounding 


skin 


Features of SLI 


LDS, varicose eczema, atrophie blanche 


Veins 




Empty, guttering on elevation 


Full, usually varicose 


Swelling 




Usually absent 


Often present 



Symptoms 

Localized discomfort in the leg: Usually at the 
site of the visible VV, particularly after pro- 
longed standing. Prominent varices may 
be tender, particularly in menstruating 
women. 

Pain: Severe pain is unusual and suggests 
infection or arterial insufficiency. 

Swelling: A feeling of swelling is common. 

Venous claudication: This is unusual and due 
to extensive postthrombotic iliofemoral 
venous occlusion. There is bursting pain in 
the calf on walking, which is relieved only 
by elevating the leg. In addition, patients 
often complain of heaviness in the calf with 
ambulation. 

Itching: This is common and may lead to 
scratching, infection, and ulceration. 

Physical Examination 

Varicose veins: Note the distribution of 
varices and any surgical scars. 



Corona phlebectatica (ankle/malleolar flare): 
One of the earliest skin manifestations of 
CVI comprises dilated intra/subdermal 
veins at or just below the medial malleolus. 
Overlying skin is thin and fragile leading 
to a blue-bleb appearance. Trauma 
frequently leads to hemorrhage and 
ulceration. 

Lipodermatosclerosis: The skin is brown 
(red or purple) and indurated due to 
hemosiderin and plasma protein deposi- 
tion, leading to dermal fibrosis. 

Atrophie blanche: Thin and pale skin due to 
the thrombotic obliteration of papillary 
capillaries; often at the site of previous 
ulceration. 

Varicose eczema: Scaly dry (or weeping) 
skin that is often intensely pruritic and 
can demonstrate blanching erythema 
(mimicking cellulitis). 

Edema: A common presentation in patients 
with CEAP class 3 or greater CVI. Chronic 
venous insufficiency may coexist with 
other diseases that cause edema, such as 



109 



CHRONIC VENOUS INSUFFICIENCY, VARICOSE VEINS, LYMPHEDEMA, AND ARTERIOVENOUS FISTULAS 




congestive heart failure, and must be 
considered when evaluating CVI patients 
(Table 10.3). 

Hemorrhage: Can be alarming, even life 
threatening, may be spontaneous or follow 
trauma. Direct pressure and elevation 
always arrest venous hemorrhage. As 
recurrent bleeding is almost inevitable, 
the patient should be hospitalized for 
definitive treatment. 

Ulceration: Most CVUs can be easily differen- 
tiated from other forms of ulceration. 

Arterial circulation: If pedal pulses are impal- 
pable, measure the ankle-brachial index 
(ABI). An ABI of <0.8 mandates referral to 
a vascular surgeon. The ABI is unreliable in 
diabetic patients. 

Investigations 

Virtually all patients with CVI require further 
investigation, and duplex ultrasound (DU) has 
largely replaced all other modalities in routine 
clinical practice. It allows Doppler velocity 
information to be color coded and superim- 
posed in real time upon a gray scale (B-mode) 
image. It determines the location and severity of 



Table 10.3. Etiological classification of lymphedema 



Primary 
lymphedema 



Secondary 
lymphedema 



Congenita (onset <2 years old): 

sporadic 
Congenita (onset <2 years old): 

familial (Milroy's disease) 
Praecox (onset 2-35 years): 

sporadic 
Praecox (onset 2-35 years): 

familial (Meige's disease) 
Tarda (onset after 35 years of age) 

Bacterial infection 
Parasitic infection (filariasis) 
Fungal infection (tinea pedis) 
Exposure to foreign-body 

material (silica particles) 
Primary lymphatic malignancy 
Metastatic spread to lymph nodes 
Radiotherapy to lymph nodes 
Surgical excision of lymph nodes 
Trauma (particularly degloving 

injuries) 
Superficial thrombophlebitis 
Deep venous thrombosis 



reflux, the location of the SPJ and nonfunctional 
perforators, and whether the deep veins are 
patent. Plethysmography involves the assess- 
ment of venous function through the measure- 
ment of limb volume and nowadays is primarily 
a research tool. Photo (PPG) and air (APG) 
plethysmography are probably the most 
popular techniques. Ambulatory venous pres- 
sure is measured by cannulating and transduc- 
ing a dorsal foot vein; it remains the research 
reference standard. Ascending venography 
determines the presence of residual thrombus, 
the extent of recanalization,and the distribution 
of collaterals. Contrast medium is injected into 
a dorsal foot vein and directed into the deep 
veins by the placement of an ankle tourniquet. 
The iliac system and the vena cava may not be 
visualized, in which case a separate injection 
can be made in the common femoral vein (CFV) 
(cavography). Descending venography involves 
injecting contrast medium into the CFV with 
the subject positioned at 60 degrees with the 
head up in order to assess reflux. Venography is 
largely reserved for patients being considered 
for deep venous reconstruction because it is 
superior to DU in determining the presence and 
extent of the PTS. Ovarian vein reflux and pelvic 
varices can be visualized by placing a catheter 
into the ovarian or internal iliac veins via the 
CFV approach. As well as this imaging being 
diagnostic, it also enables the ovarian vein to be 
embolized in women suffering from pelvic 
congestion syndrome. Ulcers that fail to heal, 
tend to bleed, or have unusual features should 
be biopsied at base and margin under local 
anesthesia. 



Nonsurgical Management 

The mainstay of treatment is compression with 
or without superficial venous surgery in the 
great majority of patients who have CVI due to 
reflux. A small minority of patients who have 
deep venous obstruction may benefit from 
surgical or endovascular reconstruction. There 
is considerable controversy over the role of 
sclerotherapy. 



Dressings 

No particular dressing or topical agent has 
been shown unequivocally to significantly 




» 



110 



VASCULAR SURGERY 



hasten CVU healing. However, they do have 
different physical properties, and the surgeon 
needs to have a basic grasp of the underlying 
science (and art) of wound care. Enzymatic 
agents (e.g., streptokinase-streptodornase) 
undoubtedly digest the constituents of slough. 
However, they are relatively ineffective against 
deep necrosis or hard eschar. There is evidence 
that they speed up healing, and may damage the 
wound environment. Hydrocolloid dressings 
come in many forms, and are generally imper- 
meable to gases, water vapor, and bacteria. They 
produce a moist, acidic, low-oxygen tension 
wound environment that has been shown exper- 
imentally to enhance wound healing. Patients 
like these dressings because they are easy to use, 
and patients can bathe with the dressing in situ. 
The dressings absorb exudate (reducing the fre- 
quency of dressing changes, smell, risks of 
cross-infection, and costs) and may provide 
superior pain relief. However, in randomized 
controlled trials where both treatment arms 
have received equal and adequate compression, 
hydrocolloid dressings have not be shown to 
improve overall healing compared to any other 
dressing. Bead dressings (such as cadexomer 
iodine and dextranomer) comprise hydrophilic, 
polysaccharide materials that absorb large 
amounts of fluid and slough. The former also 
releases iodine in to the wound. Although they 
may speed up desloughing, they have not been 
shown to enhance healing. Paste bandages com- 
prise a plain weave cotton fabric impregnated 
with zinc oxide paste, either alone or with 
calamine, calamine and clioquinol, coal tar, or 
ichthammol. These additives are designed to 
soothe venous eczema but are actually a 
common cause of contract allergy, and patch 
testing is recommended. Paste bandages do not 
retain moisture, and for this reason, and to 
apply compression, additional layers of bandag- 
ing are required. The Unna boot is a paste 
bandage containing glycerin that hardens into 
semirigid dressing. In trials where equal 
amounts of compression are applied, no form of 
paste bandage has been shown to improve 
healing over other forms of dressing. Their 
principal benefit is provision of inelastic com- 
pression. Alginate dressings absorb exudate and 
create a moist wound environment but have not 
been proved to speed healing. Biological dress- 
ings comprising cultured human epithelium or 
fibroblasts may act as a source of growth factors 



and act as a scaffold for the patient's own epithe- 
lial cells. But they are extremely expensive and 
as yet unproven. 

Topical Agents 

Dermatitis is common and maybe endogenous 
(varicose or venous stasis dermatitis) or 
exogenous due to topically applied substances 
(contact dermatitis). Dermatitis is extremely 
morbid, associated with nonhealing, and maybe 
irritant or allergic due to cell-mediated, delayed 
hypersensitivity. Early patch testing is manda- 
tory. The use of bland paraffin preparations 
greatly reduces the risks of dermatitis. In 
patients with marked exudate, zinc oxide paste 
can be used to protect the surrounding skin. 
Acute dermatitis must be treated with removal 
of the offending allergen and topical steroid 
therapy. Topical antibiotics should be avoided. 

Physical Therapy 

Prolonged bed rest with leg elevation will heal 
virtually all CVUs. However, it is logistically 
impossible, and associated with decubitus com- 
plications, and as it does not address the under- 
lying hemodynamic abnormality, recurrence is 
virtually inevitable. Exercise therapy aimed at 
improving calf muscle pump function maybe of 
benefit and trials are under way. 

Compression Therapy 

Compression undoubtedly retards the develop- 
ment and progression of CVI. However, it is still 
more of an art than a science, and the quality of 
scientific reporting remains low. Elastic band- 
aging (the four-layer bandage) is favored in the 
United Kingdom, whereas in mainland Europe 
and North America inelastic bandaging (the 
Unna boot) is preferred. The four-layer bandage 
comprises orthopedic wool (to protect the bony 
prominences and to absorb any exudates); crepe 
bandage (to compress and shape the wool, and 
to provide a firm base for the compression 
bandages), elastic bandage (e.g., Elset™, Seton, 
applied at 50% stretch) and a self-adhesive elas- 
ticated bandage (e.g., Coban™, 3M to add to 
compression and fix the bandaging in place). 
This bandage typically exerts 40mmHg at the 
ankle and 20mmHg just below the knee. Once 
the ulcer is healed, the patient should be pre- 



111 



CHRONIC VENOUS INSUFFICIENCY, VARICOSE VEINS, LYMPHEDEMA, AND ARTERIOVENOUS FISTULAS 




scribed stocking. There is no evidence that 
extending compression above the knee confers 
benefit. Compliance is a major problem. 

Sclerotherapy 

The role of sclerotherapy is controversial, with 
practitioner's views based largely on profes- 
sional background and country of origin rather 
than on clinical comparative studies. Some scle- 
rotherapists believe they can treat all VVs, but 
most accept the superiority of surgery in the 
presence of main stem, SFJ, or SPJ incompe- 
tence. However, the advent of foam sclerosants 
may revolutionize the management of such 
disease. The aim is to place a small volume of 
sclerosant in the lumen of a vein empty of 
blood, and then appose the walls of that vein 
with appropriate compression. The vein then 
fibroses closed without the formation of clot. 
Some practitioners use magnifying loupes for 
smaller veins, and there is increasing interest in 
injecting larger veins under ultrasound guid- 
ance (echosclerotherapy). The vein must be kept 
empty of blood both during and after the injec- 
tion to prevent thrombophlebitis. Adequate 
compression is difficult in the perineum, upper 
thigh, and popliteal fossa, especially in the 
obese. Patients should be mobilized immedi- 
ately afterward. In the U.K., most surgeons use 
detergents that act by directly damaging the 
endothelium. In mainland Europe and North 
America, hypertonic saline is also popular. Err 
on the side of caution with regard to volume and 
concentration until the patient's response can 
be assessed. The complications of injection 
sclerotherapy include anaphylaxis (<0.1%), 
allergic reactions (uncommon), ulceration 
(extr avascular injection), arterial injection (rare 
and serious), pigmentation (extravasation), 
superficial thrombophlebitis (inadequate com- 
pression), and DVT (inadequate mobilization). 

Surgical Management 

There is growing evidence that saphenous 
surgery improves the quality of life in patients 
with VVs, and augments the healing and 
reduces the recurrence of CVU better than com- 
pression alone (Dwerryhouse et al., 1999). For 
optimal results, it is necessary to define the 
extent and severity of venous disease, usually by 
means of DU, prior to surgery. Surgery for CVU 



is different from that for uncomplicated VVs in 
a number of important ways. The patients are 
older and often have multisystem, medical 
comorbidity; the risks, especially DVT, are 
higher. Patients may require inpatient optimiza- 
tion of cardiorespiratory function, treatment of 
dermatitis, edema reduction, and desloughing 
of the ulcer. The effect of deep venous reflux on 
the efficacy of superficial venous surgery is con- 
troversial and incompletely defined. Deep reflux 
due to PVI may reverse once superficial reflux 
has been eradicated. However, most agree that 
patients with extensive PTS gain less benefit 
from surgery. Secondary VVs that are acting as 
collaterals must not be removed. Although post- 
operative compression therapy has been shown 
to reduce VVs and CVU recurrence, compliance 
is poor. 

Varicose Vein Surgery 

Long Saphenous Surgery 

Safe and effective surgery depends on observing 
a few sound principles. In a patient of normal 
build the SFJ lies directly beneath the groin 
crease; in the obese it lies above. An incision 
made below the crease is likely to be too low. 
Resist the temptation to operate through an 
excessively small incision. Do not divide any 
vein until the SFJ has been unequivocally iden- 
tified. Unless all tributaries are taken beyond 
secondary branch points, a network remains of 
superficial veins connecting the veins of the 
thigh with those of the perineum, the lower 
abdominal wall, and the iliac region. These 
cross-groin connections are a frequent cause of 
recurrence. Ligate the LSV deep to all tributar- 
ies flush with the CFV using nonabsorbable 
transfixion suture to reduce neovascularization 
through the stump. Directly ligate, and if large, 
consider stripping, any high anterolateral or 
posteromedial or thigh branches to reduce 
hematoma formation and recurrence. There is 
evidence to show that stripping the LSV to a 
hand's breadth below the knee significantly 
reduces recurrence by disconnecting the thigh 
perforators and saphenous tributaries and by 
removing the conduit that will allow neovascu- 
larization in the groin to reconnect with the 
remaining superficial venous system of the 
thigh and calf. Confining stripping to just below 
the knee, and to a downward direction, reduces 




» 



112 



VASCULAR SURGERY 



saphenous nerve injury. The theoretical advan- 
tages of invagination stripping in terms of 
reducing blood loss, hematoma, nerve injury, 
and scars have not been confirmed in trials. 

Short Saphenous Surgery 

Saphenopopliteal junction ligation can prove to 
be a challenging procedure, especially when 
performed for recurrent disease. Always mark 
the junction preoperatively with DU (some 
surgeon still prefer venography). The SSV can 
be found by following the Giacomini vein, which 
is a superficially placed tributary of the SSV that 
runs up the thigh to join the LSV. This may be 
large and confused with the SSV, especially if the 
SPJ is absent; the importance of this will be 
apparent on the preoperative DU. Be aware that 
traction on the SSV can tent up and damage the 
mobile and thin-walled popliteal vein. Palpation 
of the artery gives an indication of the depth of 
dissection. Beware the common peroneal nerve 
under medial edge of biceps femoris, which is 
at risk from overzealous lateral retraction as 
well as from a careless stitch when closing the 
popliteal fascia. The SSV is often closely associ- 
ated with sural nerve injury and is also at risk, 
particular if stripping is undertaken. There are 
still a number of controversial issues regarding 
the SSV. Is it always necessary to ligate the SSV 
flush with the popliteal vein? Experience sug- 
gests that this counsel of perfection is hard to 
achieve in a significant proportion of patients 
without risk of collateral damage. This raises the 
question of what should be done with the gas- 
trocnemius and other, often large and refluxing, 
muscular veins? Should the SSV be stripped or 
is it permissible just to remove a segment 
through the popliteal fossa wound? It seems 
likely, although there is no proof, that SSV strip- 
ping, for the same reasons as for the LSV, would 
reduce recurrence. However, there is concern 
about sural nerve injury. 

Perforator Ligation 

Although the advent of DU-guided subfascial 
endoscopic perforator surgery (SEPS) has 
rekindled interest in perforator ligation, there is 
no evidence that it alters the natural history of 
venous disease. Clearly, the only way to resolve 
this issue once and for all is to perform a 



randomized controlled trial of compression vs. 
compression and saphenous surgery vs. com- 
pression, saphenous surgery, and SEPS. 

Surgery for Recurrent Veins 

Recurrent LSV VVs arise because of inadequate 
dissection of, or neovascularization at, the SFJ in 
the presence of a nonstripped or incompletely 
stripped LSV. Standard teaching is to approach 
the SFJ through nonoperated tissues (usually 
from a lateral approach that first exposes the 
common femoral artery) so that the CFV can be 
skeletonized of branches using nonabsorbable 
sutures for 1 to 2 cm above and below the junc- 
tion. The top of the LSV is dissected from the 
mass of scar tissue so that it can be stripped. 
However, this can be a difficult and potentially 
morbid operation. When the preoperative DU 
indicates neovascularization as opposed to an 
intact SFJ, the LSV can be located at the knee, a 
stripper passed up toward the groin, and the 
vein stripped without a formal redissection. 

Complications 

Fortunately, major complications following VV 
surgery are relatively rare. However, up to 20% 
of patients may suffer some form of minor mor- 
bidity, such as hematoma, lymphatic leak, pain, 
saphenous neuritis, and venous thrombosis. In 
the U.K., VV surgery is the commonest cause of 
litigation against general and vascular surgeons. 
This not a field for the unsupervised, inexperi- 
enced surgeon and it behooves surgeons who 
undertake VV surgery to carefully audit their 
management, techniques, and outcomes. 

Deep Venous Reconstruction 

These procedures have not gained widespread 
acceptance largely because there is little data to 
support their efficacy. Several different tech- 
niques have been described for suturing the 
edges of "floppy" valve cusps to the vein wall, 
rendering the valve competent. Autologous 
valve transplantation interposes a segment of 
axillary or brachial vein, containing a competent 
valve, into an incompetent deep vein, usually the 
popliteal. Procedures using synthetic, mixed, 
and animal valves are still experimental. An 
incompetent superficial femoral vein can be 
transected and anastomosed end to end or end 



113 



CHRONIC VENOUS INSUFFICIENCY, VARICOSE VEINS, LYMPHEDEMA, AND ARTERIOVENOUS FISTULAS 




to side to a profunda femoris or long saphenous 
vein that has a competent valve (vein transposi- 
tion). An obstructed femoral segment may be 
bypassed by anastomosing a transected, compe- 
tent LSV to the side of the popliteal vein. Again, 
satisfactory long-term patency rates have been 
reported in small series. 

Conclusions 

Although the most difficult cases of leg ulcera- 
tion are multifactorial in origin, CVI is the 
single most common underlying pathology. As 
such, there is hope that the prevalence of CVU 
may decline in the future as a result of improved 
thromboembolic prophylaxis and treatment. 
For the moment, however, CVU is a common 
and disabling condition that is often resistant to 
conservative therapy, prone to recurrence, and 
very expensive to manage. There needs to be a 
low threshold for referral to a vascular surgeon, 
preferably through a one-stop assessment clinic 
where a thorough venous and arterial duplex- 
based assessment can be performed. This will 
enable patients who might benefit from surgical 
intervention to be identified and treated early. It 
will also enable ongoing outpatient treatment to 
be based on an in-depth understanding of the 
pathophysiological mechanisms responsible in 
each patient. Great progress in the management 
of CVU has been made over the last decade 
because of an increased understanding of the 
pathophysiology and the availability of data 
from clinical trials that have provided a 
scientifically robust platform on which to base 
treatment algorithms. Despite all this, further 
research is required into the epidemiology and 
natural history of CVU, models of care, primary 
prevention, and pathogenesis. 

Lymphedema 

The Lymphatic System 

The lymphatic system performs the following 
functions: 

1. It removes water, electrolytes, low- 
molecular-weight moieties (polypeptides, 
cytokines, growth factors), and macromol- 
ecules (fibrinogen, albumen, globulins, 
coagulation and fibrinolytic factors) from 



the interstitial fluid (ISF) and returns them 
to the circulation. 

2. It permits the circulation of lymphocytes 
and other immune cells. 

3. It returns intestinal lymph (chyle), which 
transports cholesterol, long chain fatty 
acids, triglycerides, and the fat-soluble 
vitamins (A, D, E, and K), directly to the 
circulation, bypassing the liver. 

Lymph from the lower limbs and abdomen 
drains via the cisterna chyli and thoracic duct 
into the left internal jugular vein at its 
confluence with the left subclavian vein. Lymph 
from the head and right arm drains via the right 
lymphatic duct into the right internal jugular 
vein. Lymphatics accompany veins everywhere 
except in the cortical bony skeleton and central 
nervous system, although the brain and retina 
possess cerebrospinal fluid and aqueous humor, 
respectively. The lymphatic system comprises 
lymphatic channels, lymphoid organs (lymph 
nodes, spleen, Peyer's patches, thymus, tonsils), 
and circulating elements (lymphocytes and 
other mononuclear immune cells). 

Lymphatics originate within the ISF space 
from specialized endothelialized capillaries 
(initial lymphatics) or nonendothelialized 
channels such as the spaces of Disse in the liver. 
Initial lymphatics are unlike arteriovenous cap- 
illaries in that they are blind-ended, are much 
larger (50 |J.m) and allow the entry of molecules 
up to 1000 kd in size. This is because the 
basement membrane of these lymphatics is 
fenestrated, tenuous, or lacking intra- and 
intercellular endothelial pores. Lymphatic capil- 
laries are anchored to interstitial matrix by 
filaments. In the resting state they are collapsed, 
but when ISF volume and pressure increases, 
they are held open by these filaments to facili- 
tate increased drainage. Initial lymphatics drain 
into terminal (collecting) lymphatics that 
possess bicuspid valves and endothelial cells 
rich in the contractile protein actin. Larger col- 
lecting lymphatics are surrounded by smooth 
muscle. Valves partition the lymphatics into seg- 
ments (lymphangions) that contract sequen- 
tially in order to propel lymph into the lymph 
trunks. Terminal lymphatics lead to lymph 
trunks comprising endothelium, basement 
membrane, and a media of smooth muscle 
cells that are innervated with sympathetic, 




> 



114 



VASCULAR SURGERY 



parasympathetic, and sensory nerve endings. 
About 10% of lymph arising from a limb is 
transported in deep lymphatic trunks that 
accompany the main neurovascular bundles. 
The majority of lymphatic flow, however, is con- 
ducted against the venous flow from deep to 
superficial in epifascial lymph trunks. Super- 
ficial trunks form lymph bundles of various 
sizes, are located within strips of adipose tissue, 
and tend to follow the course of the major 
superficial veins. 

The distribution of fluid and protein between 
the vascular and ISF spaces depends on hydro- 
static and oncotic pressures (Starling's forces), 
together with the relative impermeability of the 
blood capillary membrane to molecules over 
70 kd. In healthy subjects there is net capillary 
filtration, which is removed by the lymphatic 
system. Small particles enter the initial lym- 
phatics directly; larger particles are phagocy- 
tosed by macrophages and transported through 
the lymphatic system intracellularly Lymph 
flows against a small pressure gradient due to 
transient increases in interstitial pressure sec- 
ondary to muscular contraction and external 
compression, the sequential contraction and 
relaxation of lymphangions, and the prevention 
of reflux due to valves. Lymphangions respond 
to increased lymph flow in much the same way 
as the heart responds to increased venous 
return in that they increase their contractility 
and stroke volume. Transport in the main lymph 
ducts also depends on intrathoracic (respira- 
tion) and central venous (cardiac cycle) pres- 
sures. In the healthy limb, lymph flow is largely 
due to intrinsic lymphatic contractility aug- 
mented by exercise, limb movement, and exter- 
nal compression. However, in lymphedema, 
where the lymphatics are constantly distended 
with lymph, these external forces assume a 
much more important functional role. 

Definition and Pathophysiology 

Lymphedema maybe defined as abnormal limb 
swelling due to the accumulation of increased 
amounts of high-protein ISF secondary to 
defective lymphatic drainage in the presence of 
(near) normal net capillary filtration (Szuba and 
Rockson, 1997). In order for edema to be clini- 
cally detectable, the ISF volume has to double. 
About 8L of lymph is produced and, following 
resorption in lymph nodes, about 4L enters the 



venous circulation. In one sense, all edema is 
lymphedema in that it results from an inability 
of the lymphatic system to clear the ISF com- 
partment. However, in most types of edema this 
is because the capillary filtration rate is patho- 
logically high and overwhelms a normal lym- 
phatic system, resulting in the accumulation of 
low-protein edema fluid. By contrast, in true 
lymphedema, capillary filtration is normal and 
the edema fluid is relatively high in protein. 
Both mechanisms frequently coexist, as in 
patients with CVI. 

Lymphedema can result from lymphatic 
aplasia, hypoplasia, dysmotility (reduced con- 
tractility with or without valvular insuffi- 
ciency), obliteration by inflammatory, infective 
or neoplastic processes, or surgical extirpation 
(Table 10.4). Whatever the primary abnormality, 
the resultant physical or functional obstruction 
leads to lymphatic hypertension and distention 
with further secondary impairment of contrac- 
tility and valvular competence. Lymphostasis 
and lymphotension lead to the accumulation in 
the ISF of fluid, proteins, growth factors and 
other active peptide moieties, glycosaminogly- 
cans, and particulate matter, including bacteria. 
As a consequence, there is increased collagen 
production by fibroblasts, an accumulation 
of inflammatory cells (predominantly macro- 
phages and lymphocytes), and activation of ker- 
atinocytes. The end result is protein-rich edema 
fluid, increased deposition of ground substance, 
subdermal fibrosis, and dermal thickening and 
proliferation. Lymphedema, unlike all other 
types of edema, is confined to the epifascial 
space. Although muscle compartments may 
be hypertrophied due to the increased work 
involved in limb movement, they are character- 
istically free of edema. 

Two main types of lymphedema are 
recognized: 

• Primary, in which the cause is unknown 
(or at least uncertain and unproved) but 
often presumed to be due to congenital 
lymphatic dysplasia 

• Secondary, in which there is a clear under- 
lying cause such as inflammation, malig- 
nancy, or surgery 

Primary lymphedema is usually further subdi- 
vided on the basis of the presence of a family 
history, age of onset, and lymphangiographic 
findings (see below). 



115 



CHRONIC VENOUS INSUFFICIENCY, VARICOSE VEINS, LYMPHEDEMA, AND ARTERIOVENOUS FISTULAS 




Table 10.4. Differential diagnosis of the swollen limb 
Nonvascular or lymphatic General disease states 



Local disease processes 



Venous 



Arterial 



Retroperitoneal fibrosis 

Gigantism 

Drugs 



Trauma 
Obesity 

Deep venous thrombosis 
Postthrombotic syndrome 

Varicose veins 

Klippel-Trenaunay 
syndrome and other 
malformations 

External venous 
compression 
Ischemia- reperfusion 

Arteriovenous 

malformation 
Aneurysm 



;e 



Cardiac failure from any cause; liver failure; hypoproteinemia due 
to nephrotic syndrome, malabsorption, protein losing 
enteropathy; hyperthyroidism (myxedema); allergic disorders 
including angioedema and idiopathic cyclic edema; prolonged 
immobility and lower lib dependency 

Ruptured Baker's cyst; myositis ossificans; bony or soft tissue 
tumors; arthritis; hemarthrosis; calf muscle hematoma; 
Achilles tendon rupture 

May lead to arterial, venous and lymphatic abnormalities 

Rare; all tissues are uniformly enlarged 

Corticosteroids; estrogens; progestogens; monoamine oxidase 
inhibitors; phenylbutazone; methyldopa; hydralazine; 
nifedipine 

Painful swelling due to reflex sympathetic dystrophy 

Lipodystrophy, lipoidosis 

There may be an obvious predisposing factor such as recent 
surgery; the classical signs of pain and redness may be absent 

Swelling, usually of the whole leg, due to iliofemoral venous 
obstruction; venous skin changes, secondary varicose veins on 
the leg and collateral veins on the lower abdominal wall; 
venous claudication may be present 

Simple primary varicose veins are rarely the cause of significant 
leg swelling 

Rare; present at birth or develops in early childhood; comprises 
an abnormal lateral venous complex, capillary nevus, bony 
abnormalities, hypo(a)plasia of deep veins, and limb 
lengthening; lymphatic abnormalities often coexist 

Pelvic or abdominal tumor including the gravid uterus; 
retroperitoneal fibrosis 

Following lower limb revascularization for chronic and 
particularly chronic ischemia 

May be associated with local or generalized swelling 






Popliteal; femoral; false aneurysm following (iatrogenic) trauma 



Epidemiology 

Lymphedema is estimated to affect around 2% 
of the population and causes significant physi- 
cal symptoms and complications, as well as 
emotional and psychological distress, which can 
lead to difficulties with relationships, school, 
and work. Many sufferers choose not to seek 
medical advice because of embarrassment and 
a belief that nothing can be done. Patients 
who do come forward, especially those with 
non-cancer-related lymphedema, often find 
they have limited access to appropriate expert- 
ise and treatment. Lymphedema is often mis- 
diagnosed and mistreated by doctors, who 
frequently have a poor understanding of the 
condition, believing it to be primarily a cos- 



metic problem. However, early diagnosis and 
treatment are important because relatively 
simple measures can prevent the development 
of disabling late disease, which is often very 
difficult to treat. 

Clinical Assessment 

In most cases the diagnosis of primary or sec- 
ondary lymphedema can be made, and the con- 
dition differentiated from other causes of a 
swollen limb, on the basis of history and exam- 
ination without recourse to complex investiga- 
tion. Unlike other types of edema, lymphedema 
characteristically involves the foot. The contour 
of the ankle is lost through infilling of the sub- 
malleolar depressions; a "buffalo hump" forms 




> 



116 



VASCULAR SURGERY 



on the dorsum of the foot, the toes appear 
square due to confinement of footwear, and 
the skin on the dorsum of the toes cannot 
be pinched due to subcutaneous fibrosis 
(Stemmer's sign). Lymphedema usually spreads 
proximally to knee level and less commonly 
affects the whole leg. In the early stages, lym- 
phedema "pits," and the patient reports that the 
swelling is down in the morning. This repre- 
sents a reversible component to the swelling, 
which can be controlled. Failure to do so allows 
fibrosis, dermal thickening, and hyperkeratosis 
to occur. In general, primary lymphedema 
progresses more slowly than secondary lym- 
phedema. Chronic eczema, fungal infection of 
the skin (dermatophytosis) and nails (ony- 
chomycosis), Assuring, verrucae, and papillae 
(warts) are frequently seen in advanced disease. 
Ulceration is unusual except in the presence of 
chronic venous insufficiency. 

Lymphangiomas are dilated dermal lymphat- 
ics that blister onto the skin surface. The fluid is 
usually clear but may be blood stained, and in 
the long term they thrombose and fibrose, 
forming hard nodules, raising concerns about 
malignancy. If they are <5 cm across, they 
are termed lymphangioma circumscriptum; if 
more widespread, lymphangioma diffusum. If 
they form a reticulate pattern of ridges, they are 
termed lymphedema ab igne. Lymphangiomas 
frequently weep (lymphorrhea, chylorrhea), 
causing skin maceration and act as a portal 
for infection. Protein-losing diarrhea, chylous 
ascites, chylothorax, chyluria, and discharge 
from lymphangiomas suggest lymphangiectasia 
(megalymphatics) and chylous reflux. 

Ulceration, nonhealing bruises, and raised 
purple-red nodules should lead to suspicion of 
malignancy. Lymphangiosarcoma was origi- 
nally described in postmastectomy edema 
(Stewart-Treves syndrome) and affects about 
0.5% of patients at a mean onset of 10 years. 
However, lymphangiosarcoma can develop in 
any long-standing lymphedema but usually 
takes longer to manifest (20 years). It presents 
as single or multiple bluish/red skin and subcu- 
taneous nodules that spread to form satellite 
lesions that may then become confluent. The 
diagnosis is usually made late, and confirmed 
by skin biopsy. Amputation offers the best 
chance of survival, but even then most patients 
live less than 3 years. It has been suggested 
that lymphedema leads to an impairment 



of immune surveillance and so predisposes 
to other malignancies, although the causal 
association is not as definite as it is for 
lymphangiosarcoma. 

Primary Lymphedema 

It has been proposed that all cases of primary 
lymphedema are due to an inherited abnormal- 
ity of the lymphatic system, sometimes termed 
congenital lymphatic dysplasia. However, it is 
possible that many sporadic cases of primary 
lymphedema occur in the presence of a (near) 
normal lymphatic system and are actually 
examples of secondary lymphedema for which 
the triggering events have gone unrecognized. 
These might include seemingly trivial (but 
repeated) bacterial or fungal infections, insect 
bites, barefoot walking (silica), DVT, or episodes 
of superficial thrombophlebitis. In animal 
models, simple excision of lymph nodes or 
trunks leads to acute lymphedema that resolves 
within a few weeks, presumably due to collater- 
alization. In animals, the human condition can 
only be mimicked by inducing extensive lym- 
phatic obliteration and fibrosis. Even then, there 
may be considerable delay between the injury 
and the onset of edema. Primary lymphedema 
is much commoner in the legs than the arms. 
This may be due to gravity and a bipedal 
posture, the fact that the lymphatic system of 
the leg is less well developed, or the increased 
susceptibility of the leg to trauma or infection. 
Furthermore, loss of the venoarteriolar reflex 
(VAR), which protects lower limb capillaries 
from excessive hydrostatic forces in the erect 
posture, with age and disease (CVI, diabetes) 
may be important. 

Primary lymphedema is often classified on 
the basis of apparent genetic susceptibility, age 
of onset, or lymphangiographic findings (Table 
10.5). None of these is ideal, and the various 
classification systems in existence can appear 
confusing and conflicting as various terms and 
eponyms are used loosely and interchangeably. 
This problem has hampered research and 
efforts to gain a better understanding of under- 
lying mechanisms, the effectiveness of therapy, 
and prognosis. Primary lymphedema, where 
there appears to be a genetic susceptibility or 
element to the disease, may be further divided 
into those cases that are familial (hereditary), 
where typically the only abnormality is lym- 



117 



CHRONIC VENOUS INSUFFICIENCY, VARICOSE VEINS, LYMPHEDEMA, AND ARTERIOVENOUS FISTULAS 




Table 10.5. Lymphangiography classification of primary lymphedema 





Congenital hyperplasia 


Distal obliteration 


Proximal obliteration 




(10%) 


(80%) 


(10%) 


Age of onset 


Congenital 


Puberty (praecox) 


Any age 


Sex distribution 


Male > female 


Female > male 


Male = female 


Extent 


Whole leg 


Ankle, calf 


Whole leg, thigh only 


Laterality 


Uni = bilateral 


Often bilateral 


Usually unilateral 


Family history 


Often positive 


Often positive 


No 


Progression 


Progressive 


Slow 


Rapid 


Response to compression 


Variable 


Good 


Poor 


therapy 








Comments 


Lymphatics are increased 


Absent or reduced distal 


There is obstruction at the 




in number, although 


superficial lymphatics; 


level of the aortoiliacor 




functionally defective; 


also termed aplasia or 


inguinal nodes; if associated 




there is usually an 


hypoplasia 


with distal dilatation, the 




increased number of 




patient may benefit from 




lymph nodes; may have 




lymphatic bypass 




chylous ascites, 




operation; other patients 




chylothorax,and 




have distal obliteration as 




protein-losing 




well 




enteropathy 







phedema and there is a family history, and those 
cases that are syndromic, where the lym- 
phedema is only one of several congenital 
abnormalities and is either inherited or spo- 
radic. Syndromic lymphedema maybe sporadic 
and chromosomal [Turner's (XO karyotype), 
Klinefelter's (XXY), Down (trisomy 21) syn- 
drome], or clearly inherited and related to an 
identified or presumed single gene defect 
[lymphedema-distichiasis (autosomal domi- 
nant)], or of uncertain genetic etiology (yellow- 
nail and Klippel-Trenaunay- Weber syndromes). 
Familial (hereditary) lymphedema can be 
difficult to distinguish from nonfamilial lym- 
phedema because a reliable family history may 
be unobtainable, the nature of the genetic pre- 
disposition is unknown, and the genetic 
susceptibility may translate into clinical disease 
only in the presence of certain environmental 
factors. Although the distinction may not 
directly affect treatment, the patients are often 
concerned lest they be passing on the disease 
to their children. Two main forms of familial 
(hereditary) lymphedema are recognized: 
Noone-Milroy (type I) and Letessier-Meige 
(type II). It is likely that both eponymous dis- 
eases overlap and represent more than a single 
disease entity and genetic abnormality. Milroy's 
disease is estimated to be present in 1 in 6000 
live births and is probably inherited in an 



autosomal-dominant manner with incomplete 
(about 50%) penetrance. In some families, the 
condition maybe related to abnormalities in the 
gene coding for a vascular endothelial growth 
factor (VEGF) on chromosome 5. The disease is 
characterized by brawny lymphedema of both 
legs (and sometimes the genitalia, arms, and 
face) that develops from birth or before puberty. 
The disease has been associated with a wide 
range of lymphatic abnormalities on lymphan- 
giography. Meige's disease is similar to Milroy's 
disease except the lymphedema generally devel- 
ops between puberty and middle age (50 years). 
It usually affects one or both legs but may 
involve the arms. Some, but not all, cases appear 
to be inherited in an autosomal-dominant 
manner. Lymphangiography generally shows 
aplasia or hypoplasia. 

Lymphedema congenita (onset at or within 2 
years of birth) is commoner in males, more 
likely to be bilateral and to involve the whole leg. 
Lymphedema praecox (onset from 2 to 35 years) 
is three times commoner in females, has a peak 
incidence shortly after menarche, is three times 
more likely to be unilateral than bilateral, and 
usually only extends to the knee. Lymphedema 
tarda develops, by definition, after the age of 35 
years and is often associated with obesity, with 
lymph nodes being replaced by fibrofatty tissue. 
The cause is unknown. Lymphedema develop- 




> 



118 



VASCULAR SURGERY 



ing for the first time after 50 years should 
prompt a thorough search for underlying 
(pelvic, genitalia) malignancy. It is worth noting 
that, in such patients, lymphedema often com- 
mences proximally in the thigh rather than 
distally 

Secondary Lymphedema 

This is the most common form of lymphedema. 
There are several well-recognized causes, in- 
cluding infection, inflammation, neoplasia, and 
trauma. 

Filariasis is the commonest cause of lym- 
phedema worldwide, affecting up to 100 million 
individuals. It is particularly prevalent in Africa, 
India, and South America, where 5% to 10% 
of the population may be affected. The vivipa- 
rous nematode Wucheria bancrofti, whose only 
host is humans, is responsible for 90% of cases 
and is spread by the mosquito. The disease is 
associated with poor sanitation. The parasite 
enters lymphatics from the blood and lodges 
in lymph nodes, where it causes fibrosis and 
obstruction, due partly to direct physical 
damage and partly to the immune response of 
the host. Proximal lymphatics become grossly 
dilated with adult parasites. The degree of 
edema is often massive, in which case it is 
termed elephantiasis. Immature parasites 
(microfilariae) enter the blood at night and can 
be identified on a blood smear, a centrifuged 
specimen of urine, or in lymph itself. A com- 
plement fixation test is also available and 
is positive in present or past infection. 
Eosinophilia is usually present. Diethylcarba- 
mazine destroys the parasites but does not 
reverse the lymphatic changes, although there 
may be some regression over time. Once the 
infection has been cleared, treatment is as for 
primary lymphedema. Public health measures 
to reduce mosquito breeding, protective cloth- 
ing, and mosquito netting may be usefully 
employed to combat the condition. 

Endemic elephantiasis (podoconiosis) is 
common in the tropics and affects more than 
500,000 people in Africa. The barefoot cultiva- 
tion of soil composed of alkaline volcanic rocks 
leads to destruction of the peripheral lymphat- 
ics by particles of silica, which can be seen in 
macrophages in draining lymph nodes. Plantar 
edema develops in childhood and rapidly 
spreads proximally. The condition is prevented, 



and its progression slowed, by the wearing of 
shoes. 

Lymphangitis and lymphadenitis can cause 
lymphatic destruction that predisposes to 
lymphedema complicated by further acute 
inflammatory episodes (AIEs). Interestingly, in 
such patients lymphangiography has revealed 
abnormalities in the contralateral, unaffected 
limb, suggesting an underlying, possibly inher- 
ited, susceptibility. Lymphatic and lymph node 
destruction by tuberculosis is also a well- 
recognized cause of lymphedema, especially in 
developing countries. 

Treatment (surgery, radiotherapy) for breast 
carcinoma is probably the commonest cause of 
lymphedema in developed countries but is 
decreasing in incidence as surgery becomes 
more conservative. Lymphoma may present 
with lymphedema, as may malignancy of the 
pelvic organs and external genitalia. Kaposi's 
sarcoma developing in the course of human 
immunodeficiency virus (HlV)-related illness 
may cause lymphatic obstruction and is a 
growing cause of lymphedema in certain parts 
of the world. 

It is not unusual for patients to develop 
chromic localized or generalized swelling fol- 
lowing trauma. The etiology is often multifacto- 
rial and includes disuse, venous thrombosis, 
and lymphatic injury or destruction. Degloving 
injuries and burns are particularly likely to 
disrupt dermal lymphatics. Tenosynovitis can 
also be associated with localized subcutaneous 
lymphedema, which can be a cause of trouble- 
some persistent swelling following ankle and 
wrist sprains and repetitive strain injury. 

It is important to appreciate the relationship 
between lymphedema and CVI. As both condi- 
tions are relatively common and often coexist in 
the same patient, it can be difficult to unravel 
which components of the patient's symptom 
complex are due to each pathology. There is no 
doubt that superficial venous thrombophlebitis 
(SVT) and DVT can both lead to lymphatic 
destruction and secondary lymphedema, espe- 
cially if recurrent. Lymphedema is an important 
contributor to the swelling of the postphlebitic 
syndrome. It has also been suggested that lym- 
phedema can predispose to DVT and possibly 
SVT through immobility and AIEs. Certainly, 
tests of venous function (duplex ultrasonogra- 
phy, plethysmography) are frequently abnormal 
in patients with lymphedema. 



119 



CHRONIC VENOUS INSUFFICIENCY, VARICOSE VEINS, LYMPHEDEMA, AND ARTERIOVENOUS FISTULAS 




It is not uncommon to see patients (usually 
women) with lymphedema in whom a duplex 
ultrasound scan has revealed superficial reflux 
(such reflux is present subclinically in up to a 
third of the adult population). Although isolated 
superficial venous reflux rarely, if ever, leads 
to limb swelling, such patients are frequently 
misdiagnosed as having venous rather than 
lymphedema, and mistakenly subjected to VV 
surgery. Not only does such surgery invariably 
fail to relieve the swelling, it usually makes it 
worse as saphenofemoral and saphenopopliteal 
ligation, together with saphenous stripping, 
compromise still further the drainage through 
the subcutaneous lymph bundles (which follow 
the major superficial veins) and draining 
inguinal and popliteal lymph nodes. 

Rheumatoid and psoriatic arthritis (chronic 
inflammation and lymph node fibrosis), contact 
dermatitis, snake and insect bites, and retroperi- 
toneal fibrosis are all rare but well-documented 
causes of lymphedema. Pretibial myxedema is 
due to the obliteration of initial lymphatics by 
mucin. Factitious lymphedema is caused by 
application of a tourniquet (a "rut" and sharp 
cut-off is seen on examination) or "hysterical" 
disuse in patients with psychological problems. 
Generalized or localized immobility due to any 
cause leads to chronic limb swelling that can 
be misdiagnosed as lymphedema. Examples 
include the elderly person who spends all day 
(and sometimes all night) sitting in a chair 
(armchair legs), the hemiplegic stroke patient, 
and the young patient with multiple sclerosis. 

Lipedema presents almost exclusively in 
women and comprises bilateral, usually sym- 
metrical, enlargement of the legs and, some- 
times, the lower half of the body due to the 
abnormal deposition of fat. It may or may not 
be associated with generalized obesity. There 
are a number of features that help to differ- 
entiate the condition from lymphedema, but 
lipedema may coexist with other causes of limb 
swelling. It has been proposed that lipedema 
results from, or at least is associated with, fatty 
obliteration of lymphatics and lymph nodes. 

Investigation 

It is usually possible to diagnose and manage 
lymphedema purely on the basis of the history 
and examination, especially when the swelling is 
mild and there are no apparent complicating 



features. In patients with severe, atypical, and 
multifactorial swelling, investigations may help 
confirm the diagnosis, inform management, and 
provide prognostic information. A full blood 
count, urea and electrolytes, creatinine, liver 
function tests, chest radiograph, and blood 
smear for microfilariae may be indicated. Direct 
lymphangiography involves the injection of 
contrast medium into a peripheral lymphatic 
vessel and subsequent radiographic visualiza- 
tion of the vessels and no des. It remains the gold 
standard for showing structural abnormalities 
of larger lymphatics and nodes. However, it can 
be technically difficult, is unpleasant for the 
patient, may cause further lymphatic injury, 
and has largely become obsolete as a routine 
method of investigation. Indirect lymphangiog- 
raphy involves the intradermal injection of 
water-soluble nonionic contrast into a web 
space, from where it is taken up by lymphatics 
and then followed radiographically It shows 
distal lymphatic but not normally proximal 
lymphatics and nodes. Isotope lympho- 
scintigraphy has largely replaced lymphangiog- 
raphy as the primary diagnostic technique 
in cases of clinical uncertainty. Radioactive 
technetium-labeled protein or colloid particles 
are injected into an interdigital web space and 
specifically taken up by lymphatics, and serial 
radiographs are taken with a gamma camera. 
The technique provides a qualitative measure of 
lymphatic function rather than quantitative 
function or anatomical detail. A single axial 
computed tomography (CT) slice through the 
mid-calf has been proposed as a useful diag- 
nostic test for lymphedema (coarse, nonen- 
hancing, reticular honeycomb pattern in an 
enlarged subcutaneous compartment), venous 
edema (increase volume of the muscular com- 
partment), and lipedema (increased subcuta- 
neous fat). Computed tomography can also be 
used to exclude pelvic or abdominal mass 
lesions. Magnetic resonance imaging (MRI) can 
provide clear images of lymphatic channels and 
lymph nodes, and can be useful in the assess- 
ment of patients with lymphatic hyperplasia. It 
can also distinguish venous and lymphatic 
causes of a swollen limb. In cases where 
malignancy is suspected, samples of lymph 
nodes may be obtained by fine-needle aspira- 
tion, needle core biopsy, or surgical excision. 
Skin biopsy will confirm the diagnosis of 
lymphangiosarcoma. 




> 



120 



VASCULAR SURGERY 



Management 

Ideally, a multiprofessional team comprising 
physical therapists, nurses, orthotists, physi- 
cians (dermatologists, oncologists, palliative 
care specialists), surgeons, and social services 
should deliver the care. Although surgery itself 
has a very small role, surgeons (especially breast 
and vascular) are frequently asked to oversee 
the management of these patients. Early 
diagnosis and institution of management are 
essential because at that stage relatively simple 
measures can be highly effective and will 
prevent the development of disabling late-stage 
disease, which is extremely difficult to treat. 
There is often a latent period of several years 
between the precipitating event and the onset of 
lymphedema. The identification, education, and 
treatment of such at-risk patients can slow 
down, even prevent, the onset of disease. In 
patients with established lymphedema, the 
three goals of treatment are to relieve of pain, 
reduce swelling, and prevent the development of 
complications. 

On initial presentation 50% of patients with 
lymphedema complain of significant pain. The 
pain is usually multifactorial, and its severity 
and underlying cause(s) vary depending on the 
etiology of the lymphedema. For example, 
following treatment for breast cancer, pain 
may arise from the swelling itself, radiation and 
surgery induced, nerve (brachial plexus and 
intercostobrachial nerve), bone (secondary 
deposits, radiation necrosis) and joint disease 
(arthritis, bursitis, capsulitis), and recurrent 
disease. The detailed treatment of such patients 
is beyond the scope of this chapter but involves 
the considered use of nonopioid and opioid 
analgesics, corticosteroids, tricyclic antidepres- 
sants, muscle relaxants, antiepileptics, nerve 
blocks, physiotherapy, adjuvant anticancer ther- 
apies (chemo-, radio-, hormonal therapy), as 
well as measures to reduce swelling if possible. 
In patients with non-cancer-related lym- 
phedema, the best way to reduce pain is to 
control swelling and prevent the development of 
complications. 

Physical therapy for lymphedema comprising 
bed rest, elevation, bandaging, compression 
garments, massage, and exercises was first 
described at the end of the 19th century, and 
through the 20th century various eponymous 
schools developed. Although there is little doubt 



that physical therapy can be highly effective 
in reducing swelling, its general acceptance 
and practice has been hampered by a lack of 
proper research and confusing terminology. 
The current preferred term is decongestive lym- 
phedema therapy (DLT) and comprises two 
phases. The first is a short intensive period of 
therapist-led care, and the second is a mainte- 
nance phase where the patient uses a self-care 
regimen with occasional professional interven- 
tion. The intensive phase comprises skin care, 
manual lymphatic drainage (MLD), multilayer 
lymphedema bandaging (MLLB), and exercises. 
The length of intensive treatment depends on 
the disease severity, the degree of patient com- 
pliance, and the willingness and ability of 
the patient to take more responsibility for the 
maintenance phase. However, weeks rather 
than months should be the goal. 

The patient must be carefully educated in the 
principles and practice of skin care. The patient 
should inspect the affected skin daily, with 
special attention paid to skin folds where mac- 
eration may occur. The limb should be washed 
daily, the use of bath oil (e.g., balneum) is rec- 
ommended as a moisturizer, and the limb must 
be carefully dried afterward. A hair drier, on low 
heat, is more effective and hygienic, and less 
traumatic, than a towel. If the skin is in good 
condition, daily application of a bland emollient 
is recommended to keep the skin hydrated. If 
the skin is dry and flaky, then a bland ointment 
[e.g., 50/50 white soft paraffin/liquid paraffin 
(WSP/LP)] should be used twice daily, and if 
there is marked hyperkeratosis, then a kera- 
tolytic agent such as 5% salicylic acid can be 
added. Many commercially available soaps, 
creams, and lotions contain sensitizers, and, as 
patients with lymphedema are highly suscepti- 
ble to contact dermatitis (eczema), are best 
avoided. Apart from causing intense discomfort, 
eczema acts as an entry point for infection. 
Management comprises avoidance of the aller- 
gen (patch testing maybe required) and topical 
corticosteroids. Fungal infections are common, 
difficult to eradicate, and predispose to AIEs. 
Chronic application of antifungal creams leads 
to maceration, and it is better to use powders 
in shoes and socks. Ointment containing 3% 
benzoic acid helps prevent athlete's foot and can 
be used safely over long periods. Painting at-risk 
areas with an antiseptic agent such as eosin may 
be helpful. Lymphorrhea is uncommon but 



121 



CHRONIC VENOUS INSUFFICIENCY, VARICOSE VEINS, LYMPHEDEMA, AND ARTERIOVENOUS FISTULAS 




extremely troublesome. Management comprises 
emollients, elevation, compression, and some- 
times cautery under anesthesia. 

Apart from lymphangiosarcoma, AIEs are 
probably the most serious complications of 
lymphedema and frequently lead to emergency 
hospital admission. About 25% of primary and 
5% of secondary lymphedema patients are 
affected. The AIEs start rapidly, often without 
warning or precipitating event, with tingling, 
pain, and redness of the limb. Patients feel viral, 
and severe attacks can lead to the rapid onset of 
fever, rigors, headache, vomiting, and delirium. 
Patients who have suffered previous attacks can 
usually predict the onset, and many learn to 
carry antibiotics with them and self-medicate at 
the first hint of trouble. This may stave off a 
full-blown attack and prevent the further lym- 
phatic injury that each AIE causes. It is rarely 
possible to isolate a responsible bacterium, but 
the majority are presumed to be streptococcal 
or staphylococcal in origin. The diagnosis is 
usually obvious but dermatitis, throm- 
bophlebitis, and DVT are in the differential. 
Benzyl (intravenous) or phenoxymethyl (oral) 
penicillin, and flucloxacillin (or clindamycin in 
severe attacks), are the antibiotics of choice and 
should be given for 2 weeks. Rest reduces lym- 
phatic drainage and the spread of infection, 
elevation reduces the edema, and heparin pro- 
phylaxis reduces the risk of DVT. Co-amoxiclav 
can be taken by patients who self-medicate. The 
use of long-term prophylactic antibiotics is not 
evidence-based but is probably reasonable in 
patients who suffer frequent attacks. However, 
the benefits of scrupulous compliance with 
physical therapy and skin care cannot be under- 
estimated. 

Several different techniques of MLD have 
been described and the details are beyond the 
scope of this chapter. However, they all aim to 
evacuate fluid and protein from the ISF space 
and stimulate lymphangion contraction. Thera- 
pists should perform MLD daily; they should 
also train the patient or caregiver to perform a 
simpler, modified form of massage termed 
simple lymphatic drainage (SLD). In the inten- 
sive phase, SLD supplements MLD, and once the 
maintenance phase is entered, SLD will carry on 
as daily massage. 

Elastic bandages provide compression, pro- 
duce a sustained high resting pressure, and 
compress more as limb swelling reduces. 



However, the sub-bandage pressure does not 
alter greatly in response to changes in limb cir- 
cumference consequent upon muscular activity 
and posture. By contrast short-stretch bandages 
exert support through the production of a semi- 
rigid casing, where the resting pressure is low 
but changes quite markedly in response to 
movement and posture. 

It is generally believed that nonelastic MLLB 
is preferable (and arguably safer) in patients 
with severe swelling during the intensive phase 
of DLT, whereas compression (hosiery, sleeves) 
is preferable in milder cases and during the 
maintenance phase. Whether the aim is to 
provide support or compression, the pressure 
exerted must be graduated (100% ankle/foot, 
70% knee, 50% mid-thigh, 40% groin), and the 
adequacy of the arterial circulation must be 
assessed. As it is rarely possible to feel pulses in 
the lymphodematous limb, noninvasive assess- 
ment of ABI using a handheld Doppler ultra- 
sound device is usually necessary. The details of 
MLLB are beyond the scope of this chapter; 
however, it is highly skilled and in order to be 
effective and safe, it needs to be applied by a 
specially trained therapist. It is also extremely 
labor intensive, needing to be changed daily. 
Compression garments form the mainstay of 
management in most clinics. The control of 
lymphedema requires higher pressures (30 to 40 
mmHg in the arm, 40 to 60mmHg in the leg) 
than are typically used to treat CVI. The patient 
should put the stocking on first thing in 
the morning before rising. It can be difficult 
to persuade patients to comply. Donning 
lymphedema-grade stockings is difficult, and 
many patients find them intolerably uncomfort- 
able, especially in warm climates. Furthermore, 
although intellectually they understand the 
benefits, emotionally they may find wearing 
them presents a greater body image problem 
than the swelling itself. 

Enthusiasm for pneumatic compression 
devices has waxed and waned. Unless the device 
being used allows the sequential inflation of 
multiple chambers up to 50 mm Hg, it will prob- 
ably be ineffective for lymphedema. Patient 
benefit is maximized and complications mini- 
mized if these devices are used under the direc- 
tion of a physical therapist as part of an overall 
package of care. 

Lymph formation is directly proportional to 
arterial inflow, and 40% of lymph is formed 




> 



122 



VASCULAR SURGERY 



within skeletal muscle. Vigorous exercise, espe- 
cially if it is anaerobic and isometric, tends to 
exacerbate lymphedema, and patients should 
be advised to avoid prolonged static activities, 
for example, carrying heavy shopping bags or 
prolonged standing. By contrast, slow rhythmic 
isotonic movements (e.g., swimming) and mas- 
sage increase venous and lymphatic return 
through the production of movement between 
skin and underlying tissues (essential to the 
filling of initial lymphatics) and augmentation 
of the muscle pumps. Exercise also helps to 
maintain joint mobility. Patients who are unable 
to move their limbs benefit from passive exer- 
cises. When at rest, the lymphedematous limb 
should be positioned with the foot/hand above 
the level of the heart. A pillow under the mat- 
tress or blocks under the bottom of the bed 
encourage the swelling to go down overnight. 

There are considerable, and scientifically 
inexplicable, differences in the use of specific 
drugs for venous disease and lymphedema 
between different countries. The benzpyrenes 
are a group of several thousand naturally occur- 
ring substances, of which the flavonoids have 
received the most attention. Enthusiasts argue 
that a number of clinical trials have shown 
benefit from these compounds, which are 
purported to reduce capillary permeability, 
improve microcirculatory perfusion, stimulate 
interstitial macrophage proteolysis, reduce ery- 
throcyte and platelet aggregation, scavenge free 
radicals, and a exert an antiinflammatory effect. 
Detractors argue that the trials are small and 
poorly controlled, with short follow-up and soft 
end points, and that any benefits observed can 
be explained by a placebo effect. Diuretics are of 
no value in pure lymphedema. Their chronic use 
is associated with side effects including elec- 
trolyte disturbance and should be avoided. 

Surgery 

Surgery benefits only a small minority of 
patients with lymphedema. Operations fall into 
two categories: bypass procedures and reduc- 
tion procedures. The rare patient with proximal 
ilioinguinal lymphatic obstruction and normal 
distal lymphatic channels might benefit, at least 
in theory, from lymphatic bypass. A number of 
methods have been described including the 
omental pedicle, the skin bridge (Gillies), anas- 
tomosing lymph nodes to veins (Neibulowitz), 



the ileal mucosal patch (Kinmonth), and, more 
recently, direct lymphovenous anastomosis. 
The procedures are technically demanding, not 
without morbidity, and there is no controlled 
evidence to suggest that these procedures 
produce an outcome superior to the best 
medical management alone. Limb reduction 
procedures are indicated when a limb is so 
swollen that it interferes with mobility and 
livelihood. These operations are not cosmetic in 
the sense that they do not create a normally 
shaped leg and are usually associated with 
significant scarring. Four operations have been 
described: 

1. Sistrunk: A wedge of skin and subcuta- 
neous tissue is excised and the wound 
closed primarily. This is most commonly 
employed to reduce the girth of the thigh. 

2. Homan: Skin flaps are elevated; subcuta- 
neous tissue is excised from beneath the 
flaps, which are then trimmed to size to 
accommodate the reduced girth of the 
limb and closed primarily. This is the most 
satisfactory operation for the calf. The 
main complication is skin flap necrosis. 
There must be at least 6 months between 
operations on the medial and lateral sides 
of the limb, and the flaps must not pass 
the midline. This procedure has also been 
used on the upper limb but is contraindi- 
cated in the presence of venous obstruc- 
tion or active malignancy. 

3. Thompson: One denuded skin flap is 
sutured to the deep fascia and buried 
beneath the second skin flap (the so-called 
buried dermal flap). This procedure has 
become less popular as pilonidal sinus for- 
mation is common, the cosmetic result is 
no better than that obtained with the 
Homan's procedure, and there is no evi- 
dence that the buried flap establishes any 
new lymphatic connections within the 
deep tissues. 

4. Charles: This operation was initially 
designed for filariasis and involved exci- 
sion of all the skin and subcutaneous 
tissues down to the deep fascia with cover- 
age using split skin grafts. This leaves a 
very unsatisfactory cosmetic result, and 
graft failure is not uncommon. However, it 
does enable the surgeon to reduce greatly 
the girth of a massively swollen limb. 



123 



CHRONIC VENOUS INSUFFICIENCY, VARICOSE VEINS, LYMPHEDEMA, AND ARTERIOVENOUS FISTULAS 




Arteriovenous Fistulas 

Classification 

Arteriovenous fistulas (AVFS) are broadly 
classified into congenital and acquired. 
Acquired fistulas may be traumatic, iatrogenic, 
or associated with malignancy, aneurysmal 
disease, and infection. These are not considered 
further. Congenital AVFs are further classified 
as hemangiomas or malformations. The former 
are characterized by endothelial hyperplasia, 
are not present at birth, and grow in early child- 
hood, but in 90% of cases involute by the age 
of 5 to 10 years. The latter exhibit normal 
endothelial cell kinetics, are always preset at 
birth, grow and continue to grow with the child, 
and may enlarge dramatically at puberty or in 
pregnancy. Whenever vascular lesions appear 
and grow rapidly, it is important to exclude 
malignancy. Malformations may be high flow 
(predominantly cardiac and great vessel anom- 
alies) or low flow (arterial, venous, lymphatic, or 
mixed). The processes leading to the develop- 
ment of the mature vascular tree are largely 
unknown, but presumably congenital fistulas 
represent a localized disorder of vessel forma- 
tion. Occasionally they may be familial, and 
some abnormalities have been mapped to 
certain chromosomal loci. 



Symptoms and Signs 

The clinical presentations are protean and 
depend on the nature of the lesion, anatomical 
site, size, and flow. 



Malformations 

Although malformations are present at birth, 
they do not usually become symptomatic, and 
even go unnoticed, until later at life. They typi- 
cally present at puberty or during pregnancy or 
following trauma to the part. There is usually 
an obvious swelling and discoloration, and 
there maybe limb enlargement. Adults typically 
present with a lump, VVs, (ischemic) ulceration, 
bleeding, or an inequality in limb length. That 
which is visible is often the "tip of the iceberg," 
and the deep component may cause pain, pres- 
sure on local strictures, organ dysfunction, and 
internal bleeding. On examination there is dis- 



coloration and a lump. If arterial, it is typically 
firm, pulsates, and is associated with a thrill and 
a murmur and sometimes pulsatile draining 
veins. If primarily venous, it is soft and com- 
pressible, and reduces in size and enlarges upon 
elevation and dependency, respectively. 

Hemangiomas 

Hemangiomas present at or within a few weeks 
of birth. They are said to be present in 10% 
of Caucasian children on their first birthday, to 
be three times commoner in girls, to be multi- 
ple in 20%, and to affect the head and neck 
(60%), trunk (25%), and extremities (10%). If 
superficial, they are bright (strawberry nevus), 
and if deep dark (cavernous hemangioma), red. 
They are firm and rubbery and cannot be 
emptied of blood on compression or elevation. 
After an initial phase of rapid growth, they 
begin to involute at about 6 to 12 months of 
age when the red color turns to purple, 
gray/while flecks appear, the lesion becomes 
softer, and the overlying skin wrinkles. Resolu- 
tion is typically completed in 50% at 5 years, 
70% at 7 years, and 90% or more by 10 years. 
Apart from cosmetic concerns there may be 
ulceration and bleeding. On the face they may 
block vision, and depending on where they are 
sited may cause mass effects. Large heman- 
giomas involving internal organs may cause 
heart failure and anemia. 

Diagnosis and Investigation 

The diagnosis can usually be made on clinical 
examination. Handheld Doppler helps to con- 
firm if there is an arterial component, and DU 
provides more detailed anatomical and func- 
tional information. In particular, DU permits 
detailed assessment of the venous system in 
patients with Klippel-Trenaunay syndrome 
where the deep venous system maybe hypoplas- 
tic or even absent, having been replaced by an 
abnormal laterally placed venous malformation. 
Duplex ultrasound has largely replaced venog- 
raphy. Phleboliths may be seen on plain x-rays 
and are only usually seen in venous lesions. 
Magnetic resonance imaging, rather than CT, is 
now regarded as the definitive investigation for 
assessing the extent of AVF. Angiography is 
performed only where there is an intention to 
treat radiologically 




> 



124 



VASCULAR SURGERY 



Management 

Management is complex, multidisciplinary (vas- 
cular, plastic, maxillofacial, and orthopedic sur- 
geons, interventional radiologist, cardiologist), 
difficult, and highly tailored to the individual 
patient. Many patients (and parents) simply 
require reassurance, and in general it pays to be 
as conservative as possible. Only 10% of heman- 
giomas fail to resolve spontaneously and many 
malformations remain asymptomatic. Pallia- 
tion, not cure, is the principal aim, and it is 
important to ensure that the treatment is not 
worse than the disease. Venous lesions may 
be treated with compression bandaging and 
hosiery. A small minority of patients are suit- 
able for excisional surgery. Complete excision is 
rarely possible, and usually the aim is to remove 
the most symptomatic part. Such surgery can be 
difficult and bloody, and recurrence is common. 
For these reasons, most patients are treated 
radiologically either by transcatheter emboliza- 



tion (arterial lesions) or by direct injection 
(venous lesions), usually under general anesthe- 
sia. This is a highly skilled and specialized 
branch of interventional radiology, and great 
care must be taken to avoid collateral damage. 
Extremities are particularly vulnerable. Surgical 
skeletonization of the arterial inflow to the 
lesion is now avoided, as recurrence is inevitable 
and such intervention prevents radiological 
intervention. Strong indications for interven- 
tion are hemorrhage, distal ischemia due to 
steal, and ulceration due to ischemia or venous 
hypertension. 

References 

Cornu-Thenard A, Boivin P, Baud JM, De Vincenzi I, 
Carpentier PH. (1994) J Dermatol Surg Oncol 
20:318-26. 

Dwerryhouse S, Davies B, Harradine K, Earnshaw JJ. (1999) 
J Vase Surg 29:589-92. 

Szuba A, Rockson SG. (1997) Vase Med 2:321-6. 



11 



Vascular Trauma 

Kathleen J. Ozsvath, R. Clement Darling III, 
Laila Tabatabai, Sacha Hamdani, Alun H. Davies, 
and Meryl Davis 




Trauma is the leading cause of death in patients 
under the age of 44 in the United States. North 
American trauma centers have an incidence of 
penetrating trauma of 35% as compared with 
5% to 8% in Europe, the difference being ex- 
plained by the higher rate of weapon-related 
crimes in North America. The treatment of trau- 
matic vascular injuries today is based on prin- 
ciples gained during the military conflicts of the 
20th century; previously treatment of vascular 
injuries was limited to the staunching of bleed- 
ing by cautery, compression, and ligature. The 
concept of repair was documented in anecdotal 
reports only. William Hunter in 1759 recounts 
the first successful vascular repair on a brachial 
artery using a farrier's stitch. 

Reports of vein grafting were available in 
the early 20th century; however, these tech- 
niques were not suitable for the injuries en- 
countered during World War I, in which the 
amputation rate was noted to be 72.5%. In 1946 
DeBakey and Simeone published a review of 
World War II experiences with vascular surgery. 
They concluded that ligation was "of stern 
necessity," required to control hemorrhage; 
attempts at vascular repair were superior to 
ligation and led to an amputation rate of 
49%. 

The application of vessel reconstruction in 
the Korean War, despite the mean lag time of 
over 6 hours between injury and repair, reduced 
the lower limb amputation rate to 13% (Hughes, 
1959); a comparable figure was achieved during 
the Vietnam War. The Vietnam Vascular Regis- 



try was established during the Vietnam War; 
surgeons were able to document and analyze the 
long-term management and outcome of vascu- 
lar trauma (Rich and Hughes, 1969). The 
significantly improved long-term results of vas- 
cular repair were attributed to faster evacuation 
of casualties within 3 hours of injury, thereby 
allowing surgeons to treat injuries that had pre- 
viously been otherwise fatal. Operations were 
performed by experienced surgeons using auto- 
logous vein grafts. 

The rise of civilian trauma in the United 
States has resulted in surgeons becoming more 
adept and experienced at dealing with vascu- 
lar injuries (Mattox et al., 1989). Penetrating 
injuries have been the number one cause of 
vascular trauma in both urban and rural com- 
munities, whereas blunt trauma has accounted 
for approximately 50% of vascular injuries, 
most commonly secondary to road traffic 
accidents. 

Regardless of the etiology of the vessel injury, 
the essential principles of treatment are emer- 
gency resuscitation at the scene, triage and 
rapid transport to an appropriate hospital, vig- 
orous resuscitation, diagnosis, and definitive 
surgery. Ideally, vascular injuries should be 
treated by surgeons with expertise in vascular 
reconstruction and trauma, in an environment 
with the necessary ancillary support services to 
allow the best results. 

This chapter reviews the workup and treat- 
ment of traumatic vascular injuries involving 
the head and neck, chest, abdomen, and extrem- 



125 




> 



126 



VASCULAR SURGERY 



ities, and discusses the approach to iatrogenic 
injuries and the endovascular treatment of vas- 
cular injuries. 



Mechanisms of Injury 

Vascular injuries can be divided into penetrat- 
ing and blunt. Penetrating injuries include stab- 
bing and gunshot wounds. Stab wounds are 
usually clean with minimal soft tissue injury; 
however, in the neck and upper limb concurrent 
nerve damage must also be suspected. 

Gunshot wounds are classified according to 
the bullet velocity. Low velocity bullets have 
a velocity of less than 250 m/s, whereas 750 to 
900 m/s represents the speed of high-velocity 
bullets. Low-velocity bullets injure the tissue 
through which they pass, whereas high-velocity 
bullets create a cavitational effect, thereby caus- 
ing a suctional effect contaminating the entire 
wound. If a high-velocity bullet strikes bone, 
there is extensive comminution with a large exit 
wound. Shotgun injuries particularly to a limb 
can cause vascular damage at several levels. 
Bombs, mines, and rocket-propelled grenades 
produce complex injuries that frequently result 
in amputation. 

Blunt vascular trauma is often caused by 
deceleration in road traffic accidents and is fre- 
quently associated with other injuries. Femoral 
shaft fractures and fracture dislocations of the 
knee carry a 10% to 40% incidence of vascular 
injury. It is important to realize that although 
the artery can remain intact, intimal damage 
with concomitant thrombosis risk can occur. 
Blunt trauma to the upper limb is often asso- 
ciated with avulsion injuries to the brachial 
plexus. Extensive crush injuries to the limbs are 
associated with a poor prognosis due to soft 
tissue damage and reperfusion injury. 

Iatrogenic injuries are increasing in inci- 
dence as a consequence of invasive procedures. 
Cardiological and radiological catheterization 
cause between 60% and 76% of all iatrogenic 
injuries. Orthopedic procedures including joint 
replacements may cause vascular injury, com- 
monly to the external iliac, common femoral, or 
popliteal arteries. In such cases where iatrogenic 
arterial injury is suspected, expeditious referral 
to a vascular surgeon must be made. 



Diagnosis 



The clinical manifestations of vascular injury 
are divided into hard and soft signs (Table 11.1). 
The management of penetrating limb trauma in 
the presence of one hard sign is exploration in 
the operating room. In blunt trauma, closed 
head injuries, spinal damage, and cervical and 
brachial plexus injuries may complicate the 
clinical findings; a careful neurological exami- 
nation is mandatory. 

Clinical examination in blunt and complex 
penetrating trauma may be unreliable, and 
trauma centers advise early angiography. The 
role of duplex ultrasound in vascular trauma is 
less clear; currently it has a major role in the 
diagnosis of occult vascular injuries and in 
postoperative assessment. Doppler arterial pres- 
sure index (API) is a useful tool. It is the systolic 
arterial pressure in the injured extremity 
divided by the arterial pressure in a noninjured 
arm. A result of <0.90 has been found to have a 
sensitivity of 95% and specificity of 97% for 
major arterial injury (Johansen et al., 1991). 

Once the diagnosis of major vascular injury 
has been made, the majority of patients require 
exploration and repair. The principles of emer- 
gency vascular repair are to control bleeding 
and prevent limb ischemia. Hemorrhage is 
usually apparent, but ischemia maybe insidious 
and must be sought. Time is paramount; it is 
generally accepted that more than 6 to 8 hours 
of warm ischemia time makes limb survival 
unlikely. If there is significant concurrent nerve 
damage, then limb reconstruction may not be 
appropriate and amputation should be per- 
formed. The input of multidisciplinary teams is 
vital to ensure optimal treatment. 



Table 11.1. The clinical manifestations of vascular injury are 
divided into hard and soft signs 



Hard signs 


Soft signs 


No pulse 


Hematoma (small) 


Thrill or bruit 


History of bleeding at scene 


Active bleeding 


Hypotension 


Hematoma (large/ 


Nerve damage 


expanding) 




Distal ischemia 





127 



VASCULAR TRAUMA 




Vascular Injuries in the Neck 

Wounds of major vessels are often lethal, and 
frequently these patients have multiple injuries. 
This is particularly true in penetrating in- 
juries of the brachiocephalic vessels, where air- 
way compromise, hemorrhage, and neurological 
damage secondary to impaired brain blood flow 
may occur, along with associated injuries to the 
pharynx, esophagus, and brachial plexus. 

In the neck penetrating trauma is more 
common than blunt trauma, although vascular 
injuries caused by blunt trauma can be more 
difficult to diagnose and treat. Blunt injuries 
include steering wheel injuries, deceleration 
forces, and crushing blows. 

Injuries to the carotid artery occur in 6% of 
penetrating injuries to the neck and make up 
22% of all vascular neck injuries, whereas 3% to 
5% of carotid injuries are secondary to blunt 
trauma (Demetriades et al., 1996). The mortal- 
ity rate of patients with penetrating neck vas- 
cular trauma is 5% to 20%. Patients presenting 
with obvious signs of penetrating vascular 
injury (expanding hematoma, pulsatile bleed- 
ing) should be rapidly explored operatively 

Treatment of vascular injuries in the neck is 
based on dividing the neck into three zones: 
zone I is 1cm above the manubrium including 
the thoracic outlet, zone II extends from the 
upper limit of zone I to the angle of the man- 
dible, and zone III lies between the angle of the 
mandible and the base of the skull (Fig. 11.1). 

Zone I injuries are best explored through a 
median sternotomy, which can then be extended 
superiorly along the anterior border of the ster- 
nocleidomastoid muscle as needed. Penetrating 
trauma to vessels in zone II can be approached 
directly for definitive repair. Zone III injuries 
can be difficult to access and sometimes require 
subluxation of the mandible or mandibular 
osteotomy. Patients with zone III asymptomatic, 
angiographically documented injuries are now 
being managed with endovascular stenting or 
embolization (Feliciano, 2001). 

Arterial defects are repaired with primary 
repair, transposition, patch, or bypass. Patients 
found to have an acutely occluded carotid artery 
are anticoagulated in an effort to prevent clot 
propagation. Neurologically impaired patients 
with carotid injuries are aggressively treated 
surgically. Patients with small intimal defects 




Figure 11.1. Treatment of vascular injuries in the neck is based 
on dividing the neck into three zones: zone I is 1 cm above the 
manubrium to include the thoracic outlet, zone II extends from 
the upper limit of zone I to the angle of the mandible, and zone 
III lies between the angle of the mandible and the base of the 
skull. 



and dissections of the carotid artery who are 
otherwise stable may be followed with physical 
examination and follow-up duplex ultrasound, 
because these lesions may resolve without inter- 
vention. Venous injuries may be dealt with by 
ligation. In the unusual circumstance of both 
internal jugular veins having been damaged it is 
imperative that one vein be reconstructed. 
Patients with blunt carotid artery injuries can 
be difficult to diagnose due to either other 
distracting injuries or altered mental status. 
Carotid artery blunt injuries are associated with 
a high mortality and poor neurological out- 
comes in most patients. 

Mechanisms of blunt carotid artery in- 
juries include hyperextension, direct injury, 
basilar skull fractures, and intraoral injuries. 
Patients may present with carotid artery dissec- 
tions, thrombosis, pseudoaneurysms, carotid- 
cavernous sinus fistulas, or arterial disruption. 
Carotid artery dissections and thrombosis are 
treated with anticoagulation. Cogbill et al. 
(1994) reported that two thirds of patients with 
carotid dissections eventually have normal 
studies on follow-up, whereas one third pro- 
gress. Pseudoaneurysms may be treated surgi- 




> 



128 



VASCULAR SURGERY 



cally or radiologically depending on their size. 
Carotid-cavernous sinus fistulas are generally 
treated by endovascular techniques. Complete 
disruption of the carotid artery is associated 
with a high mortality. 

Injury to the vertebral arteries is rare. The 
anatomical location of the injury determines the 
best operative approach when intervention is 
required. Proximal vertebral artery exposure is 
performed by a transverse supraclavicular inci- 
sion or an anterior cervical incision. The cervi- 
cal vertebral artery is best approached through 
an incision along the anterior border of the ster- 
nocleidomastoid muscle, and can be extended 
to the posterior auricular area. Once the verte- 
bral artery takes its course from the C2 foramen 
and enters the skull, the exposure becomes very 
difficult. Ligation or occlusion of the vertebral 
artery is usually the treatment of choice. Per- 
cutaneous techniques have been successfully 
employed in managing arteriovenous fistulas, 
pseudoaneurysms, and occlusions (Halbach 
et al, 1993). 

Thoracic Vascular Injuries 

Thoracic vascular trauma constitutes approxi- 
mately 10% of vascular trauma (Bongard et al., 
1990). Although penetrating trauma remains 
the most common etiology of thoracic vascular 
injuries, deceleration injuries and crush injuries 
can result in major thoracic vascular trauma. 
Injuries to the vessels in the thoracic cavity 
can lead to rapid blood loss and hemodynamic 
collapse; many patients die before reaching 
the hospital. It is often more appropriate to 
bypass the nearest hospital and transfer the 
patient to a unit able to manage cardiothoracic 
problems. 

These patients may require immediate treat- 
ment for tension pneumothorax or cardiac tam- 
ponade. Thoracotomy may be required as an 
emergency procedure and is usually carried out 
via a left anterolateral thoracotomy incision 
(which can be extended across the sternum) or 
a posterolateral thoracotomy. Access to carotid 
arteries and innominate artery can be obtained 
through a median sternotomy. Middle and distal 
subclavian artery injuries can be controlled 
with infra- and supraclavicular approaches. 

Clinical findings suggesting thoracic injury 
include external evidence of severe chest 



bruising, reduced or absent lower limb pulses, 
raised jugular venous pressure, and unex- 
plained hypotension. The chest radiograph may 
show a widened mediastinum, fracture of the 
first or second rib, hemothorax, and thoracic 
spine injury. 

In stable patients the investigations of choice 
are chest radiographs (suggesting great vessel 
or aortic injury), spiral computed tomography 
(CT) with contrast, angiography, and trans- 
esophageal echocardiography. 

The most common intrathoracic injury 
caused by deceleration involves disruption of 
the descending thoracic aorta at the isthmus. 
Sudden death is common, and it is estimated 
that 90% die before reaching the hospital; of 
those who reach the hospital, 25% die within 
24 hours. 

Commonly the site of aortic disruption is 
distal to the left subclavian artery. Access is 
gained via a left posterior thoracotomy with 
cardiopulmonary bypass. The incidence of 
paraplegia is approximately 8%. Recent endolu- 
minal treatment has been described and shown 
to be successful with a lower mortality rate. 

The subclavian arteries are relatively well 
protected from blunt trauma. Patients may 
present with absent distal pulses, and injury 
should be suspected in patients with a first rib 
fracture or traction injury to the brachial 
plexus. In stable patients, careful clinical assess- 
ment of the brachial plexus and magnetic reso- 
nance imaging (MRI) should be performed 
prior to surgical exploration. 

Direct repair of the vessel, patch, and pros- 
thetic or autologous interposition grafts are 
possible choices in repairing aortic and great 
vessel injuries. Some centers are now opting to 
use endovascular techniques (Ohki et al., 1997). 
At present this method of treatment should be 
pursued only in institutions with adequate sur- 
gical and radiological expertise. 

Abdominal Vascular Injuries 

One third of patients with vascular trauma pre- 
sent with abdominal vascular injuries. These 
patients are more commonly victims of pene- 
trating injuries, with mortality rates averaging 
50%. Deceleration and compression injuries are 
common blunt injuries and may cause damage 
to the renal or superior mesenteric arteries and 



129 



VASCULAR TRAUMA 




portal vein tributaries. Vessels can be injured 
by transection or partial transection, or have 
intimal defects causing thrombosis. Major vas- 
cular injury in the abdomen is often associated 
with injuries to other intraabdominal organs. Of 
those patients who reach the hospital, the mor- 
tality postsurgery remains high (50% to 70% 
for aortic injury and 30% to 53% for vena caval 
injury). 

Resuscitation is based on Advanced Trauma 
Life Support (ATLS) guidelines, with unstable 
patients being transferred promptly to the oper- 
ating room (Fig. 11.2). Intrathoracic injuries are 
associated in up to 25% of patients with gunshot 
wounds of the abdomen. 

The assessment of stable patients with intra- 
abdominal pathology has been extensively dis- 
cussed; ultrasound and CT are the modalities of 
choice. Angiography in the stable patient with 
blunt injuries may be useful in documenting 
unusual injuries. 

In unstable patients with significant hemor- 
rhage, laparotomy should be performed and the 
abdomen packed, and systematic evaluation 
of the abdomen undertaken. Large defects in 
the gastrointestinal tract should be temporarily 
controlled with soft bowel clamps to reduce 
contamination. If hemorrhage is not controlled, 
it maybe necessary to cross-clamp the proximal 
supraceliac aorta via the lesser sac; alternatively 
(via the left thorax) the descending thoracic 
aorta may be cross-clamped. The goal is to 
obtain proximal and distal control of the hem- 
orrhaging vessels so that repair or ligation can 
quickly be undertaken. Hypothermia and coag- 
ulopathy is a serious risk; therefore, expeditious 
control is important. Aortic injuries can be 
repaired with a transverse primary repair, a 



patch with autologous or prosthetic material, or 
interposition grafting with prosthetic material. 
The use of antibiotic-soaked grafts may over- 
come concerns regarding the use of prosthetic 
material in the presence of penetrating injuries. 

The decision to explore a retroperitoneal 
hematoma depends on the mechanism of injury 
and stability of the patient. For a retroperitoneal 
hematoma caused by blunt trauma, a conser- 
vative approach is advised especially when it 
is associated with pelvic fractures. This can 
be addressed by external fixation of the pelvis, 
angiography, and coil embolization. In contrast 
an expanding or pulsatile retroperitoneal 
hematoma requires immediate exploration. 

Retrohepatic caval injuries are difficult to 
control and carry a high mortality. Mobilization 
of the liver with division of the right triangular 
ligament and right thoracotomy with dissection 
of the diaphragm may be required. Bleeding 
from the porta hepatis can be controlled by 
compression of the hepatic artery and portal 
vein (Pringle's maneuver). 

The mortality rate for patients with 
significant superior mesenteric artery (SMA) 
injury can be high (58% mortality rate). Injuries 
to the proximal SMA are technically challenging 
due to the proximity of the pancreas and the 
presence of multiple short branches. Control of 
the suprameso colic vessels can be obtained by 
medial visceral rotation via the left paracolic 
gutter. Injuries to the celiac artery and branches 
may present with an expanding hematoma dis- 
placing the stomach and pancreas forward. 
Celiac axis vessels may be ligated if there is 
a patent SMA. Retropancreatic SMA control 
may have to be obtained by transection of the 
pancreas. 



Haemodynamically 
Unstable 



Figure 11.2. Algorithm for the 
management of a patient with 
abdominal trauma. 



Theatre 



Resuscitation as per 
ATLS guidelines 




Haemodynamically 
Stable 



Ultrasound 




Angiography 




> 



130 



VASCULAR SURGERY 



Injury or transection of the SMA may require 
either autologous or prosthetic bypass. Bowel 
viability may be difficult to assess intraopera- 
tively; therefore, second-look laparotomy in 
these patients is advisable. 

Inframesocolic vessel injuries can be ap- 
proached by mobilizing the small intestines and 
transverse colon superiorly. The inferior mesen- 
teric artery can be ligated in most patients, as 
collaterals exist that provide adequate blood to 
the intestine. 

Access to the inferior vena cava, right renal 
vein, suprarenal aorta, and porta may be gained 
by an extended mobilization of the duodenum, 
head of pancreas, and right colon (Kocher 
maneuver). The infrarenal aorta is exposed by 
retracting the small bowel to the right and incis- 
ing the retroperitoneum from the root of the 
mesocolon to the pelvis. Simple stab wounds to 
the aorta may be closed primarily; more exten- 
sive injuries may require a prosthetic patch or 
bypass graft. In the presence of significant con- 
tamination, an axillobifemoral bypass may be 
preferred after ligation of the aorta. Iliac vessel 
injuries can be approached via the retroperi- 
toneum; such injuries carry a high mortality 
(10% to 40%) and morbidity including limb 
loss. 

Renal artery injury may occur after rapid 
deceleration, resulting in acute renal artery 
thrombosis or laceration. The diagnosis of 
thrombosis is often made late, and if explored 
12 hours after the injury, renal salvage is often 
limited. In patients with a functioning kidney 
on the opposite side, a "watch and wait" policy 
can be adopted. 

The control of venous hemorrhage can be 
difficult. Simple injuries to the inferior vena 
cava can be repaired primarily with intermittent 
digital pressure. In severe hemorrhage, ligation 
of the infrarenal vena cava may be necessary; in 
contrast, injury to the suprarenal vena cava 
requires reconstruction. 

Extremity Vascular Injuries 

Experience both with trauma patients and with 
noninvasive techniques are paramount in the 
management of extremity vascular trauma. A 
low threshold for angiography should be main- 
tained to diagnose ischemia. 



In those patients requiring surgical inter- 
vention, vessels are repaired by primary repair, 
interposition grafting, or bypass with vein 
(from the uninjured limb) or prosthetic mate- 
rial; 31% of patients with arterial trauma have 
concomitant venous injuries. Repair of venous 
injuries has been found to improve the outcome 
of patients with combined arterial and venous 
injuries (Martin et al., 1994; Pappas et al., 
1997). 

Grossly contaminated wounds and massive 
soft tissue and bone injuries require a multi- 
disciplinary team approach to management. 
Orthopedic injuries should be reduced with 
external fixators, and arterial and venous shunts 
can be used to minimize the ischemia time. Soft 
tissue coverage either by extraanatomical by- 
pass or muscle flaps maybe necessary to protect 
vascular repairs. 

Debridement of devitalized tissue is impor- 
tant for postoperative wound care. Fasciotomies 
are also of great importance in those patients 
with extremity injuries who have suffered 
delayed repair, extensive tissue injury, swelling, 
elevated compartment pressures, and prolonged 
hypotension. The development of compartment 
syndrome can lead to myoglobinuria, renal 
failure, and skeletal muscle necrosis. 

Controversies arise in the presence of exten- 
sive tissue damage, vascular injuries, and con- 
comitant nerve injuries. Unfortunate patients 
may be facing the future with a viable limb that 
is nonfunctioning and painful after multiple 
operations. These patients may be best served 
with a primary amputation. This is a difficult 
decision to make, and therefore should be made 
only after extensive discussion with the patient 
and family, and after the expertise of the com- 
bined team has been sought. 

Injuries of the subclavian and axillary vessels 
rarely cause upper extremity ischemia due to 
the rich collateral network at the shoulder. 
Penetrating and blunt injuries can both lead 
to brachial plexus injuries. Brachial artery 
injuries below the level of the profunda brachii 
may not present with ischemia due to the col- 
lateral supply around the elbow. Isolated injury 
to the ulnar or radial arteries may be treated 
with ligation in the presence of a complete pal- 
mar arch. 

Injuries to the femoral vessels occur in 70% 
of all arterial injuries, with penetrating trauma 



131 



VASCULAR TRAUMA 




being the most common etiology. In 20% to 35% 
of cases in which the popliteal artery is injured, 
the popliteal vein and tibial nerve are also 
involved. Blunt injuries to the knees leading 
to posterior dislocation can result in popliteal 
artery injury in 30% to 40% of patients. Single 
tibial vessel injury can generally be dealt with by 
ligation; however, if more than one vessel is 
involved, repair is advocated. 

Iatrogenic Vascular Injuries 

With an increasing use of percutaneous tech- 
niques, there is also a higher risk of iatrogenic 
vascular injuries. In a survey of 10,500 cases 
following femoral artery puncture, the in- 
cidence of complications was 0.44% (Dorfman 
and Cronan, 1991). For cardiac catheterization 
the incidence was 0.55%, whereas peripheral 
angiography resulted in a complication rate of 
0.17%. 

The most important risk is bleeding, which 
may be controlled with direct pressure after a 
catheter has been removed. Occasionally, surgi- 
cal repair is undertaken to repair the punctured 
vessel; direct repair is adequate in most 
instances. Retroperitoneal bleeding is generally 
self- limiting; however, when the patient has 
been taking anticoagulants blood or pharmaco- 
logical products maybe required to correct clot- 
ting abnormalities. 

Pseudo aneurysms can also complicate punc- 
tures in 0.5% to 5.5% of diagnostic femoral 
punctures. Most of these pseudoaneurysms 
thrombose spontaneously. Many false aneur- 
ysms respond well to ultrasound-guided com- 
pression, which has now been superseded by 
compression with concomitant injection of 
procoagulant products. Surgical intervention 
must be undertaken acutely in the presence of 
a femoral neuropathy, hemodynamic instabil- 
ity, overlying skin necrosis, and extremity 
ischemia. 

Arteriovenous fistulas usually present late 
and complicate up to 2% of cardiac catheteriza- 
tions. Although most of these fistulas throm- 
bose, a small percentage can lead to congestive 
heart failure or limb ischemia requiring either 
radiological or surgical intervention. 

Vascular injury secondary to intraarterial 
injection of drugs can lead to extensive soft 



tissue infection, mycotic aneurysm formation, 
and gangrene. 



Endovascular Treatments 

With the emergence and development of endo- 
vascular techniques, arterial trauma is being 
treated at some centers with coil embolization, 
intravascular stent grafts, and covered stent 
grafts. The use of endovascular techniques has 
minimized the need for extensive operative dis- 
sections and anesthesia; this is especially impor- 
tant in injuries involving vessels difficult to 
assess. Endovascular techniques are best uti- 
lized in institutions equipped and ready to 
handle trauma in this fashion. 



Conclusion 

Improvements in patient transport to a level- 
one trauma center combined with prehospital 
care have allowed patients to present earlier for 
treatment. The multidisciplinary team approach 
to the management of these patients has led to 
better outcomes. 

In vascular reconstructive surgery autologous 
bypasses should be used wherever possible 
(vein being harvested from the uninjured limb). 
In the event of this not being possible, the newer 
antibiotic-soaked prosthetic grafts is advocated. 

Interventional radiology with deployment of 
endoluminal stents will continue to develop its 
role in vascular trauma. It is important that vas- 
cular surgeons remain involved in all aspects of 
vascular trauma to facilitate improvements. 



References 



Bongard F, Dubrow T, Klein S. (1990) Ann Vase Surg 4:415-8. 
Cogbill TH, Moore EE, Meissner M, et al. (1994) J Trauma 

37:473-9. 
Demetriades D, Asensio JA.Velmahos G,ThalE. (1996) Surg 

Clin North Am 76:661-83. 
Dorfman GS, Cronan JJ. (1991) Radiology 178:629-30. 
Feliciano DV. (2001) World J Surg 25:1028-35. 
Halbach VV, Higashida RT, Dowd CF, et al. (1993) J Neuro- 

surg 79:183-91. 
Hughes CW. (1959) Milit Med 124:30-46. 
Johansen K, Lynch K, Paun M, Copass M. (1991) J Trauma 

31:515-9; discussion 519-22. 




» 



132 



VASCULAR SURGERY 



Martin LC, McKenney MG, Sosa JL, et al. (1994) J Trauma 

37:591-8; discussion 598-9. 
Mattox KL, Feliciano DV, Burch J, et al. (1989) Ann Surg 

209:698-705; discussion 706-7. 
Ohki T, Veith FJ, Marin ML, Cynamon J, Sanchez LA. ( 1997) 

Semin Vase Surg 10:272-85. 



Pappas PJ, Haser PB, Teehan EP, et al. (1997) J Vase Surg 

25:398-404. 
Rich NM, Hughes CW. (1969) Surgery 65:218-26. 



12 

Complications in Vascular Surgery 

Jeremy S. Crane, Nicholas J.W. Cheshire, and 
Gilbert R. Upchurch, Jr. 




Complications in vascular surgery are often life 
threatening. However, with treatment options 
for vascular disease ever widening, patient 
selection for operative procedures is governed 
not only by evidence-based treatment options 
but also by immediate and long-term compli- 
cations. The major impact complications of 
vascular surgery, in particular long-term com- 
plications, have on patient quality of life, inpa- 
tient hospital costs, psychological problems, 
and health care resources cannot be stressed 
enough. 

A useful working definition of a vascular 
surgical complication is a procedure-related 
adverse event that harms a patient (Table 12.1). 
Many of the same complications that are ubiq- 
uitous in general surgery are pertinent to vas- 
cular surgery. 



Cardiac and Pulmonary 
Complications 

The most important cause of mortality and 
morbidity after major peripheral vascular sur- 
gery is perioperative cardiac complications. 
Atherosclerosis is a systemic arterial vascular 
disorder typically involving multiple vascular 
territories in the same patient. Accordingly, 
coronary artery disease is the most frequently 
associated vascular territory affected in patients 
with peripheral arterial disease. Consequently, 
cardiac manifestations of atherosclerosis in the 



perioperative and postoperative occur, leading 
to myocardial ischemia. Naturally, the type of 
surgery is related to cardiac risk; procedures 
associated with blood pressure and cardiac 
rhythm changes with significant blood loss are 
at high risk (e.g., aortic aneurysm repair), 
whereas carotid endarterectomy carries moder- 
ate cardiac risk. 

Patients at high risk of a coronary event may 
be considered for preoperative stress testing 
or may be directly referred for coronary angio- 
graphy with possible coronary bypass or 
angioplasty prior to vascular surgery. Cardiac 
complications influence the outcome of vascu- 
lar surgery to such an extent that the existence 
of cardiac disease may lead the vascular surgeon 
to pursue an alternative surgical technique, 
such as extraanatomical bypass to treat aortic 
disease, or to cancel vascular surgical interven- 
tion altogether. 

The magnitude of the effect of coronary 
disease following vascular surgery is such that 
even 2 years following vascular surgery, 19% 
of patients experience a late cardiac event 
(Krupski, 1993). 

In addition, many elderly vascular surgery 
patients have a degree of underlying pulmonary 
disease. Therefore, postoperative respiratory 
complications are exceedingly common after 
major vascular surgery. Predisposing patient 
risk factors that may give rise to respiratory 
problems include advancing age, cigarette smok- 
ing, and the presence of chronic obstructive 
pulmonary disease. Predisposing procedure- 



133 




134 



VASCULAR SURGERY 



Table 12.1. Classification of complications by outcome 

Minor complications 
No therapy, no consequence 
Nominal therapy, no consequence; includes overnight 
admission for observation only 

Major complications 

I Requires therapy, minor hospitalization (<48 hours) 
Requires major therapy, unplanned increase in level of 
care, prolonged hospitalization (>48 hours) 
Permanent adverse sequelae 
Death 



specific risk factors include emergency proce- 
dures, lengthy general anesthesia, thoracic or 
upper abdominal incisions, massive blood 
transfusions, and a postoperative period of pro- 
longed immobility. Similar to cardiac disease, 
screening for pulmonary dysfunction preope- 
ratively can reduce postoperative respiratory 
morbidity. A thorough history to ascertain 
whether the patient suffers from dyspnea on 
exertion or has significant sputum produc- 
tion, coupled with a plain chest radiograph, 
identifies most respiratory disease. Pulmonary 
function tests and arterial blood gas samples 
further aid diagnosis. 

Preoperative therapy involving chest physio- 
therapy, smoking restriction, antibiotics, and 
bronchodilators can greatly reduce postopera- 
tive pulmonary complications. The severity of 
postoperative pulmonary complications can 
range from benign minor atelectasis, which is 
usually self-limiting, to a fulminant acute respi- 
ratory failure that carries a very high mortality. 
Accordingly, recognition and management of 
postoperative cardiac and pulmonary complica- 
tions are essential for the vascular surgeon. 



Complications of Arterial 
Reconstruction 

Vascular surgical complications arising specifi- 
cally from peripheral arterial reconstruction 
are becoming more prevalent. This is due to 
increasing numbers of operations being under- 
taken, with up to 50,000 patients being admitted 
annually to hospitals in the United Kingdom for 
treatment of lower limb ischemia. Due to the 
growing aging population coupled with the 
ever-expanding role of vascular surgery, sur- 
geons will be faced with increasing numbers of 
complications following lower extremity revas- 
cularization. As these complications may be life 
or limb threatening, treating these complica- 
tions is one of the most significant challenges 
facing the contemporary vascular surgeon. 

Specific complications associated with peri- 
pheral arterial reconstructions usually occur 
within predictable time frames, and can be 
classified as immediate, early, intermediate, or 
late (Table 12.2). For the purpose of this chapter, 
immediate complications refer to those events 
occurring intraoperatively or within the first 
24 hours after surgery, early complications 
occur less than 30 days after surgery, inter- 
mediate occur within the first year, and late 
occur after the first postoperative year. Compli- 
cations occurring immediately perioperatively 
and within the first 30 days after surgery are 
most often due to technical error or poor 
patient selection. Technical errors include inad- 
equate anastomotic suturing technique, kinking 
of the graft, and the injudicious choice of a 
poor-quality, small-diameter graft. Poor quali- 
ties of either inflow or distal run-off are pre- 
dictors of poor graft longevity. Complications 
ensuing from these errors are usually anasto- 
motic hemorrhage or a partially or completely 



Table 12.2. Time frame for 


complications of peripheral vascular 


grafts 




Immediate 


Early 


Intermediate 


Late 


Anastomotic bleeding 
Graft thrombosis 
Embolic sequelae 


Myointimal hyperplasia 
Graft thrombosis 


Accelerated atherosclerosis 
Myointimal hyperplasia 
Vein graft stenosis 
Graft infection 


Graft infection 
Atherosclerosis 
Myointimal hyperplasia 
Graft aneurysm formation 
Vein graft stenosis 



135 



COMPLICATIONS IN VASCULAR SURGERY 




thrombosed graft with possible distal embolic 
sequelae. 

The wide range of vascular grafts now avail- 
able has revolutionized vascular surgery. How- 
ever, before a graft is declared competent and 
durable, long-term follow-up is needed, as graft 
failure may only become apparent several years 
after implantation. There are three types of vas- 
cular graft: autologous (venous and arterial), 
prosthetic [e.g., Dacron, polytetrafluoroethyl- 
ene (PTFE)] and biological prosthetic (e.g., 
modified human umbilical vein graft). Once 
functioning in the arterial circuit, each type of 
graft has its inherent drawbacks, leading to 
specific complications. These complications can 
be categorized into two groups. Direct compli- 
cations involve those where the graft itself fails 
(e.g., thrombosis), whereas indirect complica- 
tions are those that relate to a graft that still 
functions (e.g., graft infection, suture line false 
aneurysm). 

Autologous Grafts 

Autologous vein is the conduit of choice for 
lower limb arterial reconstruction, as this has 
been repeatedly demonstrated in large retro- 
spective studies (Bergamini, 1991). 

Due to its anatomical constancy, the long (or 
greater) saphenous vein (GSV) is generally con- 
sidered to be the vein of choice. However, when 
the GSV has been affected by thrombophlebitis, 
been removed for previous bypass surgery, or 
has become dilated and varicosed, the short (or 
lesser) saphenous vein or upper extremity vein 
(basilic or cephalic) are often used and have 
acceptable long-term patency rates. Regard- 
less of the conduit, there remains a significant 
failure rate in autologous vein graft, which is 
associated with considerable morbidity and 
mortality. 

The most common cause of autologous vein 
graft failure in the intermediate and long term 
is the development of myointimal hyperplastic 
lesions that lead to vein graft stenosis, subse- 
quent graft occlusion, and ultimately graft 
failure. Myointimal hyperplasia (MIH) is the 
morphological lesion that underlies vein graft 
stenosis. It can be described as an abnormal 
accumulation of cells and extracellular matrix 
in the intima of the vessel wall. The mechanical 



injury caused by a bypass procedure or angio- 
plasty of the peripheral arteries can initiate and 
maintain the process of MIH. The clinical rele- 
vance of MIH is pertinent, as the development 
of MIH at the outflow anastomosis of a vein 
bypass placed in the arterial system is respon- 
sible for most complications leading to revi- 
sion surgery. Proliferating smooth muscle cells 
migrate to the injury-provoked denuded endo- 
thelial layer area and forms a hyperplastic 
lesion, which is associated with vein graft steno- 
sis and obstruction of the vascular lumen. The 
pathogenesis of MIH remains under investiga- 
tion. However, it has been established that both 
mechanical and chemical factors may induce 
this process. Arterial injury is believed to stim- 
ulate the production of growth factors, such as 
platelet-derived growth factor (PDGF), which 
have been shown to stimulate the proliferation 
of arterial smooth muscle cells and the forma- 
tion of the hyperplastic lesion in the vascular 
system. A variety of cells, including endothe- 
lial cells, macrophages, platelets, and smooth 
muscle cells, have been shown to secrete growth 
factors and cytokines. Blocking the effects of 
growth factors such as PDGF or fibroblast 
growth factor (FGF), by the administration of 
antibodies against these growth factors, may 
limit the development of the hyperplastic lesion 
(Neville and Sidawy, 1998). The role of hemody- 
namic changes in the vein graft, including 
altered vessel wall shear stress and increased 
particle residence time, has also been shown to 
play an important part in MIH growth. Inves- 
tigations aimed at modifying blood flow dy- 
namics within vein grafts to diminish MIH 
development are currently under way. 

It has been theoretically proposed that 
leaving a vein graft in situ, instead of using the 
reversed vein graft technique, may lead to fewer 
subsequent complications. Due to the natural 
tapering of the vein, an in situ vein graft has a 
better diameter match between the vein and the 
native artery at the proximal and distal ends of 
the vein graft. This makes the construction of 
both the proximal and distal anastomosis easier 
to perform and therefore reduces the probabil- 
ity of technical errors. A more suitable diameter 
match leads to more favorable blood flow fea- 
tures with decreased incidence of graft throm- 
bosis, anastomotic aneurysm, and development 
of MIH. Also as the blood supply to the vein 




> 



136 



VASCULAR SURGERY 



graft through the vasa vasorum is still partially 
intact, the vein graft remains less hypoxic. 
Conversely, as the need to use a valvulotome 
to obliterate the valves is essential, damage to 
the vessel wall may ensue, possibly leading 
to increased risk of early graft thrombosis. 
However, biological studies of the integrity of 
the vein wall in in situ and reversed vein grafts 
have failed to show significant differences. It is 
important to note that several studies have 
found no significant difference in complication 
rates resulting from either technique. 

Prosthetic Grafts 

Once incorporated into the circulation, the 
luminal surface of a prosthetic graft becomes 
lined with fibrinogen, followed by fibrin and 
red and white blood cells. This layer is known 
as a pseudointima. Whereas prosthetic grafts 
perform well in a high-flow environment (e.g., 
in aortic reconstruction), their performance in 
the infrageniculate position is poor compared 
with that of autologous vein. The main problem 
leading to poor long-term graft patency is the 
increased thrombogenicity of prosthetic distal 
bypass grafts. Some have advocated the case for 
primary amputation being first-line treatment 
when autologous vein is unavailable for a distal 
bypass. 

Originally, Dacron grafts were used as the 
material of choice for distal bypass. However, 
due to very poor patency rates, PTFE took over 
as the prosthetic conduit of choice. As well as the 
complications resulting from increased throm- 
bogenicity of prosthetic grafts, graft infection 
has dire consequences, described later in this 
chapter. 

Lower Limb Graft Occlusion 

As the majority of vascular grafts in the lower 
limb are performed for critical limb ischemia, 
graft occlusion often reverts the limb back to 
its original critical state or worse. Moreover, 
ischemia may be compounded due to a reduc- 
tion in collateral flow caused by division of 
vessels during surgery, by physiological reduc- 
tion of collateral flow as a result of a successful 
graft, or by thrombosis extending distally into 
the run-off vessels. 



Diagnosis 

The diagnosis of an occluded infrainguinal graft 
is made clinically, but can be confirmed by 
duplex ultrasound. Presentation ranges from 
a sudden onset of a reduction in pain-free 
walking distance to acute limb ischemia. From 
the history, the time of onset of occlusion can be 
ascertained, and the time frame between ori- 
ginal surgery and occlusion can aid in the 
etiology (Table 12.2). 

Management 

If a thrombosed graft presents within hours or 
days of the occlusion, the limb is usually viable 
and the aim is to restore graft patency. In the 
absence of contraindications, thrombolysis is 
indicated. Hemorrhage at the site of catheter 
thrombolysis insertion occurs in 10% to 30% of 
cases and is the most common complication. 
The most catastrophic complication of throm- 
bolysis is intracranial hemorrhage that is often 
fatal, in particular in elderly patients with a 
history of stroke. Bleeding into other organs is 
also commonly recognized. Due to the high 
complication rate during thrombolysis, if an 
occluded lesion does not render the patient crit- 
ically ischemic, it can be argued that thrombol- 
ysis is not justified. 

If after successful thrombolysis an underlying 
lesion is identified, the patient should be 
heparinized and the lesion corrected. Jump 
grafting or patch angioplasty is an appropriate 
procedure. When a myointimal hyperplastic 
lesion postthrombolysis is found, transluminal 
balloon angioplasty, patch angioplasty, or inter- 
position grafting can be used, as well as jump 
grafting. Autologous vein is preferable for sec- 
ondary reconstructions. 

An emergency revascularization is the man- 
agement of a graft occlusion associated with a 
threatened limb. Loss of neurological function 
coupled with a tender and tense calf is an indi- 
cation for urgent surgery. The procedure differs 
depending on the type of graft. The aim of treat- 
ment of a thrombosed vein graft threatening the 
limb is restoration of graft flow with correction 
of any underlying abnormality. The distal anas- 
tomosis should be opened and the graft and 
distal vessels cleared of thrombus. On-table 
angiography should be performed once flow has 
been restored. Then any abnormality can surgi- 



137 



COMPLICATIONS IN VASCULAR SURGERY 




cally be corrected as previously described. With 
a prosthetic graft, the aim of an emergency 
procedure is restoration of the original graft 
function with or without correction of an 
underlying cause. The long-term patency of 
salvaged occluded autologous or prosthetic 
infrainguinal grafts is disappointing, and com- 
plete replacement with a new vein graft, reserv- 
ing graft revision for those patients' with no 
better bypass options, has been advocated. Fol- 
lowing an emergency procedure for lower extre- 
mity graft thrombosis, particularly if there has 
been significant preoperative ischemia, the sur- 
geon should have an extremely low threshold 
for performing a fasciotomy to prevent muscle 
necrosis. 



Complications of 
Aortic Reconstruction 

Reconstruction of the aorta for atherosclerotic 
aneurysmal or occlusive disease brings about a 
different spectrum of complication when com- 
pared with lower-extremity revascularization. 
The aortic graft used is either Dacron or PTFE, 
as there is no autologous conduit large enough 
to replace the aorta. Due to the high flow 
through a large-caliber vessel, prosthetic grafts 
replacing native aorta perform well and have 
well-documented excellent long-term results. 
The two most common local complications after 
aortic reconstruction are occlusion and graft 
infection (see Infected Prosthetic Graft, below). 
When an aortic graft or the limb of an aorto- 
bifemoral graft occludes, the limb is usually 
viable. Occlusion of a single limb of an aortic 
graft is most often caused by anastomotic stric- 
ture at the femoral anastomosis. If the onset of 
the occlusion is in the early phase (within 30 
days) postsurgery and the graft can be throm- 
bolyzed successfully, subsequent patch angio- 
plasty or distal jump grafting is usually 
successful. Thrombolysis of retroperitoneal 
knitted Dacron grafts must be performed with 
caution because of the risk of extravasation of 
blood into the retroperitoneum. Thrombectomy 
using ring strippers or adherent clot catheters is 
more successful than using ordinary thrombec- 
tomy balloon catheters. It is more common that 
the time of graft failure is not known and is 
likely to be of several weeks' duration. In these 



circumstances, open graft thrombectomy or 
thrombolysis is not possible, and so common 
practice involves revascularizing the affected 
limb with a femoral artery-to-femoral artery 
crossover graft taken from the contralateral 
limb. Other extraanatomical reconstructions are 
also feasible in this situation. 

When the body and limbs of an aortic graft 
are thrombosed, proximal disease advancing 
into the graft from the proximal aorta is usually 
the cause. This complication can be avoided by 
placing the initial graft on the aorta near the 
level of the renal arteries. However, once a diag- 
nosis of total aortic graft occlusion is made, 
most surgeons would undertake an axillob- 
ifemoral graft as an option; redo aortic grafting 
is major corrective surgery carrying great risk 
to the patient. Occasionally, a failed aortic graft 
will not produce new or acute symptoms 
because of the development of collateral vessels 
in a relatively mobile patient or because immo- 
bility masks claudication symptoms. If this were 
the case, it may be that the patient will present 
many months after the graft has failed, with 
either nonhealing ulcers or rest pain. In either 
case, and to thwart the onset of critical ischemia, 
a complete graft replacement is the only option, 
as at this stage the graft is usually unsalvageable. 

Infected Prosthetic Graft 

One of the most challenging complications in 
vascular surgery is a graft infection. Infection 
radically changes patient outcomes and is both 
life and limb threatening. Quantifying incidence 
of graft infection is not easy, as many reports are 
anecdotal and this is compounded by the long 
and variable time period between original sur- 
gery and evidence of graft infection. The 
incidence of graft infection, however, is influ- 
enced by anatomical site of graft, indications for 
original intervention, underlying disease, and 
the patients' host defense. The literature quotes 
figures that range between 0.7% and 7% for 
graft infection after aortic surgery. Prosthetic 
graft infection is more common after emer- 
gency procedures (e.g., ruptured abdominal 
aortic aneurysm), or when the graft is anasto- 
mosed to the femoral artery in the groin or sited 
superficially (e.g., axillofemoral bypass). When 
and how a graft becomes infected is often 
difficult to ascertain, and there are multiple risk 




138 



VASCULAR SURGERY 



Table 12.3. Risk factors for graft infection 

Local factors 

Prosthetic graft 

Groin invasion 

Emergency or lengthy surgery 

Leg ulceration or gangrene 

Postoperative wound infection 

Systemic factors 
Diabetes mellitus 
Malnutrition or obesity 
Chronic renal failure 
Malignancy 
Steroid therapy 



factors for graft infection (Table 12.3). However, 
there are three modes whereby the graft may 
become contaminated: (1) the infection maybe 
seeded onto the graft at the time of surgery 
(perioperative contamination), (2) subsequent 
bacteremia may seed the graft, and (3) direct 
spread from a nearby source (e.g., skin or duo- 
denum) may occur. 

Presentation 

Any symptoms and signs of sepsis in a patient 
with a vascular graft should alert the clinician 
to the possibility of a graft infection. With a sub- 
cutaneous graft, localized erythema and cellu- 
litis or a discharging sinus may be clinically 
visible over the length of the graft, or prosthetic 
material maybe visualized eroding through the 
skin. With a deep infected graft (e.g., an aortic 
graft), the patient may present with vague 
symptoms and have pyrexia of unknown origin. 
An infected aortic prosthesis may form an aor- 
toduodenal fistula and present with upper gas- 
trointestinal bleeding that may be mistaken for 
peptic ulceration. Less commonly, lower gas- 
trointestinal hemorrhage may be seen from 
aortoenteric fistulas into distal bowel sites. 
Alternatively, signs of septic emboli to the 
extremities may present as a purpuric rash. 
Other more rare sequelae of a graft infection 
included metastatic mycotic aneurysm forma- 
tion, anastomotic pseudoaneurysm formation, 
or anastomotic hemorrhage. With all these 
potential sequelae, the clinician must have a 
high index of suspicion in the presence of a 
prosthetic graft. 

Making a definitive diagnosis of graft infec- 
tion can be difficult. The presence of a perigraft 



collection should be determined whenever pos- 
sible. Determining which organisms are in- 
fecting a graft is also imperative to help guide 
antibiotic choice following graft removal. Once 
these two factors have been elucidated, the like- 
lihood of infection must be balanced against the 
general condition of the patient, the extent of 
the revision surgery required, and the necessity 
for immediate intervention. 

Investigations 

Having bacteriological cultures done on swabs 
taken from a discharging sinus may identify 
the organism involved. Blood cultures may 
also be positive, particularly if blood is drawn 
from the femoral artery, downstream to a pos- 
sible graft infection. The organisms most com- 
monly isolated from blood or from wounds 
are gram-positive bacteria, Staphylococcus and 
Streptococcus species. A particularly worrying 
phenomenon is the continued rise in the num- 
ber of reports of methicillin-resistant Staphylo- 
coccus aureus (MRSA) isolated from infected 
grafts. This organism and gram-negative infec- 
tions are associated with severe systemic sepsis, 
and experience in our unit and elsewhere has 
shown these organisms to carry a less favorable 
outcome in terms of limb salvage and mortality 
than infection with less virulent organisms. 
Early graft infection is usually associated with 
virulent organisms such as S. aureus, Strep- 
tococcus faecalis, Escherichia coli, Klebsiella, 
Pseudomonas aeruginosa, and Proteus or mixed 
pathogens. Late graft infections are usually 
caused by less virulent bacteria (e.g., Staphylo- 
coccus epidermidis, which is a common skin 
commensal). Interestingly, this organism has 
been cultured from grafts that have been 
removed for purposes other than infection, 
which may signify that only a small proportion 
of such contaminated drafts develop graft- 
threatening infection. The type of antibiotic 
therapy differs between centers, but it is prudent 
to discuss an appropriate antibiotic regimen 
with a microbiologist. 

There are a few imaging techniques to eluci- 
date the presence and extent of a graft infection. 
Computed tomography (CT) scanning can help 
in demonstrating fluid collections around a sus- 
pected infected graft. The presence of gas within 
a perigraft collection is pathognomonic of an 
infection; however, around an aortic graft, gas 



139 



COMPLICATIONS IN VASCULAR SURGERY 




may represent resolving thrombus. Computed 
tomography scanning may also not be able to 
differentiate free gas that is around a graft and 
gas within the bowel that is adhered closely to 
the graft. Computed tomography-guided aspi- 
ration of perigraft fluid may be undertaken to 
aid in diagnosis; however, this carries the risk of 
introducing infective seedlings into a poten- 
tially sterile field. 

A radioisotope-labeled white blood cell scan 
enables localization of occult sites of infection, 
as labeled granulocytes concentrate around an 
infected focus. Magnetic resonance angiogra- 
phy of the aorta is believed to be complemen- 
tary to a tagged white cell scan and may increase 
the likelihood of appropriately diagnosing a 
graft infection. Angiography, even though 
unable to pinpoint sites of infection, is neces- 
sary for preoperative planning for revision 
surgery. It demonstrates the nature of distal 
run-off and the state of arteries that may be 
used in an extraanatomical bypass. Occasionally 
it may aid diagnosis in an aortoenteric or aor- 
tocaval fistula. Gastrointestinal endoscopy to 
rule out aortoduodenal fistula must be the first- 
line investigation in a patient presenting with 
hematemesis or melena. 

Management 

Graft conservation involves local debridement 
of infected tissues surrounding a superficial 
graft, followed by a primary wound closure and 
local irrigation with gentamicin antibiotic solu- 
tion via suitably placed drainage catheters or 
impregnated collagen sponge or beads. Another 
method of graft preservation involves covering 
the debrided area and graft with a vascularized 
omental or muscle flap. This form of manage- 
ment is strongly proposed for localized groin 
wound infection in the presence of a patent 
graft and is said to be associated with a low mor- 
tality, high limb salvage rate and an acceptable 
incidence of recurrent infection. The decision to 
excise an infected graft should be undertaken 
carefully, and whenever possible the patient's 
condition needs to be optimized prior to the 
operation. The best-case scenario is when ade- 
quate collateralization has taken place and no 
further revascularization procedure is required 
once the graft is removed. More commonly, 
though, graft removal results in a return to the 
preoperative state, often rendering one or both 



legs ischemic, and at the time of removal of 
the infected graft further revascularization is 
required. 



Complications Related to 
Disorders of Coagulation 

The acute thrombosis of an arterial procedure 
in the perioperative period is usually seen 
before the patient exits the operating room and 
may occur up to 12 hours following surgery. 
Dealing with unexplained thrombosis intra- 
operatively or in the immediate postoperative 
period is one of the greatest uncertainties faced 
by the vascular surgeon. 

Hypercoagulability as grounds for an unex- 
plained vascular thrombosis poses a challeng- 
ing clinical problem. Graft failure in the 
perioperative period is presumed to result from 
technical errors in the construction of the 
anastomosis, problems with the choice of 
and quality of vascular conduit, or unsuitable 
patient selection. Only once these other factors 
as a cause of failure have been excluded can the 
diagnosis of abnormal coagulability be formu- 
lated. When it becomes apparent there is abnor- 
mal coagulation (i.e., heparin is not preventing 
clotting in the operating field or when there is 
an immediate thrombosis of a vascular repair), 
the diagnosis can be confirmed only by the 
analysis of a blood clotting profile. It therefore 
stands to reason that detection of clotting dis- 
orders prior to surgery is most often associated 
with a favorable surgical outcome. It is with 
experience that a surgeon develops an intuition 
as to which reconstructions are likely to succeed 
and has some ideas as to the types of complica- 
tions that may occur. Likewise, with experience 
a surgeon develops a feel for the character- 
istic presentations of atherosclerotic occlusive 
disease. When abnormal or unexplained throm- 
bosis is diagnosed (e.g., a thrombosed supra- 
renal aorta or upper extremity thrombosis in a 
patient who is not diabetic and has little evi- 
dence of atherosclerotic occlusive disease else- 
where), the surgeon should investigate clotting 
disorders as a possible cause. Unusual angio- 
graphic findings, particularly young patients 
who present with an occlusion in one limb with 
pristine vasculature in the contralateral limb, 
should prompt an investigation into the coagu- 




> 



140 



VASCULAR SURGERY 



lation system. The role of screening for poten- 
tial thrombophilia states in vascular surgical 
patients has been studied. Approximately 10% 
of all patients scheduled for a variety of vascu- 
lar surgical procedures had test results indicat- 
ing a potential hypercoagulable state. The three 
most common clotting disorders are heparin- 
induced platelet aggregation, lupus anticoagu- 
lants, and protein C deficiency. The incidence of 
infrainguinal graft occlusion within 30 days was 
27% among patients who were in the hyperco- 
agulable group compared with 1.6% in patients 
who were not in this group. At present, an estab- 
lished screening program does not exist to 
exclude the wide variety of coagulation disor- 
ders present in vascular surgical patients. We 
depend on taking a careful history and clinical 
examination to identify those patients who may 
be at a risk of abnormal clotting due to an inher- 
ent thrombophilic status. At this time this is 
probably the more cost-effective and efficient 
means to establish a hypercoagulable diagnosis 
prior to operation. 

Complications of 
Interventional Vascular 
Radiological Procedures 

As more minimally invasive interventional radi- 
ological procedures are being used as first-line 
treatment for occlusive and aneurysmal vascu- 
lar disease, the incidence of complications is 
rising accordingly. These complications can be 
divided into three categories: (1) puncture-site 
related, (2) catheter related, and (3) systemic 
complications. 

About 0.5% of femoral punctures and 1.7% 
of axillary punctures have been reported to 
require treatment for a complication. Hema- 
tomas at the puncture site are not usually con- 
sidered a complication and rarely need surgical 
treatment. Large hematomas (e.g., retroperi- 
toneal, pelvic, and anterior abdominal wall 



hematomas) that require surgical intervention 
occur more often in patients with coagu- 
lopathies and in patients who are obese. The risk 
of puncture-site pseudoaneurysm has been 
shown to increase with increasing catheter size 
and when a low puncture site is used. Preven- 
tion of puncture-site hematomas and pseudoa- 
neurysms can be achieved by using the 
smallest-caliber catheter possible to perform 
the necessary intervention. Accordingly, there 
are fewer puncture-site complications with 
angioplasty procedures when compared with 
stenting, as the catheter needed to place a stent 
is of a larger caliber. 

Catheter-related complications include the 
formation of arterial dissections, subintimal 
injections, and embolization from the catheter. 
Fortunately, as the dissections produced are ret- 
rograde and are pushed closed by the direction 
of blood flow, most of these complications are 
asymptomatic. However, some acute dissections 
become extensive chronic dissections or cause 
acute occlusion. Catheter thromboembolism 
occurs in less than 0.5% of cases. 

There are myriad systemic complications 
associated with angiographic procedures. Exam- 
ples include vasovagal syncope, cardiac arrest 
and arrhythmias, myocardial infarction, and 
nausea and vomiting. Contrast reactions can be 
reduced by adequate hydration and by using low 
osmolar contrast in high-risk patients. Rarely 
radiation injury in the form of a skin burn o ccurs 
when the procedure is lengthy and the patient is 
exposed to over 2 Gy. 



References 



Bergamini TM, Towne JB, Bandyk DF, Seabrook GR and 

Schmitt DD. (1991) J Vase Surg 13:137-47; discussion 

148-9. 
Hannon RJ, Wolfe JH, Mansfield AO. (1996) Br J Surg 83: 

654-8. 
Krupski WC, Layug EL, Reilly LM, Rapp JH, Mangano DT. 

(1993) J Vase Surg 18:609-15; discussion 615-7. 
Neville RF.SidawyAN. (1998) Semin Vase Surg 11:142-8. 



13 



Vascular Access 

David C. Mitchell and C. Keith Ozaki 




Vascular access is required to assist in the man- 
agement of patients requiring frequent venous 
or arterial cannulation. Its principal role is to 
provide circulatory access for hemodialysis, al- 
though such techniques are also used to pro- 
vide chemotherapy, intravenous nutrition, and 
access for plasma exchange. 

The two techniques used are either implant- 
able synthetic lines, which may have one or 
more lumina, or a surgically created fistula 
between the arterial and venous circulation. 
This latter approach may involve joining artery 
and vein together, the so-called autogenous 
arteriovenous (AV) access [arteriovenous fistula 
(AVF)], or the insertion of a synthetic bridge 
graft between artery and vein (nonautogenous 
AV access). 



Principles of Vascular Access 

The guiding principles of access surgery are to 
use AVF in preference to synthetic grafts, which 
are preferred to in-dwelling central venous 
catheters. Access should be sited as far distally 
in the chosen limb as possible. The principle is 
to conserve veins to permit proximal revision if 
the initial procedure fails. An exception is very 
old patients, who often have poor distal vessels. 
Some young patients may have strong wishes to 
avoid visible forearm scars. The nondominant 
limb should be used wherever possible. 

In patients who may require vascular access, 
venepuncture, or the insertion of intravenous 



cannulae in the main named veins of the upper 
limb, should be avoided. If central venous 
catheters are required, they should be sited in 
the jugular and not the subclavian veins to avoid 
central venous stenosis in the draining veins of 
the upper limb. 

Standards 

The United States has led the way in this area 
with the publication of specific guidelines 
[National Kidney Foundation-Disease Out- 
comes Quality Initiative (NKF-DOQI) (www. 
kidney.org/professionals/doqi/guidelines)]. 
These guidelines were a response to the rapid 
growth in patient numbers and costs associated 
with treatment for renal failure. A multidiscipli- 
nary group of nephrologists, nurses, vascular 
and transplant surgeons, and interventional 
radiologists reviewed the world literature. The 
resulting document was approved by all the rep- 
resented national societies prior to adoption. 

The principal recommendations are that the 
majority of patients starting dialysis should do so 
using a native vessel AVF at the wrist. Grafts 
require six times as many interventions as AVF to 
achieve the same patency, and their use is dis- 
couraged. Central catheters have inferior patency 
and are discouraged for "permanent" access. 
Their principal role should be for emergency or 
temporary access. Since the distribution of these 
standards, placement rates for simple fistulas has 
increased 35% in the United States. 



141 




> 



142 



VASCULAR SURGERY 



The Europeans have not produced a similar 
document, although it is hoped that a National 
Service Framework for renal disease manage- 
ment in the United Kingdom will set standards 
for vascular access within the National Health 
Service. 

Observational studies have helped to define 
standards. The Dialysis Outcomes and Practice 
Patterns Study (DOPPS) was a prospective 
global study of practice in seven countries 
involving 309 facilities and over 9500 patients. 
It showed that there are significant differences 
between varying health cultures in the devel- 
oped world. The AVF rates varied from 0% to 
87% in the United States, and 39% to 100% 
in Europe. Graft usage in new hemodialysis 
patients ranged from 2 to 24%. Central venous 
lines were particularly common in the United 
Kingdom and the United States but rare in Japan 
and Italy. 

These differences are not dictated solely by 
differences between patients, but principally by 
clinician preference. This study has clarified 
what is being done at present to provide access. 
By demonstrating widely differing practice, 
it informs the debate over ideal practice and 
can guide future trials to answer questions 
about treatment where widely differing views 
are held. 

Interpretation of the literature is further com- 
plicated by differences in reporting of the re- 
sults of access surgery. A recent study from the 
United States should help to standardize report- 
ing in the future (Sidawy et al., 2002). 



Types of Access 



The principal modes of vascular access are 
autogenous AVF, synthetic AV bridge grafts 
(nonautogenous AV access), and in-dwelling 
synthetic central venous catheters. Considera- 
tion of patient demographics, anatomical suit- 
ability, and other factors are important in the 
preoperative planning for access (Table 13.1). 

Arteriovenous Fistula 

Most clinicians agree that the most durable 
form of vascular access is the AVF. The prefer- 
ential site is between the cephalic vein and 
the radial artery at the anatomical snuffbox, or 
the wrist, in the nondominant upper limb, the 



Table 13.1. Preoperative planning for autogenous or 
nonautogenous arteriovenous fistula (AVF) placement 

Consider patient age, projected life span, time to 

initiation of dialysis, dominant hand 
Elicit history of previous access attempts/subclavian 

vein cannulations; consider contrast venography to 

rule out central venous pathologies 
Check Allen's test, upper extremity blood pressures, 

arterial pulse exam — > if abnormal then segmental 

arm pressures, plethysmography — > if abnormal 

then consider ultrasound or angiography 
Venous exam with tourniquet — > if no clear conduit 

then duplex; protect that vein from 

puncture/trauma 
Ensure patient is optimized for operation (e.g., address 

cardiac, metabolic, volume status, nutritional, 

infectious issues) 
Consider risks of steal in elderly and diabetic patients 

undergoing proximal access construction 



Brescia-Cimino fistula (Brescia et al., 1966). The 
end of the vein is joined to the side of the radial 
artery, usually under local anesthesia. 

The success of this procedure is dependent on 
the quality of the vein and artery and the tech- 
nical skill of the surgeon. Where good vessels 
are not clinically evident, duplex ultrasonogra- 
phy or venography may help to identify the 
best sites for access formation. The suggested 
sequence for AVF placement is listed in Table 
13.2). 

Widely varying success rates are quoted, but 
about 60% of wrist AVFs mature and become 
useful for dialysis. The AVFs are robust once 
established, but may take many (typically 6 to 
12) weeks to mature. Many factors may affect 



Table 13.2. Suggested sequence for AVF placement 

1. Autogenous AVF in hand/forearm (nondominant 
before dominant) 

2. Upper arm autogenous AVF (usually cephalic before 
basilic) 

3. Forearm nonautogenous AVF 

4. Upper arm nonautogenous AVF 

5. Upper arm or thigh autogenous (using transposed 
or translocated saphenous/superficial femoral vein) 
AVF 

6. Thigh nonautogenous AVF 

7. Central configurations such as axillary artery to 
contralateral axillary vein, subclavian artery to 
subclavian vein 



143 



VASCULAR ACCESS 




maturation, such as the size of artery and vein, 
change in flow, and the presence or absence of 
arterial disease (e.g., diabetes). In patients in 
whom the fistula is failing to become prominent 
within a few weeks, duplex ultrasound scanning 
can examine flow rates and identify problems. 
Surgeons willing to undertake revision may 
produce higher success rates. The principal 
reasons for failure are damaged or inadequate 
veins and (more rarely) inadequate arteries. In 
such cases, alternative forearm veins may be 
used. The basilic vein on the ulnar side of the 
forearm is often large and may not have been 
traumatized by venesection. This can be mobi- 
lized and swung across the forearm to the radial 
artery, or anastomosed to the adjacent ulnar 
artery if large enough, although the failure rates 
for this procedure are higher. 

Any vein can be damaged by repeat venesec- 
tion or placement of in-dwelling catheters. Such 
veins develop areas of fibrosis that cannot dilate 
when subjected to arterial flows. For this reason, 
in those in whom the need for access can be anti- 
cipated, every effort should be made to avoid 
needling the cephalic vein, the antecubital veins, 
or the subclavian vein. 

The radial artery at the wrist is often insuffi- 
cient in the elderly and particularly those with 
diabetes. The artery usually shows evidence of 
arteriosclerosis with calcification. The artery is 
unable to increase its flow rate in response to 
fistula formation. The result is failure of the vein 
to enlarge, or thrombosis. In this situation, it 
may be better to place the fistula more proxi- 
mally in the limb, typically at the elbow. Pre- 
operative evaluation can assist in identifying 
suitable patients for distal AVFs (Malovrh, 
2002). 

The standard procedure at the elbow is an 
end of cephalic vein to side of brachial artery 
fistula. This can usually be fashioned through 
a transverse antecubital incision or two lon- 
gitudinal incisions under local anesthesia. The 
depth of the basilic vein within the arm com- 
plicates its use as a direct fistula. This vein, 
however, may be easily mobilized through a lon- 
gitudinal incision in the medial arm. The basilic 
vein is then divided distally, tunneled subcuta- 
neously, and anastomosed to the brachial artery 
in the distal arm. The procedure can be per- 
formed using minimally invasive techniques. 
Such transposed fistulas may give good service 
over many years. This technique may also be 



applied to the lower limb in patients with good 
arterial circulation, by transposing the long 
saphenous vein subcutaneously in the thigh and 
anastomosing it to the distal superficial femoral 
artery. 

Synthetic Access Grafts 
(Nonautogenous 
Arteriovenous Access) 

Where veins are inadequate, or dialysis is 
needed urgently (within 2 weeks), AVF may be 
inappropriate. In such circumstances, access can 
be rapidly established using synthetic bridge 
grafts between suitable arteries and veins. The 
most common material for this is polytetra- 
fluoroethylene (PTFE), which may be cannu- 
lated within a week if required. Dacron is not 
widely used for access as it is difficult to needle, 
but newer composite grafts that are reported 
to bleed less after cannulation are available. 
The role of these grafts has yet to be clearly 
established. 

As veins are often deficient, grafts tend to 
be placed more proximally in the limb than 
AVFs. They may be placed in either straight 
or curved/looped configurations. No data exist 
demonstrating that one configuration is signi- 
ficantly better than any other. Grafts should be 
appropriately sized with the arterial anastomo- 
sis not bigger than 6 mm to avoid excessive flows 
and either vascular steal or high output cardiac 
failure. 

Although easier to establish, PTFE grafts are 
known to have higher thrombotic and septic 
complication rates, and to need more frequent 
revision than native AVFs. For this reason, it is 
advised that AVFs are preferred. This has never 
been formally tested in a prospective random- 
ized trial. In the elderly patient with limited 
life expectancy, a rapidly established graft may 
provide superior access with sufficient durabil- 
ity, compared to several unsuccessful attempts 
to establish an AVF. 



Central Venous Catheters 

A well-organized renal failure service should 
anticipate the need for access in patients ap- 
proaching end-stage disease, and appropriate 
access surgery should be planned in advance. 




> 



144 



VASCULAR SURGERY 



This counsel of perfection is not always realized 
as patients may present acutely after a long 
period of stability. 

Those requiring emergency access (e.g., im- 
mediate need for dialysis) need the insertion 
of a dual-lumen central venous catheter. These 
should be left in place for as short a time as pos- 
sible. Definitive access surgery should follow as 
a matter of urgency. 

Central catheters become infected easily, and 
often have to be removed or re-sited. In addi- 
tion, they stimulate fibrosis in the veins and can 
cause stenosis. For this reason, subclavian lines 
are to be avoided, as the loss of the subclavian 
vein prejudices further access in the ipsilateral 
upper limb. Lines should be placed via the 
jugular route if at all possible. If access is needed 
only for a few days, then in the absence of suit- 
able jugular veins the femoral veins may be 
used. 

Patients needing more permanent catheters 
(longer than 3 weeks) should have tunneled 
catheters inserted. Placement considerations 
are the same as for temporary lines. Tunneled 
catheters should be cuffed, as the cuffs reduce 
the risk of infection from the site of skin 
entry. 



Failure to Mature 

Failure to mature is the most common problem 
seen in AVFs. Fistulas demonstrate significant 
increase in flow within 48 hours and enlarge 
thereafter. Most successful AVFs are capable 
of being needled in about 6 to 8 weeks after 
formation, although longer may be required. 
Detecting those that will fail to mature may be 
difficult. Ideally, AVFs should be placed well 
in advance of the time that they are likely to 
be required. This permits time for assessment 
and revision if the fistula is not developing 
satisfactorily. An alternative approach if time 
is pressing is to scan the fistula with ultra- 
sound soon after formation. Flow rates greater 
than 500 mL/min signify that the fistula is likely 
to develop successfully. Scanning can also be 
used for the early detection of technical 
deficiencies, stenoses, and other problems. In 
constructions that are failing to mature, some 
groups advocate early liberal use of fistulograms 
to aid in diagnosis and therapeutic planning. 
Others rely primarily on the diagnostic vascular 
laboratory, and reserve fistulography for cases 
in which duplex scanning is equivocal or 
unavailable. 



Complications of 
Vascular Access 

Arteriovenous Fistula 

Once the AVF is established, AVF revision 
rates are low at about 15% or less per annum. 
Fistulas are robust and withstand multiple 
cannulations well. 

Technical Failure 

The most immediate complication of AV access 
is that the fistula occludes shortly after forma- 
tion. This maybe due to obvious problems such 
as inadequate vessels or to technical imperfec- 
tions in the procedure. In the former, re-siting 
the fistula is the best course of action. In the 
latter situation, revision, often under local 
anesthesia, may salvage a functioning AVF. This 
is rarely an emergency and can be managed 
during the next available elective operating 
session. 



Vascular Stenosis 

Vein stenosis and aneurysmal dilatation are 
both seen following development of AVFs. The 
former may compromise both the quality of 
dialysis and the longevity of the fistula. Some 
centers advocate routine fistula scanning, but 
this has not yet been shown to be an effective 
way of monitoring fistulas for complications. 
Where obvious stenoses exist, or dialysis is 
inadequate despite an apparently satisfactory 
fistula, then ultrasound may reveal a stenosis. 
Stenoses greater than 50% or AVFs with flows 
below 500 mL/min are more likely to fail and 
may need revision. Stenoses within 1 to 2 cm of 
the anastomosis are most easily dealt with by 
surgical revision. This can be undertaken under 
local anesthesia with an occlusive tourniquet. 
The fistula is ligated and divided and reanasto- 
mosed to an adjacent portion of artery. More 
remote stenoses may often be dealt with by 
angioplasty. If this fails, then short skip grafts of 
vein may bypass a stenosis and permit continu- 
ous use of the fistula without the need for tem- 
porary lines. 



145 



VASCULAR ACCESS 




Vascular Steal and High-Output 
Cardiac Failure 

Proximal fistulas tend to have higher flow rates 
and may divert blood from the hand. The risk 
of vascular steal should be borne in mind in 
proximal fistula. It is uncommonly seen (about 
1% to 8%), but the neurological effects are irre- 
versible if the steal is not quickly corrected 
(Tordoir et al., 2004). Any suspicion of hand 
ischemia demands immediate attention, begin- 
ning with bedside examination. Temporary 
finger occlusion of the AVF can assist in deter- 
mining whether there is an inflow problem, or 
whether the arterial flow distal to the arterial 
anastomosis has been interrupted. If hand arte- 
rial perfusion returns to baseline with AVF com- 
pression, then inflow is inadequate to maintain 
flow to the hand and through the AVF. If the 
extremity remains ischemic, then there may be 
a problem with the arterial anastomosis that 
occludes outflow, or thromboembolic complica- 
tions. Segmental arm and finger pressures, fol- 
lowed by repeated exam while the access is 
occluded, confirm the diagnosis. If inflow is 
inadequate, then improvement of distal pres- 
sures and symptoms should occur while the AVF 
or graft is compressed. 

If steal physiology is demonstrated, then 
appropriate urgent imaging to screen for occult 
inflow arterial occlusive disease may be under- 
taken, and lesions addressed. Fistula revision is 
often required. If preservation of the fistula is 
not essential, taking it down and creating a 
new one is probably the procedure of choice. 
The alternatives are banding to reduce flow, or 
bypass from the artery at least 6 cm above the 
anastomosis to an artery distally and then liga- 
tion of the artery immediately beyond the anas- 
tomosis. This last procedure is known as the 
distal revascularization-interval ligation (DRIL) 
procedure. It is gaining in popularity as it takes 
flow to the distal arterial tree from above the 
fistula and prevents steal due to reversed flow in 
the artery distal to the anastomosis. 

Banding is a complex procedure, necessitat- 
ing intraoperative flow measurement. Flows 
may fluctuate during the procedure, but ideally 
the banding should produce a flow between 800 
mL and 1.5 L per minute. Success rates for this 
procedure are low, and it is not recommended if 
an alternative is available. Postoperative throm- 
bosis is common and maybe due to overvigor- 



ous banding. If this happens, the fistula is 
usually lost. 

High fistula flows can also cause high-output 
cardiac failure, which is seen more commonly in 
proximal AVFs. In these cases, fistula revision to 
reduce flows is required. The choices include 
sacrificing the fistula and siting new access, or 
attempting a flow reduction procedure. 

Infection 

Infection is rare, but if associated with bac- 
teremia, abscess, or aneurysm formation, revi- 
sion is usually required. The clinical features 
are of an area of inflammation in relation to 
the fistula, sometimes with rapid enlargement. 
Fever and rigors, particularly on dialysis, may 
be seen, but are not a universal feature. Sudden 
rapid bleeding can occur at infected needling 
sites. Immediate pressure and placement of a 
skin suture will achieve acute control, but 
should be followed by a definitive revision in 
most cases. 

Thrombosis 

Thrombosis is uncommon, but when it occurs 
it can often be retrieved by acute intervention, 
with salvage of the fistula and avoidance of tem- 
porary central lines. Overdialysis is an uncom- 
mon cause of fistula thrombosis, but should be 
suspected if no stenosis can be found. Imaging 
of the whole fistula and ipsilateral central veins 
should always follow surgical or radiological 
declotting. Dilatation of fistula or central vein 
stenosis may be required to restore normal 
function. When the stenosis is in close proxim- 
ity to the anastomosis, then surgical revision to 
an adjacent proximal arterial segment is often 
the best procedure. 

The complications associated with trans- 
posed fistulas are the same as those seen with 
other native AVFs. They tend to have high flows, 
so cardiac failure and steal are more common in 
these fistulas. 

Arteriovenous Access 
Grafts (Nonautogenous 
Arteriovenous Access) 

Arteriovenous grafts can usually be established 
easily, with a high technical success rate. How- 




> 



146 



VASCULAR SURGERY 



ever, complications are common, and the fre- 
quent need for intervention should prompt the 
surgeon and dialysis team to undertake regular 
surveillance. 



Thrombosis 

Thrombosis is usually the consequence of 
intimal hyperplasia at the venous end of the 
graft. This can be recognized by regular intra- 
dialysis monitoring of flow or pressure. It has 
been shown that monitoring of flow or pressure 
can detect developing graft stenosis. Timely 
intervention can prolong graft survival. One 
approach is to use balloon angioplasty where 
possible, keeping surgical revision to a 
minimum. Where thrombosis occurs, prompt 
declotting undertaken by either an endovascu- 
lar or open approach can retrieve graft function. 
It is important to identify and correct the cause 
of the thrombosis or further thrombosis is 
likely. There is no clear evidence whether open 
or endovascular approaches are superior in this 
situation, and each approach has its proponents. 
As grafts require relatively frequent interven- 
tions, many advocate using minimally invasive 
techniques first and reserving open surgery 
for those in whom the vascular interventionist 
fails. 



Infection 

Like any foreign material, infection is an 
ever-present hazard, especially as needles are 
frequently introduced into the graft. Septic 
episodes in dialysis patients are most likely to 
originate from the graft. Great care must be 
taken to use aseptic techniques when cannulat- 
ing grafts, as once organisms are seeded into 
a graft the only way to get rid of them is to 
replace the graft. Eradication of nasal car- 
riage of Staphylococcus aureus by use of topical 
mupirocin has been shown to reduce the rate of 
graft sepsis. 

Grafts are often placed in European prac- 
tice as a dialysis access of last resort, and there 
may be limited alternative sites if an infected 
graft is removed. Clearly if there is inflamma- 
tion along the length of the graft, then removal 
is the only option. Where localized sepsis exists, 
or a small area of the graft is exposed, local 



excision with skip grafting through uninvolved 
tissue may keep the graft functioning. The site 
of infection should be left open to drain freely, 
and the patient treated with parenteral anti- 
biotics until the inflammation has completely 
subsided. 



Vascular Steal 

This is most commonly seen in arm grafts in 
older or diabetic patients. Rates vary from 
between 1% to 19%. The approach is the same 
as steal in an autogenous AVF, with nonautoge- 
nous graft ligation or DRIL being the potential 
management approaches. 



Aneurysm Formation 

With repeated cannulation over small areas of 
a graft, there is a tendency for "aneurysm" for- 
mation (actually a pseudoaneurysm). This is a 
result of longitudinal splitting of the graft. It 
is not a problem if there is good skin coverage, 
but if enlarging rapidly or if inflamed, then 
early revision should be undertaken to prevent 
rupture and bleeding. The technique is similar 
to that used for infection, with the exception 
that excision of the aneurysmal portion of the 
graft is not always necessary in the absence of 
infection. 



Central Venous Catheters 

Infection 

Infection is the main problem from central 
venous (CV) catheters, and rigorous aseptic 
technique is required to manage dialysis. Newer, 
totally implantable catheters that are accessed 
through the skin may reduce the risk of sepsis. 
The teaching of correct hand and skin prepara- 
tion prior to needling remains an important 
part of the management of patients with CV 
catheters. 

Once infected, lines need to be changed. 
Ideally, the old line should be removed and the 
patient treated until he or she is well before 
inserting a new line. This is not always possible. 
Rotating the line to a new site is to be preferred 
in such situations, but if the situation is desper- 
ate, removing the old line and reinserting in the 



147 



VASCULAR ACCESS 




same location while treating with antibiotics 
may be successful. 



Fibrinous Encasement and Clotting 

The tips of catheters become covered with a 
"biofilm" of fibrin that may contain bacteria. 
Even if sterile, this film can build up until a large 
tube of fibrin encases the catheter tip. This can 
interfere with blood flow and obstruct dialysis. 
If the line is not infected, removal and replace- 
ment may still be necessary to reestablish dial- 
ysis. Passing guidewires down the catheter is 
not usually successful, but snaring the catheter 
tip radiologically and stripping the fibrin cuff 
can clear the offending plug. This technique can 
significantly prolong the life of central lines. 

Thrombosis is a common problem, especially 
if the line is not used frequently. Lines should 
be flushed with the correct volume of heparin 
(1000 U/mL) after each use to remove blood 
from the lumen and prevent thrombosis. Extra 
care must be paid to this in children, as it is easy 
to give excessive volumes of heparin and induce 
systemic anticoagulation and bleeding. 



The Avoidance of 
Complications: 
Access Surveillance 

Concerns about the frequent need for access 
revision have stimulated inquiry into tech- 
niques for the early detection of access com- 
plications before symptoms develop. Most 
techniques concentrate on intradialytic moni- 
toring of flow, pressure, dialysis efficacy, or a 
combination of these. The advantage of intradi- 
alytic monitoring is that it minimizes the need 
for extra hospital resources for these ill patients. 
Each technique has its proponents, and the tech- 
niques mentioned are not mutually exclusive. 

The DOQI guidelines state, "Access flow 
measured by ultrasound dilution, conductance 
dilution, thermal dilution, Doppler or other 
technique should be performed monthly. The 
assessment of flow should be performed during 
the first 1.5 hours of the treatment to eliminate 
error caused by decreases in cardiac output 
related to ultrafiltration. The mean value of 



three separate determinations performed at 
a single treatment should be considered the 
access flow. If access flow is less than 600 mL/ 
min, the patient should be referred for fistulo- 
gram. Access flow less than lOOOmL/min that 
has decreased by more than 25% over 4 months 
should be referred for fistulogram." 

This is a counsel of perfection and some 
centers would advocate duplex ultrasound prior 
to fistulography Where there is evidence of a 
stenosis of >50%, there is a significant sub- 
sequent thrombosis rate. Such fistula should 
undergo endovascular or surgical revision as 
appropriate. Juxta-anastomotic stenoses are 
often best dealt with by surgery, with endovas- 
cular dilatation being reserved for those in the 
body of the access. 



Desperate Access 



Dialysis is becoming more successful in pro- 
longing the lives of patients in renal failure. 
Unfortunately, this improvement in survival is 
not being matched by increases in transplanta- 
tion. As a result, many more people are depend- 
ent on long-term dialysis. As successive access 
procedures fail, the establishment of secure vas- 
cular access becomes increasingly difficult. 

Surgeons and interventionists have become 
ingenious at inserting grafts or lines into 
various veins, sometimes accidentally. Grafts 
can be placed in necklace fashion between one 
axilla and the contralateral one. Arteries can 
be divided and interposition grafts inserted. 
The superficial femoral vein has been utilized 
as a hemodialysis access conduit. Lines may 
be inserted directly into the inferior vena cava 
(IVC) through the back. Such procedures should 
be reserved for those in whom no alternative 
can be found after an extensive search using 
ultrasound, venography, and magnetic reso- 
nance angiography if necessary. It should be 
made clear to patients undergoing these proce- 
dures that the risks involved in establishing such 
access are high, and the consequence of failure 
may be fatal. 

Although successful arterial access can be 
obtained for numerous patients with reason- 
able durability, numerous controversies remain 
(Table 13.3). These controversies will be 
resolved with further clinical trials. 




148 



VASCULAR SURGERY 



Table 13.3. Contemporary controversies 

Role of endovascular vs. open management of the 

failing and failed autogenous AVF and 

nonautogenous AVF 
Construction of upper arm autogenous AVF prior to 

utilization of forearm nonautogenous AVF 
Role of invasive imaging (routine arteriograms and 

venograms) prior to vascular access constructions 
Adjuvant therapies to improve vascular access patency 

(anticoagulants, antiplatelet therapies, etc.) 
Unknown value of new nonautogenous AVF conduits 

(modified Dacron,homografts,etc.) 
Role of duplex surveillance in identifying the failing 

graft 



References 



Brescia MJ, Cimino JE, Appell K, Hurwich BJ, Scribner BH. 

(1966) J Am Soc Nephrol 10:193-9. 
Malovrh M. (2002) Am J Kidney Dis 39:1218-25. 
Sidawy AN, Gray R, Besarab A, et al. (2002) J Vase Surg 

35:603-10. 
Tordoir JH.Dammers R, van der Sande FM. (2004) Eur J Vase 

Endovasc Surg 27:1-5. 



14 

Outcome Measures in Vascular Surgery 

Christopher J. Kwolek and Alun H. Davies 




Background 



Initial efforts within the health care reform 
movement in the United States largely focused 
on reducing cost. This became the preeminent 
issue as health care expenditures continued to 
increase on an annual basis, reaching 14% of 
gross domestic product in 1994. Although some 
of these expenses can be attributed to improve- 
ments in diagnostic and therapeutic regimens, 
particularly in those areas highly influenced 
by new technologies, concern has also been 
expressed about the quality of the product being 
provided to patients. 

The publication of the Institute of Medicine's 
(IOM) Committee on Quality of Care report,"To 
Err Is Human: Building a Safer Health System," 
shifted the focus of attention from cost alone 
to medical errors within the U.S. health care 
system and the costs associated with these 
errors. This has led to an erosion of patient trust 
in the medical care system. It has been estimated 
that medical errors are responsible for between 
44,000 and 98,000 deaths annually, becoming 
the eighth leading cause of death in the U.S., 
ahead of other causes such as breast cancer 
and motor vehicle accidents. Over 7000 of these 
deaths were attributed to medication errors 
alone, with the total cost to society of these 
errors estimated to be $11637.6 billion a year. 
Much of the initial research describing the 
problem of medical errors was performed in the 
1990s and supported by the Agency for Health- 
care Research and Quality (AHRQ). 



The IOM report also led to the establishment 
of the Quality Interagency Coordination Task 
Force (QuIC), which was charged with coordi- 
nating the quality improvement activities in U.S. 
federal health care programs. These groups 
have been responsible for the establishment of 
standardized guidelines and protocols based on 
clinical trials, which may decrease variability in 
the patient care processes while improving care 
and reducing costs. The vast majority of the 
errors identified in these studies were systems 
related and not attributable to negligence or 
misconduct. In fact, up to 75% of the medical 
errors and 54% of the surgical errors were found 
to be preventable. 

The U.S. Veterans Administration (VA) hospi- 
tal system had already begun to look at these 
quality and outcomes issues through the estab- 
lishment of the National VA Surgical Quality 
Improvement Program (NSQIP), which has been 
responsible for assessing specific surgical out- 
comes throughout the U.S. VA hospital system, 
while using risk adjusted data to assess surgical 
morbidity and mortality and providing specific 
institutional feedback (Khuri et al., 1998). In 
fact, this program was so successful that in the 
late 1990s it was expanded to include three large 
academic medical centers at the University 
of Kentucky, the University of Michigan, and 
Emory University in Atlanta. Currently this 
program is being expanded to include multiple 
academic medical institutions across the U.S. 

In addition, large companies, which are pur- 
chasers of health care for their employees, have 



149 




150 



VASCULAR SURGERY 



Table 14.1. Current recommendations of the Leap Frog Group 

Computerized physician order entry: mandated use to 

minimize medication errors 
Intensive care unit (ICU) physician staffing: use of 

critical care certified physicians to provide exclusive 

care of ICU patients 
Evidence-based hospital referral: preferential referral of 

patients undergoing five high-risk surgical 

procedures to "high volume centers" 



Procedure 

Coronary artery bypass grafting 
Percutaneous coronary angioplasty 
Carotid endarterectomy 
Abdominal aortic aneurysm repair 
Esophagectomy 



Annual volume 
requirement 

500/year 

400/year 

100/year 

30/year 

6/year 



begun to take an interest in improving the 
quality and potentially decreasing the cost of 
health care as demonstrated by the activities of 
the Leap Frog Group (www.leapfroggroup.org). 
The stated purpose of this group of Fortune 500 
companies is to mobilize employer purchasing 
power to trigger breakthroughs in the safety 
and overall value of health care to American 
consumers. Using the results of these and other 
studies aimed at evaluating medical outcomes, 
the Leap Frog Group has already identified three 
areas to decrease errors and improve the quality 
in U.S. -based health care systems. Health care 
providers who adopt these recommendations 
will be rewarded with preferential use and 
other incentives by this group of health care 
purchasers. The current recommendations are 
listed in Table 14.1. It is estimated that imple- 
mentation of this program would save 60,000 
lives with a monetary savings of $3.8 billion a 
year. 

Although these recommendations are sup- 
ported by reviews of outcome studies in the 
current medical literature, certain limitations 
do exist. For example, volume-based outcome 
studies demonstrate that on average, high- 
volume centers have better patient outcomes for 
certain high-risk procedures such as repair of 
abdominal aortic aneurysm (AAA) or carotid 
endarterectomy (CEA). However, some high- 
volume centers may have average or poor out- 
comes, whereas some smaller centers with low 
volumes may have excellent outcomes. It maybe 
more important to measure the clinical process 



for a health care system to identify the reasons 
and processes that allow one facility to have 
excellent outcomes so that these can be utilized 
by other facilities to achieve the same excellent 
results. This concept was used by the Northern 
New England Cardiovascular Disease Study 
Group to improve the outcomes of patients 
undergoing coronary artery bypass grafting in 
northern New England (O'Connor et al., 1996). 

Outcomes Measures 

It has become important that we evaluate the 
relative value of different treatment regimens to 
include individual and societal costs and poten- 
tial risks and benefits. Individual patient per- 
spectives on quality have also become an 
important part of the evaluation process. 

There are several reasons for physicians to 
participate in this process. Ideally, our partici- 
pation should lead to an overall improvement in 
the quality of care that we deliver. In addition, 
participation in these programs will soon be 
necessary to qualify for reimbursement from 
many insurers and purchasers of health care 
such as the U.S. government and the Leap Frog 
Group. Finally, as physicians we have a societal 
responsibility to maximize the good and mini- 
mize adverse outcomes in health care, while 
best utilizing the limited resources that exist. 
If physicians choose not to participate in this 
process, then others will make these difficult 
decisions for us. 

These changes have led to the development 
of "extended outcome assessment" rather than 
just the traditional physician-oriented out- 
comes measures that we are used to. The con- 
cept of a value compass has been proposed 
to describe the interplay between traditional 
medical outcomes, patient satisfaction, func- 
tional assessment, and cost/utility outcomes 
(McDaniel et al, 2000). 

Clinical Status 

The most traditional group of medical out- 
comes measures utilized by vascular surgeons 
usually describes clinical status. These measures 
are physician oriented and include measure- 
ments of morbidity and mortality, graft patency, 
limb salvage, complications, and laboratory 
testing such as the ankle-brachial index (ABI) 



151 



OUTCOME MEASURES IN VASCULAR SURGERY 




and duplex ultrasound results. Many of these 
outcomes are important measures of technical 
success and are easily evaluable by statistical 
methods such as life table analysis. However, 
even the long-term results within this category 
such as patient survival after aneurysm repair, 
stroke-free survival after carotid endarterec- 
tomy, or amputation-free survival after lower 
extremity bypass grafting do not necessarily 
correlate with patients' overall well-being, day- 
to-day functioning, or their perceived quality of 
life. Physicians are also comfortable with these 
outcomes since the Society for Vascular Surgery 
(SVS) and the International Society for Cardio- 
vascular Surgery (ISCVS) have published guide- 
lines for measuring these types of outcomes for 
commonly performed arterial and venous vas- 
cular procedures. 

Patient Satisfaction 

Patient-oriented outcomes tend to be less fre- 
quently measured and not as well defined. None 
of these outcomes are included in the SVS- 
ISCVS reporting standards. Patient satisfaction 
with the health care process is one area being 
more closely evaluated. Although customer sat- 
isfaction surveys have long been used in the 
world of business, they are now being applied 
to the area of health care. Two areas being fre- 
quently evaluated include patient satisfaction 
with the physician-patient relationship and sat- 
isfaction with the health care delivery process. 
This may include such issues as timeliness and 
access to care, provider and staff communica- 
tion, the physical environment where care is 
delivered, and courtesy and respect shown to 
the patient. Managed care plans in the U.S. are 
now required to assess themselves using stan- 
dardized patient satisfaction surveys such as the 
Health Plan Employer Data and Information Set 
(HEDIS), which was developed by the National 
Committee for Quality Assurance (NCQA). 
Many employers and purchasers of health care 
are now utilizing this information when decid- 
ing on which health care providers to include in 
their panel of providers. 

Functional Status 

Functional status is another of the patient- 
oriented outcomes that is being more com- 
monly described in the vascular literature. These 



health assessment instruments are designed to 
quantify how illnesses and treatments affect 
different aspects of patient functioning in every- 
day life. This will allow physicians to evaluate 
not only the presence or absence of a leg or the 
patency of a graft, but how patients are func- 
tioning after a specific intervention. These meas- 
ures are also helpful when evaluating patients' 
expectations before and after specific interven- 
tions. A list of commonly used measures of func- 
tional status is included in Table 14.2. There 
are two types of assessment tools. The specific 
instrument focuses on a specific disease or client 
group, and changes in this are more likely to 
detect subtle changes in quality of life, whereas 
generic tools give a broader summary of health- 
related quality of life, hence enabling compar- 
isons with patients suffering from other disease 
processes (Table 14.3). 

The most widely used generic functional 
health assessment instrument in the United 
States is the Medical Outcomes Short Form 36 
(SF-36). This survey evaluates patient function 
in physical and social roles, limitations due to 
health or emotional problems, patient percep- 
tions of general health, mental health, bodily 
pain, and vitality. In addition, a question con- 
cerning change in health status compared to 1 
year previously is included in the survey. This 
survey combines aspects of a Quality of Well- 
Being Scale and a Functional Status Question- 
naire. The reason that this survey has become so 
useful is that large numbers of patients have 
been evaluated using this instrument, thus 
allowing researchers to compare results to the 
general population or a patient subgroup with 
specific characteristics. These results may be 



Table 14.2. Examples of measures of functional status 

General status: quality of life 

SF-36 

Euro-QOL 

Nottingham Health Profile 

Functional Status Questionnaire 

Quality of Well-Being Scale 

Sickness Impact Profile 
Disease/symptom specific: quality of life 

Walking Impairment Questionnaire: for patients 
with lower extremity arterial occlusive disease 

Charing Cross Claudication Questionnaire 

Charing Cross Venous Ulcer Questionnaire 

VascQuol 





> 



152 



VASCULAR SURGERY 



Table 14.3. Advantages and disadvantages of quality of life instruments 



Instrument 

Generic 



Specific 



Advantages 

Single instrument 

Detects different aspects of health 

Enables comparison between different conditions 

Can be used in cost analysis 

Focus on primary are of interest 

More relevant to clinicians and clinical condition 

May be more responsive 



Disadvantages 

May not focus adequately on main problem 
May lack responsiveness 

Does not take account of values attributed to levels 
of quality of life 

Not comprehensive 

May miss side effects 

Cannot compare across conditions 



helpful in predicting long-term functional out- 
comes in patients. Thus patients scoring above 
a certain level in the area of bodily pain may 
have a decreased chance of successfully return- 
ing to work, and patients with decreased overall 
scores in the areas of physical and social func- 
tion and health may have an increased 5-year 
mortality. However, the abbreviated form of the 
SF-36, the SF-12, is growing in popularity. 

In addition to general evaluations of func- 
tional status, disease-specific instruments have 
also been developed. One of the most widely 
used surveys for the evaluation of patients with 
lower extremity arterial occlusive disease is 
the Walking Impairment Questionnaire (WIQ); 
however, there are newer and more specific 
questionnaires becoming available (Chong et 
al., 2002). Because peripheral vascular disease 
has a detrimental effect on quality of life even 
in patients without the most severe forms of 
limb-threatening ischemia, these instruments 
may be very useful in evaluating the benefit to 
patients undergoing treatment for claudication. 
These surveys may be even more important for 
evaluating patients undergoing prophylactic 
interventions for the management of asympto- 
matic disease. 

Some authors have recommended that a com- 
bination of two different surveys such as the 
SF-36 and the EuroQol be used to evaluate the 
quality of life outcomes in patients with and 
without ischemic complications who are under- 
going infrainguinal bypass grafting (Tangelder 
et al., 1999). Tangelder et al. found that the com- 
bination of the SF-36 and the EuroQol provided 
useful information concerning the patients' 
quality of life after lower extremity bypass graft- 
ing. Interestingly, patients' functional outcomes 
were similar for those with asymptomatic 
graft occlusions and patent grafts, although the 



lowest outcomes were found in patients who 
underwent amputation after failed attempts 
at secondary revascularization. These results 
confirm clinical findings that are often well 
known in clinical practice but that are not shown 
by primary or secondary patency rates or limb 
salvage. 

Additional concerns have been expressed 
about the potential for patient bias with self- 
reporting of health status. However, patient 
reports of functional health status appear to 
have good face value validity. Thus patients who 
suffer from severe strokes report worse func- 
tioning in the areas of physical and general 
health than patients with mild strokes. Similarly, 
patients with venous ulcers report impaired 
social interaction, domestic activity, and emo- 
tional status with improved functioning after 
healing of their venous ulcers (Smith et al., 
2000). Also, the benefits of varicose vein surgery 
can be justified in terms of improvements in 
quality of life scores. 



Cost/Utility 

Cost outcomes are another important area of 
recent interest. Increasing pressures are being 
exerted by the government and insurers to 
optimize the quality of the health care while 
minimizing expenditures. True costs to the indi- 
vidual and society must be calculated both for 
the acute illness and over the long term. 

Practice guidelines can often be established 
based on the results from existing controlled 
clinical trials. These guidelines can then be used 
on a regional or national basis to evaluate physi- 
cians and health care organizations. Using the 
same processes of continuous quality improve- 
ment (CQI) found in major industrial manufac- 



153 



OUTCOME MEASURES IN VASCULAR SURGERY 




turing plants, these guidelines should reduce 
variation in the processes of care, thus improv- 
ing quality while decreasing cost. These guide- 
lines are designed to evaluate processes, with 
the ultimate philosophy of continuously impro- 
ving the process to ensure the best possible 
outcome. 

This process is in contradistinction to the 
more traditional concept of quality analysis 
(QA), where minimum standards or thresholds 
are set to ensure a good outcome and only out- 
liers are evaluated rigorously. The pitfall of this 
approach is that it encourages organizations to 
be just "good enough" and meet the minimum 
standards set rather than aim for being the best 
possible. In addition, it singles out only those 
groups or individuals doing a poor job rather 
than rewarding groups that are doing a good job 
and trying to reproduce those results in other 
areas. 

The value that patients and society place 
on certain outcomes and levels of functioning 
can also play a role in evaluating the cost- 
effectiveness of certain procedures. The cost- 
effectiveness of a certain intervention can be 
expressed as the net benefit to a population by 
using statistical probabilities of certain out- 
comes occurring along with specific costs asso- 
ciated with various outcomes as documented 
from the medical literature. One example of 
this is the use of the Markov decision model to 
evaluate the cost-effectiveness of performing 
carotid endarterectomy in asymptomatic pa- 
tients with >60% stenosis (Cronenwett et al., 
1997). The measurement units in this study are 
in quality-adjusted life years (QALYs) defined as 
the fraction of a year in perfect health that the 
patient believes to be equivalent in value to a 
year in the health state in question. 

In this study, Cronenwett et al. demonstrated 
that from a societal standpoint, carotid endar- 
terectomy appeared to be cost-effective for the 
young asymptomatic patient with standard 
risk factors in the hands of a surgeon with a 
2.3% 30-day perioperative stroke and death rate. 
However, the procedure was not found to be 
cost-effective for patients older than 79 years of 
age, those with a high perioperative stroke risk, 
or those with a particularly low stroke risk with 
medical management. In this study it is impor- 
tant to note that cost-effectiveness is compared 
to cost/QALY for other medical interventions, 
and that a cost of over $100,000/additional 



QALY was defined as not cost-effective. Inter- 
estingly, one could extend the evaluation even 
further to include the individual's risk aversion 
and risk taking behavior when defining the 
cost-effectiveness for that person. This is es- 
sentially what occurs every time we obtain 
informed consent from a person who is about to 
undergo a high-risk procedure. 



Conclusion 

The reasons for physicians to utilize extended 
outcomes assessment include the following: 
(1) to achieve a better understanding of the 
effectiveness of our interventions; (2) to pro- 
vide health care consumers, both patients and 
insurers, with information that will allow them 
to make better informed decisions; and (3) to 
develop public health standards that will allow 
us provide the most cost-effective care 
(McDaniel et al, 2000). 

In addition, the Accreditation Council for 
Graduate Medical Education (ACGME) in the 
United States has mandated that all residency 
training programs evaluate as part of their core 
competency requirements six areas, all of which 
involve some component involving outcomes 
measures: (1) practice-based learning, (2) 
systems-based practice, (3) medical knowledge, 
(4) patient care, (5) interpersonal and com- 
munication skills, and (6) professionalism 
(www.acgme.org/outcomes). 

Finally, the incorporation of outcomes meas- 
ures are now being studied by the task force on 
competence of the American Board of Medical 
Specialties to be used in the process of recer- 
tification. In fact, recommendations have already 
been published stating that outcomes measures 
should be included as part of the requirement 
for recertification in vascular surgery by the 
American Board of Surgery (Hertzer, 2001). 



References 



Chong PF, Garratt AM, Golledge J, Greenhalgh RM, Davies 
AH. (2002) J Vase Surg 36:764-71; discussion 863-4. 

Cronenwett JL, Birkmeyer JD, Nackman GB, et al. (1997) 
J Vase Surg 25:298-309; discussion 310-1. 

Hertzer NR. (2001) J Vase Surg 34:371-3. 

Khuri SF, Daley J, Henderson W, et al. (1998) Ann Surg 
228:491-507. 




> 



154 



VASCULAR SURGERY 



McDaniel MD, Nehler MR, Santilli SM, et al. (2000) J Vase 

Surg 32:1239-50. 
O'Connor GT, Plume SK, Olmstead EM, et al. (1996) JAMA 

275:841-6. 



Smith JJ, Guest MG, Greenhalgh RM, Davies AH. (2000) 

J Vase Surg 31:642-9. 
Tangelder MJ, McDonnel J, Van Busschbach JJ, et al. (1999) 

J Vase Surg 29:913-9. 



15 

Carotid Artery Disease 

A. Ross Naylor, Peter H. Lin, and Elliot L. Chaikof 




Epidemiology 

Stroke 

A stroke is defined as a focal (occasionally 
global) loss of cerebral function lasting for more 
than 24 hours, and which after investigation is 
found to have a vascular cause. Stroke is respon- 
sible for 4.5 million deaths worldwide, with the 
majority occurring in nonindustrialized coun- 
tries. In the United States, stroke is the third 
most common cause of death, with approxi- 
mately 160,000 Americans dying from the 
disease each year. Stroke management con- 
sumes $45 billion annually, including indirect 
costs and is responsible for more than one 
million hospital discharges per annum. 

The demographics are remarkably similar in 
the United Kingdom, where stroke is also the 
third leading cause of death (12% of deaths 
overall) and 58,000 Britons die each year. Five 
percent of the National Health Service budget is 
used in stroke care (excluding indirect costs), 
and stroke patients occupy 20% of acute hospi- 
tal beds and 25% of rehabilitation beds. 

The incidence of stroke is 2 in 1000 per 
annum, but significantly increases with con- 
current risk factors of age, sex, and ethnic 
background. Overall, the 20-year risk for a 45- 
year-old man is 3%, but this increases to 25% for 
a 40-year risk (Bonita, 1992). The annual inci- 
dence of stroke doubles for each decade over the 



age of 55 years. For those aged to 44 years, the 
annual incidence of stroke is 0.09 in 1000, 
increasing to 2.9 in 1000 for those aged 55 to 64 
and 14.3 in 1000 in those aged 75 to 84. The 
largest incidence is observed in populations 
aged >85 years, where the incidence is almost 
20 in 1000. In both the United States and the 
United Kingdom, the incidence of stroke is twice 
as high in blacks as in white. This ethnic dif- 
ference is thought to be due to the increased 
incidence in blacks of risk factors such as hyper- 
tension, diabetes, smoking, obesity, and sickle 
cell disease. 



Transient Ischemic Attack 

A transient ischemic attack (TIA) has the same 
definition as stroke but it lasts for less than 
24 hours. The 24-hour threshold is somewhat 
arbitrary, as up to 28% of TIA patients have an 
infarct on computed tomography (CT) scan, of 
which 36% are bilateral. In the U.S., the preva- 
lence of TIA in men aged 65 to 69 years is 2.7%, 
increasing to 3.6% for men aged 75 to 79 years. 
The respective prevalence figures for women are 
1.6% and 4.1%. In the U.K., the overall incidence 
of TIA is 0.4 in 1000, but this varies with age. 
The incidence is 0.25 in 1000 for those aged 45 
to 54 years, increasing to 1.61 in 1000 in the 65- 
to 74-year age group and 2.57 in 1000 in those 
aged 75 to 84 years. 



155 




156 



VASCULAR SURGERY 



Etiology of Ischemic Stroke 

Approximately 80% of strokes are ischemic and 
20% hemorrhagic. This section deals primarily 
with the etiology of ischemic carotid territory 
stroke. 

Thromboembolism 

Approximately 50% of all ischemic carotid ter- 
ritory strokes follow a carotid thrombosis or 
embolism from a carotid stenosis (Fig. 15.1) 
into territories supplied by either the middle 
cerebral artery (MCA) or the anterior cerebral 
artery (ACA). The sequelae of carotid thrombo- 
sis depend on a number of factors, including the 
status of the circle of Willis (potential for col- 
lateralization),the chronicity of the thrombosis, 




Figure 15.1. Selective intraarterial digital subtraction angio- 
gram of left carotid bifurcation showing diffuse disease of the 
upper third of the common carotid artery,a stenosis at the origin 
of the external carotid artery, and a complex severe stenosis at 
the origin of the internal carotid artery with deep ulceration. 



and the extent of the thrombosis. Once the 
internal carotid artery (ICA) has thrombosed, 
the column of thrombus usually propagates 
distally to the ophthalmic artery. However, the 
thrombus may occasionally extend beyond the 
ophthalmic artery and propagate into the circle 
of Willis. 

A very small proportion of strokes (<4%) are 
secondary to isolated cerebral hypoperfusion. 
Patients susceptible to this type of stroke include 
those with critical ICA stenoses, poor collateral- 
ization via the circle of Willis, and a secondary 
trigger such as hypotension following an acute 
cardiac event. 

Rarely, carotid thromboembolism may cause 
posterior (vertebrobasilar) territory strokes. 
This arises because, embryologically, the ICA 
provides the blood supply to the developing 
posterior cerebral hemispheres and brainstem 
via the posterior communicating and posterior 
cerebral arteries. In a very small proportion of 
the population, this anatomical arrangement 
persists into adult life. 

Intracranial Small-Vessel Disease 

Occlusion of the penetrating end arteries 
(lenticulostriate, thalamoperforate) in the 
deep structures of the brain causes discrete 
wedge-shaped infarctions of brain tissue. These 
ischemic lesions, termed lacunar infarcts, are 
responsible for up to 25% of ischemic carotid 
territory strokes. Conditions predisposing 
toward lacunar infarction include hypertension 
and diabetes. There is still considerable debate 
as to whether embolization from a carotid 
stenosis can cause lacunar infarction. 

Cardiac Embolism 

Cardiac embolism accounts for 15% of ische- 
mic carotid territory strokes. The emboli con- 
sist of fibrin, cholesterol, calcified debris, and 
atheroma depending on the underlying pathol- 
ogy. Sources include valvular heart disease, 
prosthetic valves, atrial myxoma, ventricular 
aneurysm thrombus, cardiomyopathy, and in- 
fective endocarditis. However, the commonest 
causes are atrial fibrillation and embolization 
of mural thrombus overlying a dyskinetic seg- 
ment of myocardium following a myocardial 
infarction (MI). 



157 



CAROTID ARTERY DISEASE 




Hematological Causes 

A variety of pathologies predisposing toward a 
hypercoagulable state are responsible for about 
5% of ischemic strokes. These include poly- 
cythemia, sickle cell disease, leukemia, throm- 
bocythemia, malignancies, functional protein S 
deficiency, lupus anticoagulant (antiphospho- 
lipid antibody syndrome), antithrombin III 
deficiency, and the paraproteinemias. 

Miscellaneous Causes 

Miscellaneous causes account for the remain- 
ing 5% of ischemic carotid territory strokes. 
These include migraine, oral contraceptive use, 
trauma, dissection, giant cell arteritis, Takayasu 
arteritis, systemic lupus erythematosus (SLE), 
polyarteritis nodosa, amyloid angiopathy, co- 
caine abuse, fibromuscular dysplasia, and radi- 
ation arteritis. 



Pathology of Carotid 
Artery Disease 

Atherosclerosis 

Atherosclerosis is the commonest pathology 
affecting the carotid artery. The typical plaque 
encountered at carotid endarterectomy (Fig. 
15.2) is the culmination of a sequence of patho- 
physiological processes starting with endothe- 
lial dysfunction/damage. The tendency for 
atherosclerotic plaque to form at the carotid 
bifurcation is related to a number of factors, 
including geometry, velocity profile, and shear 
stress. 

It was originally proposed that local turbu- 
lence predisposed to high wall shear stress and 
endothelial injury. This hypothesis, however, 
was refuted by Zarins et al. (1983), who showed 
that plaque formation was increased within 
areas of low flow velocity and low shear stress 
and decreased in areas of high flow velocity and 
elevated shear stress. Postmortem specimens 
showed that atherosclerosis was particularly 
pronounced along the outer (lateral) aspect of 
the proximal ICA and carotid bulb. This zone 
corresponds to areas of low velocity and low 
shear stress. The medial or inner aspect of the 
carotid bulb (associated with high blood flow 




Figure 15.2. Atheromatous plaque excised at carotid endar- 
terectomy. Note the deep area of ulceration in the plaque 
surface. 



velocity and high shear stress in the flow model) 
were relatively free of plaque formation. 

The smooth muscle cell has an important 
role in the initial stages of plaque develop- 
ment. Smooth muscle cells migrate through 
the intima, proliferate within the media, and 
promote accumulation of cholesterol and other 
lipid molecules within the evolving lesion. 
Thereafter, the macrophage becomes a source of 
growth factor production that stimulates further 
smooth muscle cell proliferation and extracellu- 
lar matrix production. Smooth muscle cells and 
macrophages initiate a secondary inflammatory 
cell reaction and are capable of ingesting lipid 
and of being transformed into vacuolated foam 
cells that are characteristic of atherosclerotic 
lesions. 

Besides these cellular components, the major- 
ity of carotid plaques have a necrotic core con- 
sisting of loose cellular debris and cholesterol 
crystals. The necrotic core is separated from the 
carotid lumen by a fibrous cap, which is com- 
posed of a rim of variable thickness comprising 
cellular components and extracellular matrix. 
The structural integrity of the fibrous cap is 
crucial to the final stage of plaque disruption 
and its clinical and pathological sequelae. It is 
now generally accepted that acute changes 
within the plaque, notably Assuring/splitting 
of the fibrous cap (Fig. 15.2), with exposure of 
the deeper lipid contents predisposes toward 
thrombosis with or without secondary emboli- 
zation. It is not known what actually pre- 
disposes toward acute plaque disruption, but 
recent studies suggest that excess matrix metal- 
loproteinase activity or cytokine expression 




> 



158 



VASCULAR SURGERY 



within the plaque may be associated with this 
process (Loftus et al., 2002). 

Another feature characteristic of advanced 
atherosclerotic plaques is intraplaque hemor- 
rhage that can occur in the absence of a dis- 
rupted fibrous cap. Symptomatic carotid disease 
is associated with increased neovascularization 
within the atherosclerotic plaque and fibrous 
cap. These vessels are larger and more irregular 
and may contribute to plaque instability and the 
onset of thromboembolic sequelae. 

Fibromuscular Dysplasia 

Fibromuscular dysplasia (FMD) is the com- 
monest nonatherosclerotic disease to affect 
the ICA. Approximately one quarter of patients 
with carotid FMD have associated intracranial 
aneurysms, and up to two thirds of these 
patients will have bilateral carotid fibromuscu- 
lar dysplasia. 

Fibromuscular dysplasia can be divided into 
three pathological subtypes. Medial fibroplasia 
is the most common (>85% of cases) and is 
usually found in long segment arteries with few 
side branches. It is characterized by stenoses 
alternating with intervening fusiform dilata- 
tions that resemble a string of beads, particu- 
larly in the upper ICA. Pathologically, smooth 
muscle cells in the outer media are replaced 
by compact fibrous connective tissue, whereas 
the inner media contains excess collagen and 
ground substance in disorganized smooth 
muscle cells. Because of its prevalence in 
females, a possible role for estrogen and prog- 
esterone has been postulated. Others have sug- 
gested that the absence of vasa vasorum in long 
nonbranching arteries such as the ICA or renal 
arteries may predispose to mural ischemia 
that leads to the development of fibromuscular 
dysplasia. 

Intimal fibroplasia accounts for less than 10% 
of cases of FMD and affects men and women 
equally. It typically appears as a focal narrowing 
in older patients and long segmental stenoses in 
younger patients. The lesion is confined to the 
intimal layer while the medial and adventitial 
structures are always normal. Its pathophysiol- 
ogy is due to irregularly aligned sub endothelial 
mesenchymal cells within a loose matrix of con- 
nective tissue. Perimedial dysplasia is char- 
acterized by accumulation of elastic tissue 
between the media and the adventitia. This 



subtype predominantly affects the ICA and 
renal arteries and may be associated with 
secondary aneurysm formation. 

Coils and Kinks of the Extracranial 
Carotid Arteries 

Redundancy of the extracranial carotid artery 
is thought to be due to abnormalities in devel- 
opment. Occasionally, the ICA may undergo 
a complete 360-degree rotation. The ICA is 
derived embryologically from the third aortic 
arch and the dorsal aortic root. In its early 
stages, a normally occurring redundancy is 
straightened as the heart and great vessels 
descend into the mediastinum. Incomplete 
descent of the heart and great vessels may result 
in the development of complex coils and kinks. 
These are bilateral in approximately 50% of 
affected patients. 

Elongation of the ICA, which can also result 
in kinking of the ICA, is usually due to degen- 
erative changes associated with increasing age 
and atherosclerosis. The loss of elasticity of the 
arterial wall due to the aging process (coupled 
with hemodynamic shear stresses) predisposes 
toward kinks of the elongated carotid artery 
between the proximal and distal fixed points of 
the skull base and thoracic inlet. 

Carotid Artery Aneurysms 

As with aneurysms anywhere else in the body, 
carotid aneurysms may be either true or false. 
In the past,most true aneurysms (Fig. 15.3) have 
traditionally been classified as part of the 
atherosclerotic process, but emerging evidence 
suggests that aneurysmal disease may be yet 
another manifestation of abnormalities of 
matrix metalloproteinase enzyme expression 
and production, unless associated with other 
distinct pathologies, for example, arteritis (giant 
cell, Takayasu) or FMD. False aneurysms may 
arise as a consequence of iatrogenic injury, blunt 
trauma, spontaneous dissection, or infection 
(e.g., prosthetic patch infection after carotid 
endarterectomy). 

Carotid Dissection 

Acute carotid dissection can complicate athero- 
sclerosis, FMD, cystic medial necrosis, or blunt 



159 



CAROTID ARTERY DISEASE 





Figure 15.3. Intraarterial digital subtraction angiogram of the 
aortic arch and great vessels. There is a large aneurysm just 
beyond the left carotid bifurcation. 



trauma, or be associated with a collagen vascular 
disorder such as Ehlers-Danlos syndrome type 
IV or Marfan syndrome. Angiographic studies 
suggest that the most likely mechanism is an 
intimal tear followed by an acute intimal dissec- 
tion, which produces luminal occlusion due to 
secondary thrombosis within the false lumen. 
This appears as a flame-shaped occlusion 2 to 3 
cm beyond the bifurcation (Fig. 15.4). Autopsy 
studies typically reveal a sharply demarcated 
transition between the normal carotid artery 
and the dissected carotid segment. The ICA is 
commonly affected, with the dissection plane 
typically occurring in the outer medial layer. 
The classical triad of clinical signs/symptoms 
following carotid dissection include (1) pain in 
the face and neck (20%), (2) a partial Horner 
syndrome (50%), and (3) retinal or cerebral 
ischemia (50% to 90%). About one in ten will 
suffer cranial nerve palsy. 

Takayasu's Arteritis 

Takayasu's arteritis (TA) is a nonspecific arteri- 
tis affecting the thoracic and abdominal aorta 
and their major branches. Although this dis- 
ease is uncommon in Western countries, it is 



more prevalent in Asia and usually affects young 
females. Its pathogenesis relates to an inflam- 
matory process involving all three layers of the 
arterial wall with proliferation of connective 
tissue and degeneration of the elastic fibers. 
Granulomatous lesions may also develop, and 
the condition may also be associated with fusi- 
form or saccular aneurysms. 

Takayasu's arteritis is classified according 
to its mode of involvement. Type I involves 
branches of the aortic arch; type Ha involves the 
ascending aorta, aortic arch, and branches; type 
lib involves the vessels involved in Ha plus the 
descending aorta; type III involves the descend- 
ing thoracic aorta and abdominal aorta (with 
or without renal arteries); type IV involves the 
abdominal aorta (with or without renal arter- 
ies) and type V involves a combination of type 
lib and IV 




Figure 15.4. Selective intraarterial digital subtraction angio- 
gram in a patient with acute carotid dissection. The internal 
carotid artery is almost completely occluded 2 to 3cm beyond 
the origin of the internal carotid artery.There is virtually no flow 
visible up the true lumen. 




> 



160 



VASCULAR SURGERY 



Giant Cell Arteritis 

Giant cell arteritis predominantly affects elderly 
women. Although it commonly involves the 
temporal arteries, it can involve other major 
vessels including the carotid artery and upper 
extremity vasculature. There is usually ten- 
derness over the inflamed artery. Laboratory 
evaluation generally reveals elevation of C- 
reactive protein and the erythrocyte sedimenta- 
tion rate. Biopsy of an affected artery shows 
characteristic giant cell granulomata with abun- 
dant T lymphocytes. However, a negative biopsy 
does not exclude the diagnosis. Management 
of giant cell arteritis usually includes the use of 
high-dose corticosteroids as well as other forms 
of immunosuppression including cyclophos- 
phamide or azathioprine. Steroids can usually 
be tailed off after 1 year of treatment. 



Radiation Arteritis 

The principal effects of radiation on arteries 
include immediate arterial spasm and endo- 
thelial denudation, intimal disruption, sub- 
intimal edema, and degeneration of collagen and 
smooth muscle. These acute changes predispose 
toward an increase in vessel wall permeability to 
circulating lipids, which produce a plaque char- 
acterized by fibrosis, fatty infiltration, and elastic 
tissue destruction. Hyperlipidemia and hyper- 
cholesterolemia appear to predispose patients 
who have received radiation therapy to develop 
accelerated atherosclerotic lesions. The sensitiv- 
ity of elastic tissue to radiation may account for 
the mechanism of structural weakening and 
eventual rupture in elastic arteries. 

As a result of the increased use of external 
radiation to treat neck malignancies, a rise in 
radiation-induced atherosclerotic disease in 
association with symptomatic carotid stenosis 
has been noted. Radiation-induced carotid 
lesions can present either in a segmental or 
diffuse manner. The affected carotid segments 
typically lie within the field of radiation treat- 
ment. One or both common carotid arteries may 
be involved, whereas the carotid bifurcation is 
often spared. The severity of carotid injury is 
related to radiation dose. Smaller doses cause 
less cellular damage, whereas larger doses may 
even lead to arterial wall necrosis. 



Carotid Body Tumor 

Carotid body tumors (CBTs) originate from the 
chemoreceptor cells located at the carotid bifur- 
cation. Because cells of the carotid body typi- 
cally detect change in the P0 2 , PC0 2 , and pH, 
CBTs have been reported to be more prevalent 
in individuals who live at high altitudes, which 
suggests that chronic hypoxia may be an etio- 
logical factor. A CBT typically presents as a 
palpable, painless mass over the carotid bifur- 
cation region in the neck. Cranial nerve palsy 
may occur in up to 25% of patients, particularly 
involving the vagus and hypoglossal nerves. 
The differential diagnosis includes cervical 
lymphadenopathy, carotid artery aneurysm, 
branchial cleft cyst, laryngeal carcinoma, and 
metastatic tumor. 

The treatment of choice of CBT is surgical 
excision. Because these tumors are highly vas- 
cularized, preoperative tumor embolization has 
been advocated by some surgeons to minimize 
operative blood loss when dealing with tumors 
>3cm in diameter, but there is no consensus on 
this strategy. An important surgical principle in 
CBT resection is to maintain the plane of dis- 
section within the subadventitial space, which 
enables complete tumor excision without inter- 
rupting carotid artery integrity. 



Carotid Trauma 

Blunt trauma to the neck can cause carotid 
artery injury either by forceful compression 
or extension of the artery. Common scenar- 
ios include motor vehicle or motorcycle 
accidents, pedestrian injuries, hanging, or 
strangulation. 

Patients with carotid trauma may present 
with focal physical findings such as neck hema- 
toma, pulsatile cervical mass, carotid bruit, or 
localized bleeding. More generalized physical 
findings include loss of consciousness and lat- 
eralizing neurological deficits. Arteriography 
remains the diagnostic test of choice for carotid 
trauma, since it has the highest sensitivity 
and specificity compared to all other imaging 
modalities. Treatment of all blunt carotid artery 
injuries usually involves anticoagulation. Anti- 
platelet therapy should be considered if sys- 
temic anticoagulation is contraindicated. 



161 



CAROTID ARTERY DISEASE 




Clinical Presentation 
of Carotid Disease 

Syndromes 

Transient Ischemic Attack 

A transient ischemic attack is a focal loss of neu- 
rological function (lasting <24 hours) that has a 
vascular cause upon investigation. It is conven- 
tional to describe a TIA as being carotid or ver- 
tebrobasilar (see below). 



Nonhemispheric Symptoms 

These are a group of nonfocal symptoms that, 
on their own, are not indicative of true carotid 
or vertebrobasilar events. Classical nonhemi- 
spheric symptoms include isolated dizziness, 
isolated vertigo, isolated syncope (drop attacks, 
blackouts), isolated double vision (diplopia), 
and presyncope (faintness). In the absence of 
any corroborative carotid or vertebrobasilar 
symptoms, they cannot be ascribed to extracra- 
nial carotid or vertebral disease alone. 



Completed Stroke 

A completed stroke is a focal (occasionally 
global) loss of neurological function, lasting 
for more than 24 hours, that is found to have a 
vascular cause upon investigation. The term 
completed refers to the fact that the severity of 
the neurological deficit has now reached its 
maximum. 



Stroke in Evolution 

A stroke in evolution is a progressive worsening 
of the neurological deficit, either linearly over a 
24-hour period or interspersed with transient 
periods of stabilization and/or partial clinical 
improvement. 



Crescendo Transient Ischemic Attacks 

This condition has never been properly defined. 
Traditionally, it refers to a syndrome compris- 
ing repeated TIAs within a short period of 
time with complete neurological recovery in 
between. At a minimum, the term should prob- 
ably be reserved for those with daily events. 



Hemodynamic Transient Ischemic Attacks 

These attacks are focal cerebral events (hemi- 
sensory or retinal) that are aggravated by exer- 
cise or hemodynamic stress and typically occur 
after short bursts of physical activity, post- 
prandially or after getting out of a hot bath. It 
is implied that they are due to severe extra- 
cranial disease and poor intracranial collateral 
recruitment. 



Clinical Features 

Ocular Symptoms 

Ocular symptoms associated with extracranial 
carotid and vertebrobasilar disease include 
amaurosis fugax (transient monocular blind- 
ness), Hollenhorst plaques, retinal/optic nerve 
ischemia, the ocular ischemia syndrome, and 
visual field deficits secondary to cortical infarc- 
tion and ischemia of the optic tracts. 

The blood supply to the retina originates 
from two sources. The short posterior ciliary 
arteries supply the outer two layers, together 
with the optic disk and optic nerve. The central 
retinal artery supplies the inner layers of the 
retina. Amaurosis fugax is a temporary loss of 
vision in one eye (likened to a shutter coming 
down), but which can also refer to graying of the 
vision. The blindness usually lasts for a few 
minutes and then resolves. Most (>90%) are 
due to embolic occlusion of the main artery or 
its upper/lower divisions. It remains unclear, 
however, why some patients always have 
repeated episodes of amaurosis and never 
hemispheric signs. 

Monocular blindness progressing over a 20- 
minute period suggests a migrainous etiology. 
Hemodynamic amaurosis is rare but can be pre- 
cipitated by heavy exercise or arising from a hot 
bath. In this situation, where there is a severe 
carotid stenosis and poor collateralization, the 
visual loss tends to start at the periphery and 
move toward the center. Occasionally, a patient 
recalls no visual symptoms, but the optician 
notes a yellowish plaque within the retinal 
vessels (the Hollenhorst plaque). This is fre- 
quently derived from cholesterol embolization 
from the carotid bifurcation and it warrants 
further investigation (Fig. 15.5). 




> 



162 



VASCULAR SURGERY 




Figure 15.5. Funduscopic examination. Note the two choles- 
terol emboli within the retinal vessels. 



Monocular visual loss due to central retinal 
artery occlusion, persisting for more than 24 
hours is analogous to cerebral infarction, with 
the occlusive process usually occurring in the 
intraneural part of the central retinal artery. In 
addition to an embolic etiology, it is also impor- 
tant to consider microatheroma, and arteritis in 
occasional patients with other atypical symp- 
toms. Bilateral visual loss (cortical blindness) 
is due to infarction of the visual cortex in the 
occipital lobe and therefore is never a carotid 
territory event. 

The optic nerve head can be rendered 
ischemic by thromboembolic phenomena, 
although it is rarely due to extracranial carotid 
artery disease. Optic nerve ischemia is usually 
due to intrinsic disease of the short posterior 
ciliary arteries and can present as an acute loss 
of either central or peripheral vision in one 
eye. If the upper or lower visual fields are lost, 
this suggests occlusion of the upper and lower 
branch divisions of the ciliary artery. 

The ocular-ischemia syndrome is a relatively 
rare condition where there is global under- 
perfusion of the eye, deteriorating monocular 
vision, and excessive proliferation of the con- 
junctival vessels. It is indicative of extremely 
poor eye collateralization. 

Homonymous hemianopia can occur as a 
consequence of ischemia at varying points in 



the optic tracts. Occipital lesions cause a con- 
gruous homonymous hemianopia (visual field 
defects exactly overlap in each eye). Homony- 
mous hemianopia involving the lower visual 
fields is usually due to a parietal infarction and 
follows ischemia of the upper fibers of the visual 
pathway that traverse the parietal lobe. Con- 
versely, temporal lobe infarcts can extend to 
involve the lower fibers of the visual pathways 
and so cause a hemianopia involving the upper 
fields. Of practical importance is the observa- 
tion that homonymous hemianopia secondary 
to occipital lesions tends to cause a very per- 
ceptible visual void to the patient. In contrast, 
the patient with homonymous hemianopia due 
to parietal or temporal lobe lesions is often 
unaware of the deficit and may bump into 
objects. 

Motor/Sensory Symptoms 

A number of cerebral pathologies can cause 
motor or sensory symptoms that may mimic 
the focal symptoms observed in TIA or stroke. 
These primarily include intracranial hemor- 
rhage, tumor, migrainous phenomena, and 
arteritis. Overall, the pattern of onset in con- 
junction with the distribution and nature of 
the symptoms enables the clinician to be more 
discriminating in developing a differential 
diagnosis. 

Ischemic events tend to have an abrupt onset, 
with the severity of the insult being apparent 
from the outset. By contrast, motor/sensory 
signs associated with migraine often progress 
from one part of the body to the next over a 
15- to 20-minute period of time. Ischemic 
TIAs rarely include positive phenomena. For 
example, the hemisensory/motor signs with 
ischemic TIAs are not usually associated with 
seizure or paresthesia but represent loss or 
diminution of neurological function. Migrain- 
ous or postictal events frequently include 
seizures, clonic contractions, and enhanced 
sensory phenomena. 

Motor/sensory deficits can be unilateral or 
bilateral, with the upper and lower limbs being 
variably affected depending on the site of the 
cerebral lesion. For example, occlusion of the 
anterior cerebral artery causes a hemiparesis, 
with the leg being more severely affected than 
the arm. The combination of a motor and 



163 



CAROTID ARTERY DISEASE 




sensory deficit in the same body territory is sug- 
gestive of a cortical thromboembolic event as 
opposed to lacunar lesions secondary to small- 
vessel disease of the penetrating arterioles. 
However, a small proportion of the latter may 
present with a sensorimotor stroke secondary to 
small-vessel occlusion within the posterior limb 
of the internal capsule. Pure sensory and pure 
motor strokes and those strokes where the 
weakness affects one limb only or does not 
involve the face are more typically seen with 
lacunar as opposed to cortical infarction. 



Higher Cortical Dysfunction 

A number of clinical phenomena, including 
speech and language disturbances, can be 
caused by thromboembolic phenomena within 
the anterior and posterior circulations. There 
are a large number of examples of higher corti- 
cal dysfunction (dysphasia, apraxia, visuospa- 
tial neglect, alexia) that are beyond the scope of 
this review. However, the most important clini- 
cal example for the dominant hemisphere is 
expressive dysphasia/aphasia, with visuospatial 
neglect being an example of nondominant 
hemisphere injury. 

Dysphasia is a language disorder as opposed 
to dysarthria, which is a locomotor speech 
problem. Differentiation between the two is im- 
portant, as dysarthria is a vertebrobasilar symp- 
tom whereas dysphasia is of carotid origin. 
In expressive dysphasia, patients know what 
they wish to say but either cannot find the 
word or produce seemingly meaningless verbal 
output. Dysarthria can be defined as a simple 
inability to "get your tongue" around the word. 
Visuospatial neglect is the result of injury to 
the nondominant parietal lobe. Here patients 
may exhibit inattention to one side of their 
body. 



Carotid or Vertebrobasilar Territory? 

Typical carotid territory symptoms (anterior 
and middle cerebral arteries) include hemi- 
sensory/motor symptoms, amaurosis fugax, 
and evidence of higher cortical dysfunction. 
Homonymous hemianopia alone is not a fea- 
ture of injury to the carotid territory. Classical 
vertebrobasilar symptoms comprise bilateral 



motor/sensory symptoms, bilateral visual loss, 
homonymous hemianopia, dysarthria, nystag- 
mus, ataxia and gait problems, dysphagia and 
dizziness, and vertigo, provided they accom- 
pany other vertebrobasilar features. 

Discrimination between carotid and verte- 
brobasilar symptoms is usually straightfor- 
ward but can be difficult. For example, 10% 
of patients with vertebrobasilar stroke have 
hemisensory/motor signs. This is because of 
anatomical variations in the vascular boundary 
zones between the carotid and vertebrobasilar 
system. Similarly, a small number of patients 
with thromboembolic carotid artery disease 
present with vertebrobasilar symptoms because 
the posterior cerebral artery is embryologically 
derived from the carotid artery as opposed to 
the vertebrobasilar system. If there is any ques- 
tion regarding interpretation of the clinical 
picture, advice from a neurologist or stroke 
physician should be sought. 

Clinical Features of 
Nonatherosclerotic Disease 

One of the characteristic features of the non- 
atherosclerotic pathologies listed earlier is the 
potential for thrombosis/stenosis and aneurysm 
formation. Accordingly, each condition can 
cause ischemic stroke/TIA, but most are rela- 
tively rare. The onset of stroke/TIA in a patient 
exhibiting other atypical features should raise 
the possibility of a nonatheromatous pathology. 
Sudden onset of temporal headache or neck 
pain associated with a neurological or visual 
deficit is suggestive of carotid dissection. The 
initial neurological symptoms are thought to 
be due to acute expansion of the dissection 
resulting in compression of cranial nerves IX, 
X, XI, or XII, followed by cerebral ischemia. 
Horner syndrome can also occur, which is 
believed to be due to disruption of periadventi- 
tial sympathetic fibers adjacent to the carotid 
artery. Systemic illness, malaise, weight loss, 
and myalgia suggest an underlying arteritis 
(Takayasu, giant cell, SLE, polyarteritis). Jaw 
claudication is an important presentation in 
giant cell arteritis. Takayasu arteritis patients 
also present with symptoms attributable to 
involved vascular beds (renovascular hyperten- 
sion, TIA/stroke, MI). 




> 



164 



VASCULAR SURGERY 



Prognosis of Ischemic Stroke 

Mortality and Persisting Disability 

Stroke is the commonest cause of persisting 
neurological disability in the Western world 
and accounts for 4.5 million deaths per annum 
worldwide. The overall mortality rate for stroke 
is about 26% at 30 days and 38% at 1 year, but 
varies according to subtype (see below). 

At 6 months, 15% to 25% of stroke survivors 
are still dysphasic/aphasic, 7% to 11% remain 
incontinent of urine or feces, up to 33% require 
help with dressing/feeding or toileting, 15% 
are unable to walk independently indoors, and 
17% are classified as moderately or severely 
disabled. 

Outcome Based on Presentation 

The Oxfordshire Community Stroke Project 
(OCSP) was the first community-based study to 
develop a bedside classification that could reli- 
ably predict outcome, vascular pathology, and 
CT scan/autopsy findings following ischemic 
stroke (Bamford et al., 1990). Patients who were 
classified as total anterior circulation infarction 
(TACI) presented with the triad of (1) hemi- 
sensory/motor deficit affecting the face, arm, 
and leg; (2) homonymous hemianopia; and 
(3) higher cortical dysfunction (e.g., dysphasia, 
visuospatial neglect). These patients had the 
largest infarction volumes on CT scan or 
autopsy, occlusion of either the extracranial ICA 
or intracranial MCA mainstem, and a 30-day 
mortality rate of 37%. Only 7% were alive and 
independent at 1 year. Patients with TACI are 
rarely ever candidates for carotid surgery. 

Patients presenting with one or two compo- 
nents of the TACI triad were classified as partial 
anterior circulation infarction (PACI). These 
patients had focal, cortical infarcts on CT scan 
and rarely had evidence of major vessel occlu- 
sion. The majority followed embolization of 
MCA branches from a carotid plaque. The 30- 
day mortality following PACI was 13%, and 71% 
were alive and independent at 1 year. Patients 
with a nondisabling PACI stroke were exposed 
to a 5% risk of recurrent stroke within 30 days 
of the initial event and a 17% risk of stroke 
within 1 year. Patients with PACI constitute 
the majority of patients being considered for 
carotid endarterectomy 



Patients classified as lacunar infarction 
(LACI) presented with symptoms and signs 
associated with disease of the deep perforating 
arteries (pure motor stroke, pure sensory 
stroke, sensorimotor stroke, and ataxic hemi- 
paresis). Patients with LACI never have evidence 
of higher cortical dysfunction and major vessel 
occlusion was not a feature. 

Patients classified as posterior circulation 
infarction (PoCI) presented with vertebrobasi- 
lar symptoms and had infarcts localized to the 
posterior circulation territory. The 30-day mor- 
tality rate was 7%, and 80% were independent at 
30 days. 



Investigation of 
Ischemic Stroke 



All patients should undergo simple baseline 
investigations (full blood count, urea/elec- 
trolytes, glucose, lipids, chest x-ray, electro- 
cardiogram). This not only enables diagnosis of 
unexpected coexistent pathologies (sickle cell 
disease, thrombocytosis, etc.), but also enables 
important risk factors to be corrected (hyper- 
tension, diabetes, ischemic heart disease, etc.). 
There is no need for more complex investiga- 
tions to be performed routinely (autoantibod- 
ies, echocardiograms, thrombophilia screens). 
These should only be undertaken if the history 
or initial investigations suggest it is appropriate. 

Duplex Ultrasound 

Noninvasive diagnostic testing should be per- 
formed in the preliminary evaluation of all pa- 
tients suspected of having suffered a stroke or 
TIA. Color duplex is now the most accessible 
screening technique for diagnosing carotid 
stenosis. One of the great advantages of duplex 
is that the machine can be brought down into a 
single visit outpatient clinic. Duplex combines 
B-mode imaging and pulsed wave Doppler sam- 
pling of velocity spectra to assess the potential 
hemodynamic significance of the carotid lesion. 
Indications for carotid duplex scanning typi- 
cally fall into three categories: symptoms, signs, 
and risk factors. Symptoms include those that 
define a classic TIA. The primary sign is the 
presence of a carotid bruit, whereas risk factors 
include diabetes mellitus, hypertension, ciga- 



165 



CAROTID ARTERY DISEASE 




rette smoking, hypercholesterolemia, peripheral 
vascular disease, and coronary artery disease. As 
the number of risk factors increases, the likeli- 
hood of an associated carotid lesion rises. 

The accuracy of a carotid duplex scan is 
largely dependent on the technician who per- 
forms the study, as well as the type of scanner 
that is used. Ultrasound criteria vary among 
units, and each vascular laboratory should vali- 
date the technical skills of the ultrasonographer 
before duplex imaging is used as the sole diag- 
nostic study. 

An increasing number of units now perform 
carotid surgery on the basis of duplex ultra- 
sound alone. However, corroborative magnetic 
resonance angiography (MRA) or diagnostic 
angiography may be required in patients with 
(1) gross calcification causing severe acoustic 
shadowing, (2) inability to image proximal or 
distal limits of plaque, (3) damped inflow wave- 
form suggestive of proximal common carotid 
disease, and (4) a high-resistance ICA waveform 
suggestive of distal severe disease. Some units 
may still wish to undertake corroborative 
studies in patients with a duplex diagnosis of 
ICA occlusion. 

Magnetic Resonance Angiography 

The principal advantages of MRA are that no 
radiation is involved, it can be combined with 
functional brain imaging, and the surgeon is 
provided with a hardcopy for easy interpre- 
tation. Disadvantages include problems with 
interpretation of images and, more importantly, 
rapid access to imaging in TIA clinics. 

The principle underlying MRA is that the 
patient is positioned within a magnetic field. 
This causes protons in the body's water mole- 
cules in the area of interest to become aligned 
with their axis to the magnetic field. Radio wave 
pulses are then applied to the region of interest 
that excite the protons and cause them to rotate. 
As the protons return to their original align- 
ment they emit energy that is picked up by 
external detectors. There are two principal 
methods for imaging blood vessels: time of 
flight (TOF) and phase contrast. 

In the TOF method, stationary tissue is dark 
because the repeated radio wave pulsations sat- 
urate their signal. In contrast, flowing blood 
emerging from the chest has not been subject to 
the radio waves and is represented by a white 



signal against the black background. Two- 
dimensional (2D) TOF involves the sequential 
acquisition of tomographic slices of data that 
are then reconstructed by a computer algorithm 
to create the axial angiogram. Three-dimen- 
sional (3D) TOF acquires multiple slices of data 
at the same time. Each has its advantages and 
disadvantages. Two-dimensional TOF is better 
at discriminating abnormalities within fields of 
slow flowing blood. Therefore, it is useful in dif- 
ferentiating subtotal occlusion from complete 
occlusion. It is, however, limited by its inability 
to reliably interpret kinks and loops, and the 
creation of flow voids tends to lead to overin- 
terpretation of stenosis degree. The latter is not 
a problem with 3D TOF. As with duplex, advo- 
cates of MRA must validate their own findings 
with either angiography or resected plaque 
specimens. 

Contrast Angiography 

Few centers now perform routine contrast 
angiography on all patients prior to surgery. 
This is partly due to the inevitable delay, but 
mainly because of the potential for angio- 
graphic-related strokes. This risk is between 1% 
and 2%. Interestingly, 50% of the operative risk 
in the Asymptomatic Carotid Atherosclerosis 
Study was attributable to the angiographic 
stroke risk. The risk of angiographic stroke is 
reduced by (1) employing arch injections where 
possible, (2) using small volumes of nonionic 
contrast material, (3) using small bore catheters, 
(4) using experienced practitioners, and (5) 
being sure the patients are well hydrated. 

Intraarterial digital subtraction arteriogra- 
phy (DSA) is still indicated in patients in 
whom there is discordance between duplex and 
MRA or problems with interpretation of these 
modalities. 



Functional Imaging 

This is another contentious issue. Some centers 
advocate routine CT or magnetic resonance 
imaging (MRI) on all stroke/TIA patients on the 
basis that unexpected pathology (neoplasms, 
vascular malformations, subarachnoid or sub- 
dural hematomas) must be considered in the 
overall differential diagnosis. Others argue that 
the yield is <2%, it incurs an unnecessary delay, 




> 



166 



VASCULAR SURGERY 



it does not alter management decisions, and it 
incurs undue cost. 

In an ideal world, all patients should undergo 
functional imaging (preferably MRI) combined 
with MRA. However, all clinicians would agree 
that patients presenting with a stroke require 
imaging within 2 weeks of onset (so as to iden- 
tify patients with intracranial hemorrhage), as 
should patients reporting atypical symptoms 
(seizure, progressive neurological symptoms, 
headaches, cranial nerve signs). Centers that 
continue to advocate routine CT/MRI must 
ensure that implementation of such a policy 
does not lead to unacceptable delays in planning 
management strategies. 

Management of Occlusive 
Carotid Artery Disease 

Best Medical Therapy 

There is more to investigating and treating 
patients with stroke or TIA than simply identi- 
fying those who might benefit from carotid 
endarterectomy More importantly, the imple- 
mentation of the best medical therapy should 
not be delegated to the most junior member of 
the team. Although some risk factors (age, sex, 
gender, family history) are nonmodifiable, there 
is an increasing body of systematic evidence to 
guide the clinician with regard to implementing 
optimal medical therapy in patients with cere- 
bral vascular disease. 



Antiplatelet Therapy 

Aspirin irreversibly blocks cyclooxygenase- 
mediated breakdown of arachidonic acid, 
thereby inhibiting the formation of thrombox- 
ane A 2 (platelet aggregator and vasoconstric- 
tor). Meta-analyses have shown no evidence that 
aspirin has any beneficial role in the primary 
prevention of stroke. However, in patients with 
a history of vascular disease, the Antiplatelet 
Trialists' Collaboration showed that aspirin con- 
ferred a 22% relative risk reduction (RRR) in all 
vascular events (nonfatal stroke, nonfatal MI, 
vascular death). A more recent meta-analysis 
has shown that aspirin confers a 15% reduction 
in stroke alone in patients presenting with 
symptomatic cerebral vascular disease. 



There has been much debate about the 
optimal aspirin dose. A balance must be struck, 
as there is a 1.0% annual risk of adverse events 
associated with aspirin therapy, with the risk 
increasing as the aspirin dose increases. In 
Europe, there is a trend toward using low-dose 
aspirin (50 to 150mg daily), whereas larger 
doses have previously been recommended in 
North America (300 to 1200 mg). Meta-analyses 
have shown that aspirin doses of <100mg daily 
confer a 13% RRR in subsequent vascular events 
in patients with a presenting history of stroke or 
TIA, falling to 9% for medium doses (300 mg 
daily) and 14% for daily doses in excess of 900 
mg. The North American Symptomatic Carotid 
Endarterectomy Trial (NASCET) originally 
observed that the incidence of perioperative 
stroke was significantly higher in patients 
receiving lower-dose aspirin. However, a sub- 
sequent randomized trial in more than 2500 
patients undergoing carotid endarterectomy 
indicated that the 30-day risk of stroke, MI, or 
death was significantly lower in patients taking 
81 mg or 325mg aspirin as compared with 650 
to 1300mg. 

Dipyridamole inhibits platelet aggregation by 
partially blocking the adenosine diphosphate 
(ADP) receptor on the platelet and by elevat- 
ing levels of cyclic adenosine monophosphate 
(cAMP) and cyclic guanosine monophosphate 
(cGMP). Dipyridamole on its own does not 
significantly reduce the risk of secondary 
stroke, but a number of studies have suggested 
that combination therapy with aspirin is more 
effective. This has been confirmed in a recent 
meta-analysis that showed that aspirin and 
dipyridamole conferred a 23% RRR in stroke. 
However, the benefit was slightly offset by the 
fact that about a quarter of patients experienced 
adverse side effects, necessitating dipyridamole 
withdrawal. 

Ticlopidine and clopidogrel have similar 
modes of action. Both inhibit platelet activation 
through inhibition of ADP binding to its platelet 
receptor, thereby blocking the GpIIb-IIIa re- 
ceptor complex that is the principal receptor 
for platelet fibrinogen binding. Although some 
studies showed a beneficial reduction in the 
risk of stroke, an overview by the Antiplatelet 
Trialists' Collaboration suggested that ticlopi- 
dine conferred no extra benefit over aspirin 
alone in terms of reducing the risk of all vascu- 
lar events. However, ticlopidine was associated 



167 



CAROTID ARTERY DISEASE 




with a 20% incidence of adverse complications 
including neutropenia in 2% and a tendency 
to increased cholesterol levels. Overall, 6% of 
patients had to stop the drug. 

Clopidogrel is the newest antiplatelet agent 
and has a greater antiplatelet effect than ticlopi- 
dine. Although the CAPRIE study observed a 
significant reduction in the incidence of vascu- 
lar events in favor of clopidogrel (75 mg daily), as 
compared with aspirin (325 mg daily), it should 
be borne in mind that the relative risk reduc- 
tion was 8.7%, the absolute risk reduction only 
0.5%, and subgroup analyses failed to show a 
significant reduction in stroke risk relative to all 
vascular events (CAPRIE Steering Committee, 
1996). In the U.K., a course of clopidogrel is 30 
times more expensive than aspirin. 

In summary, aspirin remains the antiplatelet 
agent of choice. The dose should be between 75 
and 300 mg daily, and therapy should continue 
throughout the perioperative period in patients 
undergoing carotid endarterectomy Medically 
treated patients who suffer repeated throm- 
boembolic events while on aspirin should either 
have dipyridamole (200 mg) added to the 
aspirin regime or convert to clopidogrel (75 mg 
daily). Clopidogrel is preferable to dipyridamole 
alone in patients who are aspirin intolerant. 
Surgeons, however, should be aware that clop- 
idogrel trebles the bleeding time whereas a 
combination of aspirin and full-dose clopido- 
grel increases the bleeding time by a factor of 
five as compared with aspirin alone. 

Treatment of Hypertension 

A systematic review of randomized trials com- 
prising 48,000 hypertensive patients found that 
a sustained reduction of 5mmHg in diastolic 
blood pressure over a 3-year period was asso- 
ciated with a 38% reduction in the risk of 
late stroke (Collins and MacMahon, 1994). How- 
ever, relatively few of these patients had a prior 
history of stroke or TIA. More recently, the 
Antithrombotic Trialists' Collaboration per- 
formed a review of the randomized trial data 
from 150,000 patients with cerebral or coronary 
events, and found that for every 5 mm Hg reduc- 
tion in the diastolic blood pressure, there was a 
15% relative reduction in the risk of stroke. 
Interestingly, there was no evidence of a lower 
diastolic threshold below which the risk of 
stroke did not fall. 



There remains some controversy over the 
threshold for therapeutic intervention in 
patients with hypertension. In the U.S., the 
recommendation is to maintain blood pressure 
less than 140/90 mm Hg, whereas in the U.K. 
the advice is for control to also reflect the age of 
the patient. The exception might be the diabetic 
patient with hypertension who warrants more 
careful control of blood pressure (see below). 
The one other golden rule is that patients 
undergoing carotid endarterectomy should not 
undergo surgery with uncontrolled hyperten- 
sion, as this is associated with a twofold excess 
risk of perioperative stroke. 

Smoking 

For obvious reasons, there have never been 
any randomized trials comparing stroke risk 
in patients who continue to smoke! A meta- 
analysis of epidemiological and cohort studies 
indicates that the relative risk of stroke is 
doubled in smokers as opposed to nonsmokers. 
The excess risk of stroke declines with time after 
smoking cessation and is equivalent to that of 
nonsmokers after about 5 years. 

Diabetes 

Diabetes is associated with a twofold excess 
risk of stroke, and, intuitively, one would have 
thought that careful glycemic control would be 
associated with a significant reduction in stroke 
risk. However, one of the few randomized trials 
to address this issue showed that the risk of late 
stroke appeared to be unchanged in patients 
randomized to aggressive glycemic control. The 
available evidence suggests that it is the hyper- 
tensive type 2 diabetic who is most at risk of late 
stroke. The U.K. Prospective Diabetes Study 
Group randomized hypertensive, type 2 diabetic 
patients to strict antihypertensive therapy 
(mean blood pressure 144/82 mm Hg) or less 
stringent control (mean blood pressure 154/ 
87mmHg). During follow-up, the former strat- 
egy was associated with a 44% reduction in late 
stroke. 

Treatment of Atrial Fibrillation 

About 20% of all strokes are secondary to non- 
valvular atrial fibrillation (NVAF). Approxi- 




> 



168 



VASCULAR SURGERY 



mately 5% of the population aged >65 years 
have NVAF, incurring a 3% to 5% annual risk of 
stroke. Treatment with warfarin reduces the risk 
of stroke by 68% but decisions regarding the 
role of anticoagulation must take into account 
the potential hemorrhagic risks. The annual 
risk of significant bleeding in anticoagulated 
patients is 1.3%, including a 0.3% incidence of 
intracranial hemorrhage. Patients at increased 
risk of significant hemorrhage include those 
with the following findings: a previous history 
of bleeding, age >75 years, an international nor- 
malized ratio (INR) >3.0, fluctuating INRs, and 
uncontrolled hypertension. 

Although each case must be considered 
individually, it is generally recommended that 
patients considered high risk for embolic 
stroke be warfarinized with a target INR of 
<3.0. High-risk patients (6% annual stroke risk) 
include (1) those of any age with a past history 
of TIA or stroke, rheumatic heart disease, 
ischemic heart disease, and evidence of im- 
paired left ventricular function on echocardio- 
graphy; and (2) patients aged >75 years with 
hypertension and/or diabetes. Low-risk patients 
(1% annual stroke risk), including patients 
aged <65 years with no risk factors, should be 
treated with aspirin. The medium-risk group 
(2% annual stroke risk) are more difficult to 
categorize. It comprises (1) those aged <65 years 
with a history of diabetes, hypertension, periph- 
eral vascular disease, and ischemic heart dis- 
ease who should be warfarinized; and (2) those 
aged 65 to 75 with no risk factors in whom it 
might be reasonable to treat with aspirin. 
Aspirin therapy should be considered in all 
patients with NVAF who are unable to take 
warfarin. 



Treatment of Hyperlipidemia 

In a meta-analysis of the available epidemiolog- 
ical studies (450,000 patients), no association 
was observed between cholesterol level and 
overall stroke rate. However, meaningful inter- 
pretation of these data was confounded by the 
fact that although a higher cholesterol level was 
associated with an increased risk of ischemic 
stroke, this was offset by an increased risk of 
hemorrhagic stroke in patients with lower 
cholesterol levels. A subsequent systematic re- 
view of randomized trials indicated that statin 



therapy was associated with a 25% reduction in 
the risk of late stroke, but only in those patients 
with coexistent symptomatic ischemic heart 
disease. The principal problem regarding plan- 
ning lipid lowering therapy based on evidence 
is that most of the randomized trials have been 
undertaken in patients with coronary heart 
disease. Few have specifically examined the role 
of statin therapy in patients with TIA or stroke, 
although several should be reporting in the next 
year or so. 

In the U.K., there is conflicting advice regard- 
ing statin therapy. The National Service Frame- 
work for Coronary Heart Disease recommends 
statin therapy (cholesterol to be reduced to 
<5mmol/L) in all patients with coronary heart 
disease or other occlusive disease (including 
those with TIA or stroke irrespective of cardiac 
status). In contrast, the National Clinical Guide- 
lines for Stroke recommends therapeutic 
intervention only in patients with symptomatic 
cerebrovascular disease and coronary heart 
disease. This conflicting advice will almost cer- 
tainly be revised following the recent publica- 
tion of the British Heart Protection Study. This 
trial randomized 20,000 patients with vasc- 
ular disease (including stroke/TIA) to placebo 
or simvastatin (40 mg daily). Overall, statin 
therapy conferred a 12% reduction in all mor- 
tality, a 17% reduction in vascular mortality, a 
24% reduction in all coronary events, and a 27% 
reduction in stroke. Of most importance was the 
observation that there did not appear to be any 
lower cholesterol limit below which benefit was 
not observed. 

In the U.S., statin therapy is advised in all 
patients with TIA/stroke and coronary heart 
disease. In patients with no coronary heart dis- 
ease and fewer than two vascular risk factors 
(selected from men aged >45 years, women aged 
>55 years, family history of heart disease, 
smoking, hypertension, diabetes, high-density 
lipoprotein (HDL) cholesterol <35mg/dL), the 
advice is to try dietary modification for 6 
months and to introduce statin therapy only if 
the low-density lipoprotein (LDL) cholesterol 
is >190mg/dL with a target of <160mg/dL. The 
TIA/stroke patients with no history of coronary 
heart disease (but having more than two risk 
factors) should undergo dietary modification 
for 6 months followed by statin therapy if the 
LDL cholesterol is >160mg/dL with a target of 
reducing it to <130mg/dL. 



169 



CAROTID ARTERY DISEASE 




Surgery for Occlusive disease 

Carotid endarterectomy (CEA) is one of the few 
surgical procedures to have been subjected to 
evidence-based scrutiny with large, multicenter 
randomized trials. An overview of the principal 
results from the European Carotid Surgery Trial 
(ECST), the NASCET, and the Asymptomatic 
Carotid Atherosclerosis Study (ACAS) is pre- 
sented below. One other carotid surgical pro- 
cedure (extracranial-intracranial bypass) has 
been intermittently advocated for recurrent 
TIAs/stroke in the presence of a chronically 
occluded ICA. This procedure was popularized 
during the 1980s, but a randomized trial there- 
after showed no level I evidence of benefit. The 
methodology of this study has been challenged 
but few surgeons currently advocate this form of 
surgery. 

Overview of ECST and NASCET 

In ECST and NASCET almost 6000 patients 
in over 200 centers around the world were 
randomized, comparing "best medical therapy" 
against "best medical therapy" and CEA. All 
patients had to have reported ipsilateral carotid 
territory symptoms within the preceding 6 
months; all were seen by a neurologist prior 
to randomization and all underwent CT scan- 



ning and contrast angiography. Follow-up was 
coordinated by the neurologists and stroke 
physicians. 

The basic results are summarized in Table 
15.1. Both trials showed that CEA conferred 
significant benefit in symptomatic patients with 
a 70% to 99% stenosis. The ECST found no evi- 
dence of benefit in patients with lesser degrees 
of disease. The NASCET observed a small but 
significant benefit in patients with 50% to 69% 
stenoses. The reason for these apparent discrep- 
ancies lies in the method for calculating degree 
of stenosis. The ECST compared the residual 
luminal diameter against the diameter of the 
carotid artery at the level of the stenosis (usually 
the carotid bulb). The NASCET compared the 
residual luminal diameter against the diameter 
of the ICA at least 1 cm above the stenosis. 
As a consequence, the ECST tends to systemati- 
cally overestimate stenoses (as compared with 
the NASCET method), particularly in those 
with mild/moderate disease. In reality, a 50% 
NASCET stenosis is approximately equivalent to 
a 65% ECST, whereas a 70% NASCET stenosis 
equates to an 82% ECST. 

The ECST and NASCET have identified 
predictive factors that are associated with a 
significantly higher risk of late stroke in 
medically treated patients. These include male 
sex, 90% to 94% stenosis, surface irregularity/ 
ulceration, coexistent syphon or intracranial 



Table 1 5.1 . Long-term risk of ipsilateral stroke (including perioperative stroke or death) 






Trial stenosis 


Surgical risk 


Medical risk 


ARR 


RRR 


NNT 


Strokes prevented 


(%) 


(%) 


(%) 


(%) 


(%) 




per 1000 CE As 


ECST 














<30% 


9.8 at 5 y 


3.9at5y 


-5.9 


n/a 


n/a 


n/a 


30-49% 


10.2 at 5y 


8.2 at 5 y 


-2.0 


n/a 


n/a 


n/a 


50-69% 


15.0 at 5y 


12.1 at 5y 


-2.9 


n/a 


n/a 


n/a 


70-99% 


10.5 at 5y 


19.0 at 5y 


+8.5 


45 


12 


83at5y 


NASCET 














30-49% 


14.9 at 5y 


18.7 at 5y 


+3.8 


20 


26 


38at5y 


50-69% 


15.7 at 3y 


22.2 at 3y 


+6.5 


29 


15 


67at3y 


70-99% 


8.9 at 3 y 


28.3 at 3y 


+19.4 


69 


5 


200 at 3 y 


ACAS 














60-99% 


5.1 at 5y 


11.0 at5y 


+5.9 


53 


17 


59at5y 


ACST 














60-99% 


6.4 at 5 y 


11.8 at5y 


+5.4 


46 


19 


53at5y 



ARR, absolute risk reduction; RRR, relative risk reduction; NNT, number of CEAs to prevent one ipsilateral stroke at specified time interval; n/a = 
not applicable;/, years. 




> 



170 



VASCULAR SURGERY 



disease, no recruitment of intracranial collat- 
erals, hemispheric symptoms, cerebral events 
within 2 months, multiple cerebral events, con- 
tralateral occlusion, multiple concurrent risk 
factors, and age >75 years. In contrast, a lower 
relative risk of stroke is observed in patients 
with lesser degrees of disease [70% to 79% 
stenosis (ECST), 50% to 69% (NASCET)], near 
occlusion with a string sign, smooth stenoses, 
female sex, retinal symptoms, and those pre- 
senting with lacunar stroke. 

A pooled analysis of data from the ECST, 
NASCET, and the Veterans Affairs (VA) trial 
has recently been performed (Rothwell et al., 
2003). All the angiograms were standardized to 
the NASCET method, and the overall database 
contained >6000 randomized patients. Table 
15.2 summarizes the 30-day death/ any stroke 
rates following endarterectomy for each of the 
principal trials. Overall, 7.1% of patients [95% 
confidence interval (CI) 6.3-8.1] in the three 
trials either died or suffered a stroke within 30 
days of surgery. This is an important observa- 
tion, as the track record for all participating 
surgeons was vetted before they were allowed to 
randomize patients. 

Figure 15.6 illustrates how the benefit of CEA 
increases with time and with increasing degrees 
of stenosis. By contrast, CEA was harmful in 
patients with <50% stenoses. It conferred a 
small but significant benefit in symptomatic 
patients with 50% to 69% stenoses, with the 
maximal benefit being observed in patients 
with 70% to 99% stenoses. Note, however, that 
patients with near-occlusion did not derive any 



systematic benefit from CEA. Near-occlusion 
was defined angiographically as severe stenosis 
with evidence of reduced flow in the distal ICA 
(delayed arrival of contrast in the distal ICA or 
recruitment of collateral flow toward the symp- 
tomatic hemisphere, or both) and evidence of 
narrowing of the poststenotic ICA (lumen 
diameter similar to, or less than, that of the 
ipsilateral external carotid artery and less than 
that of the contralateral ICA). Accordingly, 
there is increasing evidence that patients with 
subocclusion and distal vessel collapse should 
be treated conservatively. 



Overview of ACAS and ACST 

A number of randomized studies have evalu- 
ated the role of CEA in patients with severe 
asymptomatic carotid artery disease (MACE, 
CASANOVA, VA study), but ACAS and ACST are 
generally accepted to be the best (ACST, Lancet 
2004). The ACAS randomized 1600 patients with 
asymptomatic 60% to 99% stenoses. Table 15.1 
summarizes the principal findings. Although 
there was a 53% RRR in late stroke, this only 
equated to a 5.9% actual risk reduction at 5 
years. Clinicians in North America and main- 
land Europe have embraced the findings of 
ACAS more liberally than their U.K. or Scandi- 
navian colleagues. In the latter countries, con- 
cerns remain that the ACAS showed no benefit 
in women, there was no reduction in disabling 
stroke, and (unlike ECST and NASCET) no rela- 
tionship between increasing stenosis and stroke 



Table 15.2. Analysis of pooled data from the ECST, NASCET, and the Veterans Affairs trial: 30-day risk of death and/or stroke after 
carotid endarterectomy 



Trial 


<50% 


50-69% 


>70% 


Near occlusion 


ECST 


73/1044 
7.0% (5.4-8.6) 


37/371 

10.0% (6.9-13.1) 


17/249 

6.8% (4.0-10.8) 


3/78 

3.8% (0.8-10.8) 


NASCET 


43/663 

6.5% (4.7-8.6) 


30/421 
7.1% (4.8-10) 


14/261 

5.4% (3.0-8.8) 


5/70 

7.1% (2.4-15) 


VA309 




2/20 

10.0% (1.2-3.2) 


5/71 

7.0% (2.3-15.7) 




Combined 


116/1707 
6.8% (5.6-8.0) 


69/812 

8.5% (6.6-10.5) 


36/581 

6.2% (4.4-8.5) 


8/148 

5.4% (2.4-10.4) 



Note: The data shown are the observed percents.The 95% confidence interval is shown in parentheses. 

Source: Adapted from Rothwell et al. (2003). Here the individual data from each trial were reanalyzed and combined after having standardized 

all the angiograms to the NASCET method of stenosis measurement. 



171 



CAROTID ARTERY DISEASE 




45 1 

40 
35 
30 
25 
20 
15 
10 

5 


-5 



] ARRat3 
] ARRat5 



ARR at 8 




<30% 30-49% 50-59% 60-69% 70-79% 80-89% 90-99% near occ 
degree of stenosis 



Figure 15.6. Pooled results from European Carotid Surgery Trial (ECST),the North American Symptomatic Carotid Endarterectomy 
Trial (NASCET), and Veterans Affairs (VA) studies, showing the absolute risk reduction (ARR) in stroke at 3, 5, and 8 years relative to 
the degree of stenosis at randomization. Note that the maximal benefit was observed in patients with 90% to 99% stenoses, whereas 
patients with near occlusion and distal vessel collapse or underfilling (string sign) did not benefit from CEA. (From Rothwell et al., 
2003.) 



risk (in fact there was an inverse relationship). 
Table 15.1 also summarizes the principal results 
from the ACST. Note that the results were very 
similar to those of the ACAS. However, the key 
finding from ACST was a 50% reduction in fatal 
or disabling stroke long term. 

Carotid Endarterectomy 

Operative Principles 

The carotid bifurcation is usually approached 
via a longitudinal incision based on the anterior 
border of sternomastoid. The upper aspect of 
the incision angles posteriorly to minimize 
trauma to the greater auricular nerve. Any 
superficial veins are ligated and divided. Dissec- 
tion continues medial to the sternomastoid to 
reveal the common facial vein. This large tribu- 
tary of the internal jugular vein often overlies 
the carotid bifurcation and is a useful landmark. 
The common facial vein is divided. Round- 
toothed West retractors open up the space 
between the perilaryngeal structures and the 
jugular vein. Dissection continues medial to the 
jugular vein and the common carotid artery 
(CCA). Careful dissection is then continued 
superiorly. The external carotid artery (ECA) 



can be identified by locating its first branch (the 
superior thyroid artery). Occasionally the bifur- 
cation is rotated. The surgeon should remember 
to mobilize the ECA branches toward the first 
assistant. 

There are a number of strategies for mobiliz- 
ing the bifurcation and distal ICA. Care should 
be taken to avoid excessive mobilization of the 
bifurcation, as this leads to functional elonga- 
tion and distal ICA kinking (predisposing 
toward postoperative thrombosis) and an in- 
creased risk of procedural embolization. Most 
surgeons prefer to mobilize a discrete segment 
of ICA well above the bifurcation so as to min- 
imize the risks of dislodging an embolus into 
the cerebral circulation. The patient is systemi- 
cally heparinized and the carotid arteries cross- 
clamped. If the surgeon chooses to shunt the 
patient, the shunt is inserted now. The plaque is 
endarterectomized, tacking sutures inserted if 
required, and the ECA origin cleared of plaque 
using the eversion technique. The arteriotomy 
is closed either primarily or by a patch (vein/ 
prosthetic). Flow is restored first up the ECA, 
then the ICA. Having achieved hemostasis, the 
wound is closed with absorbable suture to re- 
constitute platysma (usually over a deep suction 
drain). Nonabsorbable suture or clips are used 
to close the skin. 




> 



172 



VASCULAR SURGERY 



Points for Debate 

Despite being a relatively straightforward oper- 
ation in concept, few operations arouse so much 
debate regarding content. A summary of the 
principal debating points follows, which high- 
lights those of most practical concern. 



Stop Antiplatelet Therapy or Not? 

There is no evidence that aspirin should be 
withdrawn during the perioperative period. An 
emerging problem, however, is the effect of the 
ADP inhibitor clopidogrel on surgical hemosta- 
sis, particularly if the patient is receiving com- 
bination antiplatelet therapy. Many surgeons 
have anecdotally observed that suture line 
bleeding is significantly increased in patients 
receiving chronic clopidogrel therapy. In the 
Leicester Royal Infirmary, clopidogrel therapy 
is stopped 1 week preoperatively and aspirin is 
restarted. 



Locoregional or General Anesthesia? 

The rationale for the use of locoregional anes- 
thesia is that the surgeon is immediately aware 
of any neurological deficit during the proce- 
dure. Accordingly, steps can be taken to treat 
this deficit. Advocates of general anesthesia 
cite ease of operation, no patient movement, 
and lower cerebral metabolic requirements to 
counter this deficit. There is no evidence that 
either anesthetic technique reduces operative 
morbidity or mortality. However, meta-analyses 
of nonrandomized trials suggest that locore- 
gional anesthesia may be associated with 
lower perioperative cardiovascular morbidity. 
A further large randomized trial (GALA) is 
currently underway in the U.K. 



Preincision Infiltration with Local Anesthesia 
and Adrenaline? 

No randomized trial data are available. The 
rationale is to minimize troublesome skin edge 
bleeding at the beginning of the procedure. 
Advocates need to be aware that subcutaneous 
and cutaneous bleeding may occur in the early 
postoperative period as the adrenaline effect 
wears off. 



Longitudinal or Transverse Incision? 

No randomized trial data are available. Trans- 
verse wounds probably confer a better cosmetic 
result, but may be associated with problems 
relating to access to the distal ICA. Longitudinal 
incisions inevitably divide more cutaneous 
nerves, leading to a larger area of anesthesia 
under the chin. The latter, however, is the 
approach of choice if there is any question of 
high carotid disease, especially in male patients 
with short fat necks! 



Anticipating Distal Disease? 

Preoperative preparation is mandatory. Nasola- 
ryngeal intubation opens the space between the 
ramus of the mandible and the mastoid pro cess. 
Nasolaryngeal intubation should probably not 
be undertaken in all patients, as there is a not 
insignificant risk of retronasal bleeding, which 
can predispose to vomiting (and aspiration) in 
the early postoperative period. If the surgeon 
feels that temporomandibular subluxation may 
be necessary (preferable to dislocation), this 
must be anticipated in advance and maxillofa- 
cial specialists must be involved. This procedure 
cannot readily be undertaken once the proce- 
dure has started. In the Leicester Royal 
Infirmary, >1200 CEAs have been performed 
over the last decade without recourse to tem- 
poromandibular subluxation. Surgeons unfa- 
miliar with dissecting in the upper reaches of 
the neck should involve more experienced col- 
leagues. High carotid dissections are associated 
with an increased risk of operative stroke, 
bleeding, and cranial nerve injury. 

When Should I Clamp the Internal Carotid 
Artery Early? 

Transcranial Doppler (TCD) allows the surgeon 
to be promptly aware of particulate emboliza- 
tion during the dissection phase of the opera- 
tion. One or two emboli warn of the likelihood 
of an unstable plaque. The dissection technique 
is then made even more meticulous, and early 
clamping is not usually necessary, provided no 
attempt is made to injudiciously dissect around 
the bifurcation. Continued embolization (des- 
pite modification in operative technique) is a 
worrisome phenomenon and merits consid- 
eration of early distal ICA clamping. There is 



173 



CAROTID ARTERY DISEASE 




usually more than enough time to complete the 
dissection safely and insert a shunt as necessary. 

Blocking the Sinus Nerve? 

There is no randomized trial evidence that 
routine blockade of the sinus nerve with ligno- 
caine significantly influences postoperative 
cardiovascular morbidity. The rationale is that 
sinus blockade minimizes intraoperative hypo- 
tension. However, this must be offset against 
the potential for postoperative reinnervation 
hypertension. In practice, once the ICA has 
been clamped, intraoperative hypotension is not 
usually a problem. 

Should I Ever Abandon a Procedure at 
This Stage? 

There are few situations warranting abandon- 
ment of the operation at this stage. The com- 
monest reason is cardiovascular instability 
(e.g., electrocardiogram evidence of myocardial 
ischemia not controlled with simple therapy, 
unstable arrhythmia). Other reasons reflect an 
unanticipated change in the risk/benefit ratios 
for the patient, that is, the benefits quoted to the 
patient preoperatively are now being exceeded 
by the potential risks as the procedure becomes 
ever more complex. Scenarios might include a 
hypoplastic carotid artery extending to the skull 
base (this can be evaluated by on-table angiog- 
raphy). Controversy then relates to whether the 
ICA should be tied off or left alone. Most would 
probably leave it alone and start the patient 
on warfarin postoperatively. Other examples 
include unexpected high disease extension in a 
patient with a unilateral asymptomatic stenosis. 
As the annual risk of stroke was only about 
2% in the AC AS and ACST trials (where high 
disease was specifically excluded), it may now be 
considered best to abandon the procedure, as 
the operative risks will be in excess of what was 
quoted preoperatively. 



What Dose of Heparin? 

Most centers administer 5000 units of intra- 
venous heparin before clamping. There is no 
evidence that heparin doses tailored to patient 
weight influences the balance between anti- 
thrombotic and hemorrhagic risk. 



Should I Shunt? 

This is an ever-enduring controversy. There is 
no evidence that a policy of routine shunting, 
selective shunting, or never shunting actually 
alters the operative risk, largely because the 
trials were small and poorly designed. Most sur- 
geons are either routine or selective shunters, 
with the choice being most likely to reflect the 
strategy of their trainer! 

What Choice of Shunt? 

The most commonly used shunts are the Javid 
(less flexible, tapered, requiring external retain- 
ing clamps) and the Pruitt-Inahara (softer, 
more flexible, smaller caliber, intraluminal 
retaining balloons). Alternatives include the 
Brener in-line shunt. No large randomized trials 
have compared the effect of shunt type on clin- 
ical outcome. Evidence suggests that 90% of 
patients have flow rates within 10% of preclamp 
levels using the Pruitt shunt. High carotid dis- 
sections probably benefit from using the Pruitt 
shunt, as the Javid requires a further centimeter 
or so of distal dissection to position the retain- 
ing clamp. This can sometimes be quite difficult 
to achieve in the upper reaches of the neck. 

Proximal or Distal End of the Shunt in First? 

No randomized trial has addressed this issue. 
Most surgeons insert the proximal (CCA) limb 
first and flush the shunt to clear debris prior to 
distal insertion. Allowing the distal ICA to back- 
vent before carefully inserting the shunt can 
also reduce distal embolism. No force should 
ever be applied during shunt insertion. Resis- 
tance should encourage the surgeon to dissect 
the carotid artery more distally to exclude an 
impacting coil. 

How Do I Know If the Shunt Is Working? 

About 3% of shunts malfunction, usually 
because of impaction of the distal limb against 
the ICA wall or carotid coil. Unless some form 
of monitoring is used, this will go unnoticed. 
Simple techniques include awake testing or 
TCD. For those without any access to monitor- 
ing, the surgeon using the Pruitt shunt can 
temporarily clamp the CCA inflow and assess 
backflow from the circle of Willis (i.e., is the 




> 



174 



VASCULAR SURGERY 



distal channel patent or not) by opening the red 
tap on the T limb. 

The Shunt Is Working But I Am Still Worried 
About Perfusion 

Advocates of TCD recommend trying to main- 
tain mean middle cerebral artery blood flow 
velocity of >15cm per second. This threshold 
broadly corresponds to that equivalent to the 
level of brain perfusion usually associated with 
loss of cerebral electrical activity. Note that this 
threshold does not equate to loss of neuronal 
function. If flow rates are considered inade- 
quate, check that the shunt limbs are neither 
impacted nor kinked. If there is no mechanical 
problem, the anesthetist can then carefully 
elevate the patient's blood pressure pharmaco- 
logically, which usually raises the intracranial 
blood flow to acceptable levels. 

Traditional or Eversion Endarterectomy? 

Traditional endarterectomy involves the 
removal of the plaque through a longitudinal 
arteriotomy extending from the CCA into the 
ICA. The plaque is usually transected proxi- 
mally and removed with cephalad dissection 
using a Watson-Cheyne dissector. The distal 
end is feathered or transected. Eversion endar- 
terectomy involves transecting the ICA from 
the bifurcation. Everting the enclosing media/ 
adventitia then expels a tube of plaque. The CCA 
is endarterectomized and the ICA reimplanted 
onto the bifurcation. Potential advantages of 
eversion endarterectomy include no need for 
patching and the ability to shorten an elongated 
ICA. However, neither technique is superior to 
the other. 



Patch or Not? 

This is another of the single-issue subjects that 
have characterized carotid surgery. Options 
include routine patching, selective patching, 
and never patching. No randomized trials have 
compared selective with routine patching. How- 
ever, a meta-analysis of the six randomized 
trials comparing routine patching with routine 
primary closure showed that patching conferred 
a three- to fourfold reduction in (1) periopera- 
tive thrombosis, (2) perioperative stroke, (3) late 
stroke, and (4) and late restenosis (Counsell 
et al., 1997). Accordingly, there is little evidence 
to support continuance with a policy of routine 
never patching. 



Choice of Patch? 

A meta-analysis by the Cochrane collaboration 
has shown that patch type does not influence 
outcome. There is, however, a perception that 
prosthetic patches are probably more thrombo- 
genic than vein. A recently published random- 
ized trial addressed this issue and observed 
no difference in the rates of embolization in 
the early postoperative period. In reality, pa- 
tient factors seem more likely to mediate 
an increased risk of thrombosis than does the 
patch type. The Achilles heel of prosthetic 
patching is infection, which complicates approx- 
imately 1% of procedures. About half become 
apparent in the first few months of surgery, the 
commonest organisms being staphylococci and 
streptococci. Evidence suggests that removal of 
the prosthetic material with autologous recon- 
struction is associated with the lowest risk of 
stroke and late reinfection. Secondary recon- 
struction with prosthetic material should be 
avoided. 



Tack the Intimal Step or Not? 

No randomized trial data are available, and atti- 
tudes usually reflect the policy of the trainer. 
The NASCET observed that tacking sutures 
were associated with a higher perioperative risk. 
This nonrandomized comparison, however, may 
simply reflect the fact that tacking sutures were 
more likely to be inserted when the surgeon 
encountered a more complex case (e.g., difficult 
to achieve a clean, tapering distal end-point in a 
thin walled vessel). 



Should Vein Be Harvested from Ankle 
or Groin? 

Vein patches are susceptible to rupture. This 
characteristically occurs on the 5th to 7th post- 
operative day and is more common in females 
and in hypertensives where the vein has been 
harvested from the ankle. Current advice, there- 
fore, is to use groin saphenous vein for patching 
because of its intrinsically stronger structure. 
Only about 15% of patients need their saphe- 



175 



CAROTID ARTERY DISEASE 




nous vein for alternative cardiovascular recon- 
structions in the next 5 years. 

Carotid Bypass? 

Carotid bypasses should not be performed rou- 
tinely but are indicated if the endarterectomy 
zone is too thin after plaque removal, if there 
is extensive atheroma and concomitant kink- 
ing, or if there is any question of infection. In 
common with infrainguinal venous recon- 
structions, about 20% of carotid bypasses using 
saphenous vein develop a recurrent stenosis 
within 24 months. This is significantly higher 
than that observed with traditional endarterec- 
tomy and patching. Accordingly, bypass patients 
require serial duplex surveillance. 

Shorten the Carotid or Not? 

Following endarterectomy, particularly if there 
is a slightly redundant distal ICA, there is a 
tendency to observe functional elongation of 
the endarterectomy zone. Uncorrected, this may 
predispose to perioperative thrombosis. Short- 
ening the endarterectomy zone may reduce this. 
Options include transection and reanastomosis 
(may be difficult in thin-walled, narrow-caliber 
vessels in female patients) or eversion plication. 
In the latter, stay sutures incorporating the 
redundancy on either side of the artery are tied, 
causing the excess arterial wall to be everted. 
The eversion is then closed with a continuous 
6:0 Prolene suture. 

Monitor or Not? 

There is considerable skepticism as to whether 
monitoring reduces stroke risk after CEA. 
Options include electroencephalogram (EEG), 
somatosensory sensory evoked potentials 
(SSEPs), TCD, near-infrared spectroscopy, 
jugular venous oxygen saturation, awake test- 
ing under locoregional anesthesia, xenon blood 
flow measurement, subjective assessment of 
backflow, and stump pressure. In practice, the 
most commonly used techniques are awake 
testing, TCD, and EEG. To date, no randomized 
trial has been performed to address this issue. 
However, for any form of monitoring to work, 
the right questions must be asked. Electroen- 
cephalography and SSEP only tell the surgeon 
that perfusion has dropped below the threshold 
for maintaining electrical function. Awake 



testing identifies any neurological deficit 
(indicative of perfusion having fallen below the 
threshold for neuronal viability), but it requires 
additional strategies (e.g., some sort of quality 
control) to identify what was actually causing 
the underlying problem. It has been the experi- 
ence at the Leicester Royal Infirmary that com- 
bining a monitoring method (TCD) with quality 
control assessment (angioscopy) has been asso- 
ciated with a 60% reduction in the operative risk 
over the last 10 years. 

Quality Control or Not? 

Quality control assessment is a slightly different 
concept from monitoring. Its use is based on 
the observation that the majority of operative 
strokes follow inadvertent technical error. 
Accordingly, detection of technical error should 
(in theory) reduce the risk. Once again, no 
randomized trials have evaluated completion 
angioscopy, angiography, or duplex ultrasound. 
Advocates of monitoring and quality control 
assessment have reported significant benefits. 
However, such a policy is helpful only if the 
choice of technique addresses the appropriate 
question. Awake testing does not prevent 
embolization during carotid dissection. Awake 
testing remains the only "gold standard" for 
determining who benefits from selective 
shunting, but does not prevent a stroke due 
to embolization of luminal thrombus follow- 
ing restoration of flow unless something like 
angioscopy is employed. 

Drain or Not? 

Surprisingly, no randomized trial has addressed 
this issue. However, most surgeons insert a 
suction drain into the neck wound for 12 to 24 
hours on the basis that it probably does no 
harm. There is certainly no evidence that drains 
increase the risk of prosthetic patch infection. 

Early Postoperative 
Management 

Establishing the Neurological Status 

Following surgery, the patient must be carefully 
monitored for three clinical variables: neurolog- 
ical status, wound stability, and blood pressure. 




> 



176 



VASCULAR SURGERY 



If general anesthesia has been used during the 
operation, the patient should be closely ob- 
served to ensure that cerebral function remains 
intact upon awakening. The initial assessment 
should include the patient's response to simple 
verbal commands and extremity movement. 
Once the patient becomes fully alert in the 
recovery room, a more detailed assessment 
including the vagus and hypoglossal nerve 
function can be performed. 



that regular review is required as (quite often) 
the hypertension is a transient phenomenon 
and may require therapy only for 4 to 6 hours. 
Sustained postoperative hypertension at 3 to 5 
days requires assessment by a cardiovascular 
physician. Caution should be exercised in dis- 
charging patients before blood pressure is 
adequately controlled, as these patients are 
probably at higher risk of suffering seizures in 
the early postoperative period. 



Neck Hematoma 

The neck incision should be regularly moni- 
tored for swelling. This is usually a result of 
hematoma formation, although surprising 
amounts of tissue edema can accumulate within 
hours of the procedure. Despite achieving com- 
plete hemostasis and placing a closed suction 
drain in the wound, delayed bleeding result- 
ing in hematoma and airway compromise can 
occur. Once an expanding hematoma is recog- 
nized, the patient should be returned to the 
operating room promptly to evacuate the 
hematoma and identify the potential bleeding 
source. If the patient develops respiratory dis- 
tress, the neck incision should be opened at the 
bedside to decompress the hematoma followed 
by orotracheal intubation to establish an airway 
if needed. Alternatively, it is useful to have infil- 
trated the neck incision with local anesthetic 
agent prior to finishing the procedure. Should 
a significant neck hematoma occur, it can be 
released without having to reintubate the 
patient. 

Hypertension and Hypotension 

Significant, sustained hypotension is not usually 
a problem following endarterectomy and tends 
to respond well to colloid replacement. Early 
postoperative hypertension is more common 
and requires careful monitoring and control. 
As a rule, most anesthetists prefer to maintain 
blood pressure within 10% of preoperative 
levels. There is anecdotal evidence from reports 
documenting outcome for carotid surgery 
under locoregional anesthesia that postopera- 
tive hypertension is less of a problem. Most 
cases of early postoperative hypertension 
respond to a single bolus or carefully titrated 
infusion of beta-blocker (e.g. labetalol). Note 



The Patient Suffers a 
Neurological Deficit 

Intraoperative 

An intraoperative stroke is defined as having 
occurred if a neurological deficit becomes 
apparent as the patient recovers from anesthe- 
sia. Usually the first sign is a prolonged delay in 
awakening. In the absence of any monitoring 
modality, it should be assumed that the patient 
has suffered either a carotid thrombosis or 
embolization into the cerebral arteries, and 
the patient should be reexplored. The aim of 
reexploration is to recognize and treat throm- 
bosis before an irreversible neurological deficit 
occurs. Delay beyond 1 hour is associated with 
a progressively worsened outcome. The second 
reason is to identify and correct a source of 
technical error responsible for platelet accumu- 
lation and secondary embolization. 



Neurological Deficit Within the First 24 Hours 

Focal neurological deficits occurring within the 
first 24 hours are still usually thromboembolic. 
In the absence of any monitoring techniques, it 
is again probably best to return the patient 
immediately to the operating room. Intracranial 
hemorrhage is extremely rare within the first 24 
hours, and a CT scan is probably unnecessary 
unless it is done immediately on the way to the 
operating room. 

In the Leicester Royal Infirmary, we have pre- 
viously observed that the majority of early post- 
operative neurological deficits were secondary 
to carotid thrombosis. Subsequent research 
showed that patients at risk of progressing to 
thrombosis had a 1- to 2-hour period of increas- 
ing embolization on transcranial Doppler 
before any neurological deficit became appar- 



177 



CAROTID ARTERY DISEASE 




ent.This association has now been corroborated 
in many centers around the world. This phase 
of sustained embolization can be arrested by 
intravenous infusion of Dextran 40 in saline, 
the dose of which has to be increased in 30% 
of patients in order to control embolization. 
No case of stroke due to early postoperative 
thrombotic occlusion has been encountered fol- 
lowing 1000 CEAs in the Leicester Unit since 
the protocol of 3 hours of TCD monitoring 
with selective Dextran therapy was instituted in 
1995. 

The algorithm in Figure 15.7 summarizes the 
current strategy in the Leicester Royal Infirmary 
for managing a neurological deficit in the first 
24 hours after a successful recovery from anes- 
thesia. Patients with hemiplegia/hemiparesis 
are more likely to have suffered a major vessel 
occlusion and are probably more likely to bene- 
fit from reexploration. Patients with a mono- 
paresis or isolated dysphasia are highly unlikely 
to have suffered major vessel occlusion, and 
there is time for more discriminating inves- 
tigation and treatment. The aim is to avoid 
unnecessary reexploration in patients with a 



normal endarterectomy zones and in those who 
have suffered embolic occlusion of the middle 
cerebral artery mainstem. Note that transcranial 
Doppler is used to detect any evidence of 
ongoing embolization and can be invaluable in 
guiding decisions regarding reexploration. 

Neurological Deficit After 24 Hours 

Strokes occurring after 24 hours have elapsed 
have a greater number of potential underlying 
causes. In addition to thromboembolism, the 
clinician must now consider the likelihood of 
hyperperfusion and intracranial hemorrhage. 
Figure 15.8 summarizes the current Leicester 
protocol in this situation. The key difference 
from the strategy for managing strokes within 
the first 24 hours is the importance of perform- 
ing an emergency CT scan. Intracranial hemor- 
rhage complicates 1% of CEAs and carries a 
very poor outcome, with up to 80% of patients 
dying or suffering a major disabling stroke. 
There is currently very little that can be done 
to treat these patients, although vascular sur- 
geons may like to ask their neurosurgical 



neurological deficit within 24 hours 



hemiplegia 



r 



TCD and Duplex assessment 

I L_ 



ICA 
occ 



ongoing 
emboli 

I 



No MCA 
emboli 



no improvement 
in 30 minutes 



deficit 
improves 



emergency 
exploration 



Dextran 
therapy 



monoparesis 



MCA main 
stem occln 



MCA 
normal 



Conservative management 

exclude cardiac sources 

CT scan at 3-5 days 



Figure 1 5.7. Algorithm for the investigation and management of patients at the Leicester Royal Infirmary who have suffered a neu- 
rological deficit within the first 24 hours of CEA, having made a normal recovery from anesthesia. CT, computed tomography; ICA, 
internal carotid artery; MCA, middle cerebral artery; TCD, transcranial Doppler. 




> 



178 



VASCULAR SURGERY 



neu. 



] eficit after 24 hours 



TCD + Duplex + CT scan 



no hemorrhage present 



emergency 
reexploration 



intracranial hemorrhage 



1 



hyperperfusion 

high MCAV 

hypertension 

seizures 

+ /- CT edema 



Figure 1 5.8. Algorithm for the 
investigation and management 
of patients at Leicester who 
have suffered a neurological 
deficit after 24 hours or more 
have elapsed after carotid 
endarterectomy (CEA). 



conservative Mx 
blood pressure control 
anticonvulsants 



colleagues whether they might ever consider 
clot evacuation. 

Stroke due to hyperperfusion syndrome is 
another poorly understood entity, and there is 
considerable controversy as to whether it would 
be better renamed the "reperfusion" syndrome. 
More recently, there has been renewed debate as 
to how much the condition is associated with 
high flow as opposed to hypertensive encepha- 
lopathy or dysautoregulation. Patients at risk of 
hyperperfusion-related stroke tend to have a 
history of severe bilateral disease and treated 
hypertension. Stroke is often preceded by 
seizure and severe hypertension. The main- 
stay of management is control of seizures and 
careful reduction of blood pressure. Early CT 
scans often show changes consistent with an 
evolving ischemic infarction, but in fact this is 
often white matter edema. Paradoxically, a lot of 
the white matter edema is present in the verte- 
brobasilar territory. 

Late Postoperative Management 

There is a surprisingly good transatlantic con- 
sensus regarding many of the principal issues 
relating to carotid surgery. Two, however, serial 
noninvasive surveillance and reintervention for 
recurrent stenoses, have aroused considerable 
debate. 



Serial Surveillance and Recurrent Stenoses 

In North America and in many centers in main- 
land Europe, follow-up carotid duplex scan- 
ning is considered mandatory. The underlying 
rationale is to monitor for recurrent lesions, as 
well as for progressive disease of the contralat- 
eral carotid artery. Patients are recommended 
to return for an initial follow-up visit approxi- 
mately 1 month following the operation to 
ensure that adequate wound healing has taken 
place. A carotid duplex scan is performed at that 
time to evaluate the carotid reconstruction and 
to provide a new baseline for future study com- 
parison. This is followed by a subsequent clinic 
visit at 6 months, at which time a repeat carotid 
duplex scan is performed. If the duplex scan 
shows satisfactory results of both carotid arter- 
ies, the patient may be seen 1 year following 
the operation. A bilateral carotid duplex scan 
should be performed at that visit and at all 
future annual examinations. 

In the U.K., parts of northern mainland 
Europe, and Scandinavia, the policy is com- 
pletely different. Here patients are reviewed at 
4 to 6 weeks, and unless they are part of an 
ongoing research program, they are discharged 
with instructions to return if any new symp- 
toms recur. The rationale underlying this 
approach is based on the following observa- 
tions. First, the annual risk of stroke after sue- 



179 



CAROTID ARTERY DISEASE 




cessful endarterectomy is only about 1% to 
2% in the operated hemisphere and 2% in the 
contralateral hemisphere. Second, it has been 
difficult to determine the actual risk of ipsilat- 
eral stroke in patients with a recurrent stenosis, 
largely because many of the patients have 
already been subjected to a prophylactic redo 
CEA. However, in Frerick's recent overview, 10 
published series documented the risk of stroke 
according to whether the patient had a recur- 
rent stenosis or not. Having corrected for the 
effects of different lengths of follow-up and 
study size, a reanalysis of the data suggests that 
the incidence of ipsilateral stroke in patients 
with a recurrent stenosis (>50%) is approxi- 
mately 5.5% at 3 years (i.e., only about 2% per 
annum). For those with no evidence of a re- 
current stenosis, the risk of ipsilateral stroke 
was 3.3% over 3 years (i.e. only about 1% per 
annum). These figures correlate closely with the 
follow-up data reported in the international 
trials. Third, acute changes in plaque morphol- 
ogy tend to precede onset of symptoms rather 
than a progressive worsening of stenosis. In 
short, if one reviews the results of the last sur- 
veillance scan before onset of neurological 
symptoms, it rarely enables reliable prediction 
of patients at risk of subsequent stroke. 



Carotid Angioplasty 

Angioplasty is an accepted component of 
vascular practice apart from the cerebral cir- 
culation. The reason for this discrepancy is 
the potential for procedural embolization and 
stroke. The first carotid angioplasty was per- 
formed in 1980. By 1992, the first overview of 
balloon angioplasty in the carotid artery was 
published. In a pooled series of 123 patients 
from 10 studies, the procedural mortality rate 
was 0%, the disabling stroke rate 0%, and the 
30-day death and any stroke rate was 0.8%. 
This study was thereafter a catalyst for the 
increasing interest in the potential for angio- 
plasty to replace endarterectomy as the gold 
standard treatment for occlusive carotid artery 
disease. 

However, despite the excellent results from 
these pioneers of angioplasty (particularly as 
they appeared safer than diagnostic angio- 
graphy alone), concerns thereafter were voiced 



that balloon angioplasty alone was probably 
not safe. This observation was based on con- 
cerns that vessel recoil, intimal dissection, 
plaque dislodgment, and particulate emboli- 
zation were now major potential problems, 
despite not having been evident in the original 
overview. It was proposed that the insertion 
of a stent would avoid the above problems 
as well as reduce the rate of re-stenosis. A 
subsequent overview of outcomes following 
primary stenting revealed a 4.3% death/stroke 
rate at 30 days in 2569 patients with either 
symptomatic or asymptomatic patients and a 
re-stenosis rate of <5%. Interestingly, there is 
now a third-generation view that these results 
can be improved even further with the use of 
cerebral protection devices. These devices 
(distal balloon, distal aspiration, distal filters 
and umbrellas, and retrograde flow during 
balloon insufflation) are currently under devel- 
opment or clinical evaluation. 

To date, the results from voluntary stent reg- 
istries and single centers around the world have 
clearly demonstrated that angioplasty is feasible 
and accepted by patients. It remains to be seen 
when and where it should replace endarterec- 
tomy. The situation regarding carotid angio- 
plasty, therefore, is analogous to that faced by 
the cardiac surgeons in the 1980s when coro- 
nary angioplasty was first introduced. Ulti- 
mately, the answer will have to be determined 
by level I randomized trials, and it is inevitable 
that angioplasty will have a role in the manage- 
ment of selected patients with cerebral vascular 
disease. Evidence suggests that the worst 
procedural outcomes are to be encountered in 
older patients, symptomatic patients, those with 
lesions longer than 10mm, and those with more 
severe degrees of stenosis. 

An overview of the published literature in 
2000 indicated that the results of angioplasty 
were generally poorer than surgery in trials 
reporting outcomes in symptomatic patients 
who should, otherwise, form the mainstay of 
current treatment (Golledge et al., 2000). Angio- 
plasty cannot simply be justified because it is 
effective in predominantly asymptomatic popu- 
lations. Using the results of ACAS as a guide, 
only 58 ipsilateral strokes would be prevented 
per 1000 angioplasties on asymptomatic pa- 
tients with 60% to 99% stenoses. 

Five randomized trials have now compared 
CEA and angioplasty in the treatment of symp- 




> 



180 



VASCULAR SURGERY 



tomatic patients with carotid artery disease. 
Four have been published in peer-reviewed 
journals, whereas the SAPPHIRE study has only 
recently been presented to the American Heart 
Association. 

The Leicester trial was abandoned after a 
small number of patients were recruited 
because of an excess risk of stroke in the angio- 
plasty group. The WALLSTENT study was aban- 
doned after 1 year, again because of a threefold 
excess risk of stroke in the angioplasty arm. The 
fourth trial was performed in a community 
hospital in the U.S. that reported no early or 
late strokes at all in either arm. The largest ran- 
domized study to date is the CAVATAS. This trial 
randomized 504 patients and found that at 1 
year there was no significant difference in the 
rate of ipsilateral stroke relative to angioplasty 
or surgery. However, the re-stenosis rate was 
significantly higher in the angioplasty group. 
The most worrisome aspect of the CAVATAS 
trial was the excessive 30-day risk of stroke or 
death in both the CEA (9.9%) and angioplasty 
patients (10.0%). The SAPPHIRE study ran- 
domized 307 patients to CEA or angioplasty. 
The 30-day risks of death/stroke were not 
significantly different (4.4% following angio- 
plasty versus 6.1% after surgery), but the major 
adverse event rate (30-day risk of death/ 
stroke/Ml) was 5.8% versus 12.6% in favor of 
angioplasty (presented at the American Heart 
Association in 2002). The inclusion of MI as a 
hard end-point is a new development in the 
history of performing randomized carotid 
surgery trials. 

Evidence suggests that following a successful 
angioplasty, the risk of recurrent symptoms and 
severe re-stenosis are low. A number of centers 
have documented a zero incidence of late stroke 
with restenosis rates of <5% at 12 months. In 
CAVATAS, the rate of re-stenosis at 12 months 
was 14%. 

In the U.K., angioplasty is generally reserved 
for use in randomized trials, although it is 
probably the optimum treatment for those few 
recurrent stenoses after CEA and particularly 
carotid vein bypasses requiring reintervention. 
In Europe and the U.S., the indications are prob- 
ably more liberal but should remain within 
institutional review board control. 



Future Perspectives 

There are a number of key issues that must be 
addressed and implemented in the next 5 years: 

• Researchers must ensuring that the popu- 
lation-based results are equivalent to those 
of the ECST and NASCET. 

• Surgeons must know and quote their own 
results rather than those from the ECST 
and NASCET. 

• The aim of treatment is not simply identi- 
fying those with operable carotid disease. 
Optimal medical therapy must be imple- 
mented in everybody. 

• High-risk symptomatic and asymptom- 
atic patient subgroups must be identified 
to optimize clinical benefit and cost- 
effectiveness. The simple fact remains that 
at 3 years, NASCET and ECST have shown 
that 70% of symptomatic patients did not 
need surgery (or by inference angioplasty), 
whereas ACAS showed that almost 90% 
of asymptomatic patients did not need 
intervention. 

• Randomized trials, as opposed to personal 
dogma and interdisciplinary turf wars, 
should determine which patients will 
benefit from surgery or angioplasty in the 
future. 



References 



Bamford J, Sandercock P, Dennis M, Burn J, Warlow C. ( 1990) 

J Neurol Neurosurg Psychiatry 53:16-22. 
Bonita R. (1992) Lancet 339:342-4. 

CAPRIE Steering Committee (1996) Lancet 348, 1329-39. 
Collins R, MacMahon S. (1994) Br Med Bull 50:272-98. 
Counsell CE, Salinas R, Naylor R, Warlow CP. (1997) Eur J 

Vase Endovasc Surg 13:345-54. 
Golledge J, Mitchell A, Greenhalgh RM, Davies AH. (2000) 

Stroke 31:1439-43. 
Loftus IM, Naylor AR, Bell PR, Thompson MM. (2000) Br J 

Surg 89:680-94. 
Rothwell PM, Eliasziw M, Gutnikov SA, et al. (2003) Lancet 

361:107-16. 
Zarins CK, Giddens DP, Bharadvaj BK, Sottiurai VS, Mabon 

RF, Glagov S. (1983) Circ Res 53:502-14. 



16 



Arch Vessel, Vertebrobasilar, and 
Upper Extremity 

Eva M. Rzucidlo and A. Ross Naylor 




The innominate, proximal common carotid and 
upper limb arteries are relatively rare sites for 
atherosclerotic disease, arteritis, and trauma. 
Accordingly, there is little evidence-based liter- 
ature to guide clinical practice. The Joint Study 
of Arterial Occlusion reported that the incidence 
of severe lesions demonstrated by arteriography 
in the innominate and proximal subclavian 
artery was 17%. However, of the 2000 operations 
performed at the University of California- 
San Francisco for supra-aortic disease, only 
7.5% were undertaken for innominate, common 
carotid, or subclavian artery lesions. 



The Innominate Artery 

Etiology 

Innominate artery (IA) occlusive disease is 
uncommon, representing only 1.7% of all 
operations performed at the University of 
California-San Francisco for occlusive lesions 
involving the brachiocephalic trunk, vertebral 
arteries, and carotid bifurcation. Most contem- 
porary studies report that 61% to 84% of 
patients with innominate artery disease have 
concomitant arch, vertebral, or carotid bifurca- 
tion lesions (Rhodes et al., 2000). 

In the United States the most common etiol- 
ogy of innominate artery occlusive disease is 
atherosclerotic disease (Fig. 16.1). Other impor- 
tant etiologies include Takayasu's arteritis 
(second most common in the United States), 



giant cell arteritis, and radiation-induced ather- 
osclerosis. Trauma is relatively uncommon, 
but may follow a deceleration injury in major 
vehicle accidents or penetrating injury. Innom- 
inate artery occlusive disease tends to occur in 
patients in their fifth and sixth decades (Azakie 
et al., 1998). In the United States, there appears 
to be a trend toward the majority being women. 
Conversely, in France, Kieffer et al. (1995) 
reported that 72% of the 148 patients undergo- 
ing innominate artery reconstruction for ather- 
osclerosis in their practice were men. 

Symptoms 

Symptoms largely depend on the underlying 
etiology, anatomy, and co-involvement of other 
major vessels. For example, in the bovine 
anomaly (Fig. 16.2) the innominate and left 
common carotid arteries arise from a common 
origin, and therefore any symptoms could 
involve either the carotid territory or the verte- 
bral system. In general, innominate artery 
lesions present with symptoms of ischemia of 
the right upper extremity, symptoms involving 
anterior cerebral circulation, symptoms refer- 
able to the posterior circulation, or combined 
upper extremity and neurological symptoms. 
Upper extremity symptoms occur in 5% to 
63% of patients and include claudication due 
to hemodynamic compromise or embolic phe- 
nomenon from unstable proximal lesions. A 
combination of upper extremity and neurolog- 
ical symptoms occurs in 30% to 40% of patients. 



181 




182 



VASCULAR SURGERY 




Figure 16.1. A: Arch angiogram showing occlusion of the innominate artery and a severe stenosis at the origin of the left common 
carotid artery (arrow). B: Delayed films following on from part A.The right subclavian and distal innominate arteries are filled via ret- 
rograde flow down the right vertebral artery. The actual innominate occlusion is relatively short. 




Figure 16.2. Bovine arch anomaly. The innominate and left 
common carotid arteries have a common origin (arrow). There is 
a large atherosclerotic lesion causing a severe stenosis of the 
bovine origin and total occlusion of the left subclavian artery. 



Cerebrovascular symptoms [transient ische- 
mic attack (TIA)/stroke] may be due to 
thromboembolism into the carotid circulation 
(hemisensory/motor signs, monocular blind- 
ness, or higher cortical dysfunction) or the 
vertebrobasilar system (bilateral sensory/motor 
signs, dysarthria, ataxia, nystagmus, homony- 
mous hemianopsia, cortical blindness, etc.). 
Neurovascular symptoms affect 5% to 90% of 
patients with severe innominate artery disease. 
Ten percent of patients present with global 
ischemia involving both the carotid and verte- 
bral territories. 



Diagnosis 

Many of the clinical features of innominate 
artery occlusive disease are of equal relevance to 
subclavian and vertebral artery disease and are 
reviewed in this section. Atherosclerotic disease 
of the supra-aortic trunk is suggested by absent 
pulses in the neck (subclavian, carotid) or arm 
(axillary, brachial) in one or both sides. Unequal 
or abnormally low blood pressures in the upper 
extremity should also be noted. Physical exam- 
ination should include palpation and ausculta- 



183 



ARCH VESSEL, VERTEBROBASILAR, AND UPPER EXTREMITY 




tion of the proximal and midcervical carotid 
pulses and palpation of the superficial tem- 
poral, subclavian, brachial, radial, and ulnar 
pulses. Carotid or subclavian artery bruits or 
thrills suggest innominate artery or other 
supra-aortic trunk stenotic lesions. Absent 
pulses are suggestive of occlusions. 

Blood pressure comparison of both upper 
extremities is mandatory. If bilateral upper 
extremity blood pressures are determined to be 
low, comparison should be made to those in the 
lower extremity. The hands should be inspected 
for cyanosis, ulcers, subungual splinter hemor- 
rhages, or livedo reticularis, which may indicate 
ulcerated proximal lesions predisposing to 
atheroemboli. In patients with subclavian steal 
(see later), a pulse delay may be detected in the 
radial artery. 

A computed tomography scan of the brain is 
an essential part of the workup of any patient 
with suspected atherosclerotic disease of the 
supra-aortic vessels. About 80% of the cerebral 
cortex is clinically silent, and the finding of 
areas of infarction (in the absence of any previ- 
ous symptoms) may be helpful in planning 
management strategies. 

Duplex evaluation of the proximal supra- 
aortic vessels may not be possible because they 
may be out of the scanning range of most 
machines. However, an experienced sonogra- 
pher will warn the surgeon of damped wave- 
forms in the common carotid or subclavian 
vessels. This should alert the surgeon to the like- 
lihood of inflow disease and the need for 
employing other imaging modalities. 

Arch arteriography remains the gold stan- 
dard for defining the anatomy of the lesion, 
extent of disease, and probable etiology (e.g., 
atherosclerosis versus arteritis). A complete 
examination should include an arch aortogram, 
selected carotid views, vertebral imaging, and 
images of the intracranial circulation. 

Concomitant coronary artery disease is 
present in 40% to 50% of patients with supra- 
aortic trunk disease. Inevitably, these patients 
have other cardiovascular risk factors including 
hypercholesterolemia, history of stroke, TIAs, 
peripheral vascular disease, hypertension, dia- 
betes, and renal insufficiency. A prior history of 
smoking is present in 78% to 100% of patients 
with supra-aortic trunk disease. Accordingly, 
cardiac evaluation is advisable, especially if any 
major reconstructions were planned. 



Treatment 

Historically, supra-aortic trunk disease was 
treated by direct reconstruction. In 1956 Davis 
et al. first described transthoracic innominate 
endarterectomy. Later, in 1958, DeBakey des- 
cribed prosthetic bypass grafting of the great 
vessels. In appropriately selected patients, both 
forms of direct reconstruction can give excellent 
results. 

Most patients with supra-aortic trunk ather- 
osclerosis have multiple supra-aortic trunk 
lesions. Multiple arch vessel involvement occurs 
in 61% to 84% of patients. The best operative 
repair of this patient population having multi- 
ple arch vessel lesions may therefore be directed 
reconstruction with bypass grafting. 

Bypass grafting has an excellent technical 
success rate and good early results with relief of 
symptoms in up to 95% of patients. Long-term 
relief of symptoms appear equally as good, with 
primary patency rates of 95% being evident. 
The two largest series were from Berguer et al. 
(1998) (100 consecutive patients followed over 
16 years) and Kieffer et al. (1995) (148 patients 
followed over 20 years). Berguer et al. reported 
5- and 10-year cumulative primary patency 
rates of 87% and 81%, respectively. Stroke-free 
survival rates at 5 and 10 years were 87% 
and 81%, respectively. Kieffer et al. reported a 
primary patency rate of 98% and 96% at 5 
and 10 years, respectively. The probability of 
freedom from ipsilateral stroke was 98% and 
96% at 5 and 10 years, respectively. Life-table 
analysis and perioperative events were used to 
calculate the probability of survival, which was 
77% and 51% at 5 and 10 years, respectively. 
Most patients died from cardiac events. 

The excellent outcomes following bypass 
grafting of supra-aortic trunk disease, however, 
does not come without a price. Perioperative 
mortality ranges from 0% to 15%. In the more 
recent studies, the rate generally is around 5% 
and is mostly attributable to cardiac causes. The 
perioperative stroke rate ranges from 0% to 8% 
giving a combined stroke and death rate of 
almost 16%. Rhodes et al. (2000) have deter- 
mined risk factors for early and late complica- 
tions in these patients. Patients with serum 
creatinine levels of >2 had a significantly higher 
combined stroke/death rate when compared to 
patients with normal creatinine (50% versus 
7%). Similarly, stroke rates were higher in 




» 



184 



VASCULAR SURGERY 



patients with preoperative evidence of a hyper- 
coagulable state (33% versus 4%). 

Although there have been no randomized 
controlled trials, endarterectomy has been 
reported to confer the same results as bypass 
grafting. Bypass grafting of supra-aortic trunk 
lesions requires the use of prosthetic material, 
often with multiple limbs, which maybe kinked 
after closure of the sternotomy wound. 

Endarterectomy has the advantage of recon- 
struction without the use of prosthetic material, 
but is technically intimidating to surgeons who 
have limited experience of operating in that part 
of the vascular system. It is essential, therefore, 
that patients are selected carefully. Anatomical 
features considered contraindications for 
endarterectomy include (1) atherosclerosis or 
calcification of the aortic arch extending into 
the origin of the innominate artery, and (2) 
close proximity of the origin of the left common 
carotid artery, which prevents safe clamping of 
the innominate artery without increasing the 
risk of embolization or of compromising cere- 
bral blood flow. Endarterectomy would not be 
an appropriate option in patients with bovine 
arches (Fig. 16.2) as cross-clamping would pre- 
dispose to inadequate left hemisphere perfu- 
sion. The San Francisco group has long been 
proponents of the endarterectomy technique 
and has partly overcome the problems imposed 
by multiple vessel disease by performing sepa- 
rate endarterectomies on each of the affected 
vessels (Azakie et al., 1998). Certain diseases 
processes (inflammatory arteritis and radiation 
arteritis) are best treated with bypass grafting, 
as the pathological process is a transmural 
process and development of an endarterectomy 
plane may be difficult. 

The emergence and technical advances in 
percutaneous angioplasty, however, have ren- 
dered many of these major reconstructions 
obsolete. Percutaneous therapy for occlusive 
lesions of supra-aortic trunk disease has several 
advantages over a surgical approach. These 
include its minimally invasive nature, avoidance 
of general anesthesia, lower cost, and, most 
importantly, the potential for lower morbidity 
and mortality. Percutaneous angioplasty for 
treatment of supra-aortic trunk lesions was 
introduced in 1980. Since that time more than 
30 small series have been published regarding 
the role of angioplasty or stenting as primary 
therapy for supra-aortic trunk lesions in more 



than 900 patients. None of the reports are ran- 
domized controlled trials (as is also the case 
with surgery), and long-term results are still not 
available. 

A comparative trial of stenting and surgical 
intervention for supra-aortic trunk lesions was 
reported in 1998 by Whitbread et al. Eighteen 
patients with symptomatic arch vessel stenosis 
or occlusion were treated with angioplasty and 
stenting, and the data compared with the pub- 
lished results following surgical procedures. The 
primary success rate was 100%, with no major 
procedural complications, and at 17-month 
follow-up all patients were asymptomatic with 
100% primary patency. The incidence of stroke 
and death was 3% and 2%, respectively. The 
authors therefore concluded that stenting 
should now be considered the first-line therapy 
for brachio cephalic obstruction. A review of the 
available literature suggests that the initial 
success rate is higher with stenoses (98%), as 
opposed to occlusions (46%). Both types of 
lesions were treated with minimal mortality 
(0.2% for stenoses and 0% for occlusions), the 
stroke rate was commendably low (0.3% for 
stenoses, 0% with occlusion), and with no 
reports of arm embolization. Recurrence of 
stenoses was similar for both stenotic lesions 
and occlusions. With a mean follow-up period, 
63 months, the incidence of recurrent stenosis 
was identical: 11% in originally stenotic lesions, 
9% for occlusions. 

Although the initial results following angio- 
plasty of supra-aortic trunk lesions are well 
documented, there is a relative lack of reports 
describing the long-term results. There are, 
however, reports of endovascular treatment of 
patients with supra-aortic trunk lesions with 9- 
and 15-year follow-up (Henry et al., 1999). These 
follow-up results are longer than most pub- 
lished surgical series. Cumulative primary 
patency was 91% to 95% at 5 years and 81% to 
84% at 10 years. Sullivan et al. (1998) prospec- 
tively studied acute and long-term results of 
angioplasty and stenting in occlusive lesions of 
the supra-aortic trunk. The initial technical 
success rate was 94%, and 80% of the technical 
failures were due to an inability to cross occlu- 
sive lesions. With initial failures included, life 
table analysis revealed 84% patency at 35 
months. None of the patients required reinter- 
vention or surgical conversion for recurrent 
symptoms. The authors concluded that angio- 



185 



ARCH VESSEL, VERTEBROBASILAR, AND UPPER EXTREMITY 





Figure 16.3. A: Severe stenosis in the right subclavian artery immediately distal to the vertebral artery (arrow). This produced arm 
symptoms only and no subclavian steal. This was successfully treated by angioplasty (B). 



plasty and stenting of the supra-aortic trunk 
vessels could be performed with relative safety 
and satisfactory midterm success. 

Endovascular treatment of supra-aortic 
trunk lesions began prior to the advent of arte- 
rial stents. Some groups have advocated the 
routine use of stents, whereas others use them 
only when angioplasty produces technically 
inadequate results. The routine use of stents 
is supported by nonrandomized evidence 
documenting improved early and long- 
term patency rates, especially when treating 
occlusions. 

One further role for endovascular interven- 
tion is in the management of the critically ill 
patient with innominate disruption or false 
aneurysm formation following trauma. The 
whole procedure can be undertaken via direct 
exposure of the right common carotid artery 
with retrograde cannulation of the carotid and 
innominate arteries. 



subclavian artery disease was only found in 
4.3%. Isolated subclavian artery stenosis (Fig. 
16.3) is most commonly found in patients with 
systemic vascular disease. The true prevalence 
of subclavian artery disease, however, is difficult 
to estimate because most patients are asympto- 
matic. Only 24% of patients with disease of the 
subclavian artery arteriography have symp- 
toms. The left subclavian artery, arising directly 
from the aortic arch, tends to be affected three 
to four times more frequently than the right. 

Other important causes of subclavian artery 
disease include aneurysm, radiation arteritis, 
Takayasu's arteritis, and the thoracic outlet syn- 
drome. Thoracic outlet syndrome is due to com- 
pression of one or more components of the 
neurovascular bundle (subclavian artery, sub- 
clavian vein, and brachial plexus) by a cervical 
rib (Fig. 16.4), fibrous band, first rib, and clavi- 
cle and anatomical anomalies of the scalene 
musculature. 



The Subclavian Artery 

Etiology 

Subclavian artery lesions are relatively uncom- 
mon, but are encountered more frequently than 
innominate or proximal common carotid artery 
lesions. The most common cause is atheroscle- 
rosis. In 4748 angiograms performed for signs 
and symptoms of ischemic cerebrovascular 
disease, the prevalence of stenosis greater than 
30% was 21%, whereas the prevalence of occlu- 
sion was 3%. Out of nearly 2000 great vessel 
reconstructions by Wylie and Effeney (1979), 




Figure 16.4. Three-dimensional magnetic resonance tomo- 
graphic image of cervical rib with connection on to first rib. 




> 



186 



VASCULAR SURGERY 



Symptoms 

As with the lower limb, symptoms can be acute 
or chronic. Thromboembolism from a subcla- 
vian artery aneurysm can cause acute upper 
limb ischemia or more focal digital ischemia. 
The latter may also originate from poststenotic 
dilatation distal to (1) an atherosclerotic proxi- 
mal subclavian stenosis or (2) a damaged artery 
in patients with thoracic outlet syndrome. The 
latter is a surgical emergency and must be rec- 
ognized and treated promptly. 

Chronic symptoms depend on the location 
of the disease. Lesions distal to the origin of the 
vertebral artery present with forearm claudica- 
tion with exercise. No neurological signs or 
symptoms are elicited from the history. Occlu- 
sive/stenotic disease proximal to the origin of 
the vertebral artery produces the subclavian 
steal syndrome. Here forearm exercise increases 
the need for increased blood flow, which is pro- 
duced via reversed flow in the ipsilateral verte- 
bral artery. 

Subclavian steal is a vertebrobasilar neuro- 
logic symptom that occurs with ipsilateral arm 
exercise. With the use of the affected arm, the 
demand for blood flow increases and the arm 
"steals" blood from the cerebral circulation 
through the ipsilateral vertebral artery. Interest- 
ingly, even if there is reversed blood flow in the 
ipsilateral vertebral artery on imaging, symp- 
toms of vertebrobasilar insufficiency may rarely 
be apparent, and the risk of posterior circula- 
tion cerebral infarction is extremely low 
(Hennerici et al., 1988) A rich collateral blood 
supply is the likely reason why symptoms may 
not become apparent. 

Patients with atherosclerotic subclavian 
artery disease tend to have a good prognosis, 
with <20% suffering significant disease pro- 
gression within 2 years of diagnosis. Moreover, 
nearly one quarter of patients who presented 
with steal (i.e., indicating marked hemody- 
namic impairment) and who were then treated 
conservatively became asymptomatic over the 
next 42 months of follow-up (Schillinter, 2002), 
indicating the potential for developing a collat- 
eral circulation in this situation. 

A second type of steal, again peculiar to the 
subclavian artery, is the coronary-subclavian 
steal syndrome. This phenomenon specifically 
occurs in patients who have had coronary 
revascularization using the internal mammary 



artery. If thereafter they should develop a severe 
stenosis or occlusion of the proximal subclavian 
artery, arm exercise can steal blood away from 
the heart. Most patients present with recurrent 
angina symptoms or cardiac failure. Treatment 
(angioplasty or carotid subclavian bypass) pro- 
duces significant improvements in blood flow to 
the heart. Clearly, cardiac surgeons should be 
aware of the potential for this problem and 
arrange for imaging of the subclavian arteries if 
there is any evidence of dizziness with arm exer- 
cise, forearm claudication, unequal blood pres- 
sures in the upper limbs, or weak radial pulses 
on palpation. Any significant lesion can there- 
after be treated (usually with angioplasty) or 
alternative conduits considered (contralateral 
internal mammary, radial artery, saphenous 
vein). 

Thoracic outlet syndrome is characterized by 
symptoms attributable to compression of the 
subclavian artery, vein, or brachial plexus. 
Arterial compression can cause claudication 
with exercise, Raynaud's phenomenon, digital 
microembolization, and even acute upper limb 
ischemia. Arterial symptoms are usually associ- 
ated with bony anomalies (e.g., cervical rib) or 
compression between the clavicle and first rib. 
Arterial symptoms secondary to thoracic outlet 
compression must be treated urgently. Com- 
pression of the subclavian vein causes heavi- 
ness/blueness of the arm and can predispose to 
subclavian vein thrombosis (Paget-Schroetter 
syndrome). Venous compression/thrombosis in 
this syndrome is often seen in patients partici- 
pating in demanding fitness regimes. The 
neurological component of thoracic outlet 
syndrome depends on whether there is com- 
pression of the lower (C8/T1) or upper (C5/C6) 
trunks of the brachial plexus. Pain or paresthe- 
sia predominate, but in severe cases muscle 
wasting can occur. 



Diagnosis 

Duplex ultrasound is now the principal imaging 
modality for examining the subclavian artery. 
Although the origin of this vessel may be 
difficult to image directly, an experienced ultra- 
sonographer will be alert to a damped inflow 
waveform suggestive of significant disease. If 
required, a corroborative or diagnostic mag- 
netic resonance angiogram (MRA) or digital 



187 



ARCH VESSEL, VERTEBROBASILAR, AND UPPER EXTREMITY 




subtraction angiography (DSA) can be 
undertaken. 

Symptoms suggestive of arterial compression 
(cold arm, fluctuating radial pulse with arm 
movements, forearm claudication, microem- 
bolization) require careful x-rays of the thoracic 
inlet to look for a cervical rib or a prominent 
transverse process on C8. The latter suggests 
that a fibrous band may be present. More 
modern tomographic imaging can demon- 
strate the size and location of any cervical rib 
(Fig. 16.4). 

Management 

Revascularization of the chronically occluded 
subclavian artery is possible with either angio- 
plasty or surgery. Surgical options include (1) 
carotid-subclavian bypass, (2) carotid-axillary 
bypass, (3) axillary-axillary bypass, and (4) 
subclavian-carotid transposition. The choice of 
technique reflects local expertise, clinician pref- 
erence, and patient presentation. 

Primary patency rates following carotid- 
subclavian bypass have been reported to be 
94%, with 87% symptom-free survival at 10 
years (Vitti et al., 1994). The principal advantage 
of subclavian transposition is that no prosthetic 
material is used. Here the proximal subclavian 
artery is mobilized, transected, and reanasto- 
mosed end-to-side onto the common carotid 
artery. It is reported to have similar outcomes to 
carotid-subclavian bypass (primary patency at 5 
years, 98%; freedom from recurrent symptoms, 
95%). However, these types of revascularization 
are not without risk: thoracic duct injury (2%), 
nerve injury (11%), stroke (6%), and wound 
hematoma (1%). The long-term durability of 
axillary- axillary bypass is inferior to carotid- 
subclavian bypass or transposition but may be 
the treatment of choice in a high-risk patient 
because it can be performed under local anes- 
thesia. For obvious reasons, this type of recon- 
struction should be avoided in any patient who 
might subsequently a median sternotomy. 

Angioplasty is the preferred option in the 
United Kingdom and most European countries. 
There is, however, no consensus as to whether 
stents should be routinely used. Angioplasty of 
the subclavian artery carries a 2% to 3% risk of 
procedural stroke. This risk can be minimized 
by passing a protective balloon up the brachial 
artery and up into the ipsilateral vertebral 



artery. This is inflated while the main subclavian 
lesion is angioplastied, and deflated once it is 
completed. It is of interest to note that following 
successful subclavian artery angioplasty, flow 
reversal in the vertebral artery is not instanta- 
neous. The reason for this delayed reversal of 
flow is not known. 

The Proximal Common 
Carotid Artery 

Etiology 

Proximal common carotid disease occurs much 
less frequently than carotid bifurcation disease, 
but 1% to 2% of patients with severe bifurcation 
disease also have significant proximal common 
carotid disease. Lesions of the right common 
carotid artery are rare in the absence of innom- 
inate artery disease and are rarer compared to 
left common carotid disease. The principal 
underlying pathologies include atherosclerotic 
disease (most common), Takayasu's arteritis, 
and, rarely, radiation-induced atherosclerosis. 

Symptoms 

Proximal common carotid lesions present with 
thromboembolic symptoms that are identical to 
those of disease at the carotid bifurcation 
(hemisensory/motor signs, temporary monocu- 
lar blindness, higher cortical dysfunction). 

Diagnosis 

The proximal common carotid artery is not 
within the scanning range of current Duplex 
technology. However, as with proximal subcla- 
vian disease, significant disease at its origin 
causes significant damping of the waveform and 
turbulence. If there is any evidence of this (or if 
the patient is being considered for bifurcation 
angioplasty), he/she will need to undergo either 
MRA or DSA. Other emerging alternatives 
include computed tomography (CT) angiogra- 
phy. Clearly the choice reflects the unit's experi- 
ence, access, and preference. 

Management 

In the past, proximal common carotid disease 
required partial sternotomy and either endar- 




> 



188 



VASCULAR SURGERY 



terectomy or bypass off the aortic arch. Other 
options included carotid-carotid bypass or 
carotid transposition. However, this is another 
situation where on-table angioplasty (following 
exposure of the mid-common carotid artery) 
affords a balance between minimal risk and 
prevention of embolization. Venting blood into 
a sheath or up the external carotid artery can 
prevent the latter. One advantage of on-table 
angioplasty is that it can be combined with 
synchronous bifurcation endarterectomy 



The Vertebral Artery 

Etiology 

The vertebral arteries arise from the proximal 
subclavian arteries. The first segment extends 
from its origin to where it enters the transverse 
cervical vertebral processes. The second seg- 
ment is intraspinal. The third segment starts at 
the upper border of the transverse process of C2 
and passes up to the atlanto-occipital mem- 
brane. The fourth segment is intradural to its 
confluence with the contralateral vertebral to 
thereafter form the basilar artery. The com- 
monest pathology is an atherosclerotic stenosis 
or occlusion of the proximal vertebral artery 
(Fig. 16.5). Alternative (less common) patholo- 
gies include fibromuscular dysplasia or 
dissection. 



Symptoms 

Classical vertebral territory symptoms (verte- 
brobasilar symptoms) include varying com- 
binations of unsteadiness of gait, diplopia, 
bilateral motor/sensory impairment, nystag- 
mus, ataxia, bilateral blindness, and dysarthria. 
These symptoms can be due to either thrombo- 
sis or embolism (often very difficult to tell), but 
the consensus view is that the majority is prob- 
ably hemodynamic. Tomographic imaging can 
sometimes help in the difficult task of discrim- 
inating between the two etiologies. 

There is much controversy about whether 
nonhemispheric symptoms (isolated diplopia, 
isolated dizziness, isolated vertigo, syncope, 
etc.) can be attributed to disease in the verte- 
brobasilar system. To date, there is no com- 
pelling evidence that they are attributable 




Figure 16.5. Occlusion of the proximal vertebral artery with 
distal refilling via collaterals. The most likely pathology is throm- 
bosis secondary to atherosclerotic disease. 



unless they definitely coexist with other, more 
typical vertebrobasilar territory symptoms. One 
other myth is the concept of "nipping of the 
intraspinal vertebral arteries causing dizziness 
on head movement." At the Leicester Royal 
Infirmary, a number of these patients have been 
examined with transcranial and extracranial 



189 



ARCH VESSEL, VERTEBROBASILAR, AND UPPER EXTREMITY 




duplex imaging, and none have ever been noted 
to have any change in flow or direction of flow 
with head movements. All were subsequently 
found to have inner ear pathologies. There is 
also no evidence that coils or kinks in the prox- 
imal vertebral artery are responsible for cere- 
brovascular symptoms unless a coexistent 
stenosis is present. 

Diagnosis 

Duplex imaging identifies evidence of damped 
flow, hypoplastic arteries (not uncommon), and 
inflow stenoses. It is not, however, particularly 
good at imaging the entire length of the artery. 
Accordingly, patients suspected of having verte- 
brobasilar insufficiency should undergo MRA 
or intraarterial DSA. 

Treatment 

The treatment of vertebral artery stenoses/ 
occlusions is to first address the anterior circu- 
lation. If there is significant internal carotid 
artery disease, carotid endarterectomy should 
be employed first. Some centers have reported 
huge series of vertebral reconstructions. 
At Leicester (apart from carotid-subclavian 
bypass), only three vertebral reconstructions 
have been performed over the last 8 years, all 
involving segment 1. Angioplasty is currently 
the emerging treatment of choice, although 
some surgeons remain strong advocates of 
surgical revascularization of segment 2 (endar- 
terectomy and patch, bypass, transposition onto 
the carotid artery). 



Technique 



All interventions should be undertaken in a spe- 
cially equipped operating room with dedicated 
fluoroscopic/angiographic capabilities. As one 
of the risks for endovascular treatment of the 
supra-aortic trunk is distal embolization, local 
infiltrative anesthesia supplemented with intra- 
venous sedation is optimal. This type of anes- 
thesia allows for continuous monitoring of the 
patient's neurologic status. 

Due to the risk of distal embolization, com- 
mon carotid lesions are best treated through a 
limited surgical incision in the neck. This allows 



for distal control of the common carotid artery 
and the ability of flushing the artery of athero- 
sclerotic emboli. Flow through the external 
carotid artery is usually sufficient for cerebral 
perfusion. If concomitant bifurcation disease 
is present (around 2% of patients), then the 
common carotid lesion should be treated first. 
Once the artery is exposed, intravenous heparin 
is given prior to obtaining proximal control, 
placement of the sheath, and crossing the lesion. 
The artery is accessed with a Potts-Cournand 
18-gauge needle in a retrograde fashion and a 
0.035-mm wire in advanced with the help of 
fluoroscopy into the aortic arch. A 35-cm-long 
sheath maybe placed to allow for more distance 
between the surgeon and radiation field, 
decreasing exposure to the surgeon's hands. 
Access of the femoral artery allows for place- 
ment of a flush catheter in the aortic arch and 
more precise localization of the ostium of the 
vessel. Selection of the innominate artery and 
selected oblique views to define the right 
common carotid and right subclavian origin is 
imperative for precise stent placement. The true 
location of the ostium is imperative to deter- 
mine optimal stent placement, with 1 to 2 mm of 
the stent protruding into the lumen of the aortic 
arch. Occlusions should be predilated to avoid 
stent dislodgment during crossing of the lesion 
or to allow for safe passage of self-expanding 
stents across the lesion. Whether balloon 
expandable stents or self-expanding stents are 
better is unknown, as there are no compara- 
tive studies. It would seem logical that if the 
lesion is tortuous or crosses the clavicle, a self- 
expanding stent, which is more flexible, would 
be preferred. Cerebral protection devices may 
change the need for surgical control of the 
common carotid artery. These devices could be 
used for stenoses, but not for occlusive lesions. 
As in internal carotid artery stenting, the lesion 
is crossed with the low-profile cerebral protec- 
tion devise prior to any manipulation of the 
lesion. This would be impossible with an occlu- 
sive lesion. Other limitations of the cerebral pro- 
tection device use would be common carotid 
size, not allowing for good apposition of the 
device to the artery. The device, therefore, 
would need to be placed in the internal carotid 
artery. 

Subclavian lesion maybe treated both from a 
femoral and brachial approach. Again, a com- 
bined brachial-femoral approach allows for 




> 



190 



VASCULAR SURGERY 



placement of a flush catheter in the aorta and 
therefore more precise placement of the stent. 
With occlusive lesion, the brachial approach 
allows for more pushability to cross the lesion. 
The caveat to this approach is risk of dissection 
into the aortic arch. In general, stenting allows 
for tacking of the dissections. Other considera- 
tions are posterior circulation embolization. It 
is therefore imperative to determine flow direc- 
tion in the vertebral artery by duplex ultra- 
sonography. Antegrade vertebral flow is delayed 
from 20 to 240 seconds, serving as an effective 
protective mechanism against embolism in the 
posterior circulation. Brachial artery cutdown 
allows for direct repair of the artery after com- 
pletion of the stenting, thus decreasing the 
risk of atherosclerotic emboli (allowing for 
flushing of the artery) and avoiding risk 
of hematoma. 



References 



Azakie A, McElhinney DB, Higashima R, Messina LM, Stoney 

RJ. (1998) Ann Surg 228:402-10. 
Berguer R, Morasch MD, Kline RA. (1998) J Vase Surg 27: 

34-41; discussion 42. 
Hennerici M, Klemm C, Rautenberg W. (1988) Neurology 

38:669-73. 
Henry M, Amor M, Henry I, Ethevenot G, Tzvetanov K, Chati 

Z. (1999) J Endovasc Surg 6:33-41. 
Kieffer E, Sabatier J, Koskas F, Bahnini A. (1995) J Vase Surg 

21:326-36; discussion 336-7. 
Rhodes JM, Cherry KJ Jr, Clark RC, et al. (2000) J Vase Surg 

31:260-9. 
Sullivan TM, Gray BH, Bacharach JM, et al. ( 1998) J Vase Surg 

28:1059-65. 
Vitti MJ, Thompson BW, Read RC, et al. (1994) J Vase Surg 

20:411-7; discussion 417-8. 
Whitbread T, Cleveland TJ, Beard JD, Gaines PA. (1998) Eur 

J Vase Endovasc Surg 15:29-35. 
Wylie EJ, Effeney DJ. (1979) Surg Clin North Am 

59:669-80. 



17 



Aneurysmal Disease 

Philip Davey and Michael G. Wyatt 




Arterial aneurysmal disease has been recog- 
nized now for over 4000 years, with the expres- 
sion "aneurysm" deriving from the Greek word 
aneurysma, meaning "a widening." Historical 
review charts a condition in which initially 
observational experiences of aneurysms have 
been gradually replaced with the established 
treatment regimes we see in clinical practice 
today. Advances made in aneurysm manage- 
ment reflect not only improved surgical acumen 
and the appliance of innovative technology but 
also the progress made in our basic scientific 
knowledge, clinical experience, diagnosis, peri- 
operative care, anesthesia, and follow-up of 
the disease. Aneurysmal disease constitutes a 
substantial component of the vascular surgeon's 
work load, and this chapter reviews the 
common pathology with reference to contem- 
porary opinion in pathogenesis, diagnosis, and 
management. 



Classification of 
Arterial Aneurysms 

The aneurysm is defined as an abnormal, 
focal dilatation of a blood vessel. An artery is 
empirically considered aneurysmal when its 
diameter becomes 50% larger than its normal 
vessel segment. This diagnosis is distinct from 
ectasia, which refers to a dilated vessel that 
has not yet reached this threshold. Similarly, 
the conditions of arteriomegaly and multiple 



aneurysms must not be confused. In the former, 
generalized dilated vasculature (>50%) is ob- 
served but, unlike the latter condition, normal- 
caliber vessel segments do not distinguish these 
conditions. 

Aneurysmal disease may be classified as 
follows: 

1. By nature: true aneurysms relate to a dis- 
tinct pathological process involving all 
three layers of the arterial wall. False or 
pseudoaneurysms do not reflect a gen- 
uine aneurysmal process at all but rather 
clinical mimicry true disease. They are, in 
fact, a vessel-associated contained blood 
collection and typically result from 
trauma. 

2. By morphology: saccular aneurysmal 
disease is present if only part of the vessel 
circumference is diseased. Entire circum- 
ferential involvement is termed fusiform. 

3. By etiology: aneurysms are frequently 
referred to with respect to an antecedent 
disease process. The list, which is not 
exhaustive, includes 

a. Degeneration (e.g., atherosclerosis, 
fibromuscular dysplasia) 

b. Infection (e.g., syphilis, bacterial, 
fungal) 

c. Trauma (e.g., iatrogenic, penetrating, 
blunt) 

d. Inflammation (e.g., Takayasu's or 
Kawasaki disease, polyarteritis nodosa) 



191 




> 



192 



VASCULAR SURGERY 



e. Connective tissue disorders (e.g., 
Marfan syndrome, Ehlers-Danlos 
syndrome) 

f. Congenital (e.g., tuberous sclerosis, 
Turner syndrome) 

4. By location: for example, abdominal aortic, 
popliteal, femoral, subclavian, thoracoab- 
dominal, etc. 

Pathogenesis of 
Aneurysmal Disease 

Despite extensive research, the exact mecha- 
nisms underlying aneurysm pathogenesis 
remain unclear. It appears the process is multi- 
factorial, involving variable components of 
altered biochemistry, immunological, mechani- 
cal, and genetic importance (Wassef et al.,2001). 
Much of our understanding of the disease 
relates to work performed on the abdominal 
aorta, and it is assumed that nonaortic aneurys- 
mal disease results from a similar pathological 
sequence of events. 

Aneurysmal disease ultimately results follow- 
ing gradual proteolytic degradation of arterial 
vessel walls. Structural integrity of arterial wall 
is dependent on adequate functional connective 
tissue elements including elastin and collagen. 
Loss of these elements weakens the vessel wall 
and thus predisposes to aneurysm formation. 
Research into abdominal aortic aneurysms 
(AAAs) has discovered local increased levels of 
matrix metalloproteinases (MMPs) that result in 
this observed connective tissue breakdown. 
Four MMP subtypes are thought to be of impor- 
tance in this process: gelatinases (MMP-2 and 
MMP-9), matrilysin (MMP-7), and macrophage 
elastase (MMP-12). In addition to this increased 
local expression, altered levels of circulating 
proteases may play a role, and research con- 
tinues into the contributions to aneurysmal 
disease from various plasminogen activators, 
serine elastases, and cysteine proteases (cathep- 
sins S and K). Abnormally low levels of protease 
inhibitors are suggested to exert the same path- 
ogenic effects, and this has also been the focus 
of much recent work. Preliminary research into 
the most abundant serine protease inhibitor, 
cystatin c, has confirmed that in patients with 
aneurysmal disease corresponding levels of this 
protein are indeed lower than normal. 



Chronic inflammation plays an important 
role in aneurysmal disease. Much of the vessel 
wall destruction is undoubtedly mediated by the 
inflammatory infiltrate composed of T cells, 
macrophages, B lymphocytes, and plasma cells, 
but the antecedent trigger for this cellular 
migration remains unclear. It has been sug- 
gested that aneurysmal disease is in fact an 
antigen-driven immune disease from work 
analyzing AAA disease. Proposed antigenic 
activators to subsequent inflammation include 
elastin, interstitial collagen, oxidized low- 
density lipoprotein, cytomegalovirus, and 
artery-specific antigenic proteins such as 
AAAP-40. Following T-cell antigen recognition, 
the inflammatory cascade begins, ultimately 
resulting in vessel wall degradation and pro- 
gression to aneurysmal disease. 

Arterial wall biomechanical stress is also 
accepted as being of considerable importance 
in both aneurysm progression and rupture. 
Model analysis of wall stress variation in differ- 
ent anatomical locations implies that altered 
hemodynamic profiles may explain the varying 
susceptibility of an arterial wall to become 
aneurysmal (e.g., increased disease incidence 
in abdominal as opposed to thoracic aorta). 
It is suggested that mechanical failure from 
excessive stress initiates and promotes ane- 
urysmal pathogenesis first by the aggregation 
of humoral factors, and then by a consequent 
focal inflammatory response and finally wall 
breakdown. 

The familial clustering of aneurysmal 
disease has led to interest in determining a 
possible genetic explanation for the process. 
Supporting evidence is derived from observa- 
tions regarding the altered disease charac- 
teristics of "familial" and "spontaneous" 
aortic aneurysmal disease. Cases of familial 
AAA present significantly younger, carry a 
greater rupture risk (especially in females) 
and are 18 times more likely to be present in 
other family members than is seen with sponta- 
neous pathology. Affected sibling pair DNA 
linkage studies aims to identify the putative 
AAA-susceptibility gene, whose expression or 
mutation promotes progression to aneurysmal 
disease. This approach to pathogenesis may 
eventually form the basis of genetic testing 
of specific increased-risk populations, 
allowing more focused surveillance and early 
intervention. 



193 



ANEURYSMAL DISEASE 




Abdominal Aortic Aneurysms 

In the 16th century the anatomist Vesalius first 
recognized aneurysmal disease affecting the 
infrarenal aorta. Limited progress was made 
over the following centuries in the treatment of 
AAA, and it was not until 1952 that Dubost 
reported the first successful treatment of the 
condition by surgical repair. In the past 50 years, 
increased experience and advancement of sur- 
gical techniques have rendered elective AAA 
repair a routine albeit major operative proce- 
dure with expected mortality rates of less than 
5% (Hallin et al., 2001). Exciting developments 
in the endovascular arena over the last decade 
have offered further options for treatment in 
elective AAA disease and promises to revolu- 
tionize management for many patients. 

The natural history of AAA is to gradually 
expand and eventually rupture. Ultimately, the 
aim of elective surgery is to prevent this from 
happening by excluding the aneurysm from the 
circulation by means of either prosthetic graft 
insertion or endovascular stent deployment. 
Unfortunately, even with improved periopera- 
tive management and operative technique, the 
great advances made in elective AAA repair 
have not been witnessed in the emergency 
setting for AAA rupture, and consequently mor- 
tality rates remain high at approximately 50%. 

Definition 

Aneurysms are arbitrarily defined as a segment 
of vessel dilatation that is at least 50% larger 
than the expected normal vessel diameter. In 
practical terms this diameter refers to the nona- 
neurysmal adjacent vessel segment. We accept a 
"normal" abdominal aorta to be approximately 
20 mm in diameter, and therefore an aorta wider 
than 30 mm would suggest aneurysmal disease. 

Epidemiology 

In all epidemiological analysis, statistical varia- 
tion can exist as a result of inconsistent 
definitions of disease, and AAA is no exception. 
Population screening surveys and postmortem 
studies have been used to estimate a current 
prevalence of AAA of between 1.3% and 12.7% 
in England (Wilmink and Quick, 1998). Marked 
increases in the prevalence of the disease are 



seen if the definition of AAA is relaxed to even 
1 mm less than stated earlier (30mm), but little 
difference exists between the two methods of 
prevalence estimation in this condition. 

The incidence of asymptomatic AAA has 
been reported at 3 to 117 per 100,000 person- 
years. These estimations are derived from the 
number of hospital admissions for elective 
asymptomatic AAA repair and have been 
observed to increase by 7% to 26% over the past 
15 years. This basically reflects changes in 
improved disease awareness, referral patterns, 
and diagnosis, more specifically the increased 
availability and use of ultrasound scanning in 
recent years. The reported incidence of rup- 
tured AAA is much more inaccurate due to high 
prehospital mortality, but quoted values are 
between 1 and 21 per 100-000 person-years. 
Data also suggests that this value is continuing 
to increase by 2.4% per year. In the United 
Kingdom, ruptured aortic aneurysms have been 
reported as the 13th commonest cause of death 
accounting for 1.2% of male and 0.6% of female 
mortality. 

Abdominal aortic aneurysm is typically a 
disease of the male population, being five times 
more common than in women. A sharp 
increased incidence is seen after the age of 50 
years, rising to apeak at 80 years of age. Abdom- 
inal aortic aneurysm in females does not tend 
to occur until about 10 years later than in men, 
and then increases linearly from the age of 60. 

In a more general sense, 5% of the over-60 
population will have an AAA, and at the age of 
67 a man is ten times more likely to die from a 
ruptured AAA than is a woman. 

Risk Factors for Abdominal 
Aortic Aneurysm 

Factors associated with an increased risk of 
aneurysmal development in the infrarenal aorta 
include increasing age, male sex, ethnic origin, 
family history, smoking, hypercholesterolemia, 
hypertension, and prior vascular disease. 
Studies suggest that of these, male sex and 
smoking are the most important, increasing 
the chances of AAA development by 4.5 and 5.5 
times, respectively. A positive family history 
(first-degree relative affected) alone doubles the 
risk of AAA presentation and is more likely if 
the affected relative is a female. Interestingly, 




> 



194 



VASCULAR SURGERY 



diabetes mellitus, which is a major risk factor 
for occlusive arterial disease, is associated with 
less risk (compared to nondiabetics) in the 
development of aortic aneurysmal disease. 

Clinical Features 

The vast majority of AAAs are asymptomatic, 
and the diagnosis is usually made on clinical 
abdominal examination or following ultra- 
sound investigation for other pathology. If 
symptoms are present, they usually relate to 
the complications of infra-aortic aneurysmal 
disease both locally and systemically 

Abdominal or back pain may be experienced, 
and this must alert the clinician to the possibil- 
ity of posterior erosion of the aneurysm into the 
neighboring vertebral bodies. Classically in this 
situation, the history of pain follows a non- 
specific and indolent course. Appropriate subse- 
quent investigations should distinguish erosion 
from the diagnosis of inflammatory AAA (see 
later). Sudden onset of severe abdominal or 
back pain in a patient with a known AAA 
who is hemo dynamically unstable suggests 
aneurysm rupture. The pain may radiate into 
the hips, and the patient will be pale, anxious, 
clammy, tachycardic, and hypotensive. An 
aneurysm rupture that is contained within the 
retroperitoneum is often seen with a similar but 
less pronounced symptom profile, affording 
more time for preoperative optimization than 
uncontained ruptures. 

Symptoms of distal ischemia may result 
directly from the presence of AAAs. Emboliza- 
tion of contents from the aneurysmal sac may 
present with acute or chronic limb ischemia, 
whereas a similar picture may be witnessed if 
the AAA thromboses in situ. In the latter case 
one would expect symptoms of bilateral leg 
ischemia. 

Less common presentations of infra-renal 
aortic aneurysmal disease relate to fistulation. 
Cases of aortoduodenal fistulas present classi- 
cally with intermittent, unexplained gastroin- 
testinal bleeding and melena or massive 
uncontrollable hematemesis. The diagnosis is 
confirmed by upper gastrointestinal endoscopy 
prompting urgent surgical repair. Isolated aor- 
tocaval fistulas are rare but present with symp- 
toms of high output cardiac failure. More 
commonly, an aortocaval fistula results as a con- 
sequence of aneurysm rupture, and the opera- 



tive finding at laparotomy is that of massive 
venous hemorrhage requiring rapid control to 
avert on-table death due to exsanguination. 

Detection of an AAA can potentially be made 
by careful abdominal palpation during clinical 
examination. The reliability of this technique 
alone is questionable, its sensitivity governed by 
a variety of factors such as aneurysm size, exam- 
iner's skill, patient's body habitus, and the 
purpose of the clinical examination itself. Even 
after acknowledgment of these variables, a 
correct diagnosis is achieved in only approxi- 
mately 50% of cases. Smaller aneurysms and 
obesity increase the likelihood of a missed 
diagnosis, whereas an false-positive diagnosis of 
AAA may be made in thin patients, tortuous 
aortas, or in those with more prominent vessels 
as a consequence of lumbar lordosis. Similarly, 
the success of manual examination in the 
assessment of aneurysm size and proximal 
extent in relation to the renal arteries is poor. 
Subcutaneous fat and overlying intestine tend to 
cause an overestimation of size, and the ability 
to admit an examining hand between the costal 
margin and pulsatile mass will not indicate 
infrarenal disease in all cases. 



Investigations for Abdominal 
Aortic Aneurysm 

Plain Film Radiography 

Plain abdominal x-rays are cheap and widely 
available but of little use in the routine diagno- 
sis of the AAA. Occasionally, calcification may 
be observed in the wall of the aneurysmal vessel 
or there may be loss of the psoas major shadow. 
In the preoperative setting their use is limited 
to exclusion of an alternate diagnosis to AAA. 
They do, however, play an important role fol- 
lowing endovascular stent graft repair of AAA 
for observation of long-term outcome and graft 
integrity. 



Ultrasonography 

Abdominal ultrasound examination is responsi- 
ble for the detection of the majority of asymp- 
tomatic AAAs and it is also used to confirm 
the suspected clinical diagnosis (Fig. 17.1). In 
addition, this modality is employed to observe 
aneurysmal expansion in patients with disease 



195 



ANEURYSMAL DISEASE 




L«08. 6 JJg 


*■**> 


■ 

i 





Figure 17.1. An abdominal ultrasound showing an aortic 
aneurysm. 



under surveillance. Ultrasound is cheap, rela- 
tively quick, widely available, and noninvasive. 
Visualization of the aorta is achieved in 97% 
of patients, and current figures demonstrate 
minimal interobserver variability. The avail- 
ability of mobile ultrasound machines allows 
assessment of the critically ill patient who 
ideally should not be transferred to the radiol- 
ogy department. 

Ultrasound, however, does have its limita- 
tions. Unfortunately, cases of suprarenal, vis- 
ceral artery aneurysmal disease and iliac vessel 
extension cannot be accurately assessed by 
ultrasound. In obese, ventilated patients, or 
those patients with excessive bowel gas, this 
unreliability is further exaggerated. Further- 
more, this modality is poor at confirming the 
presence of aneurysm rupture and hence plays 
no role in the emergency setting. 

Computed Tomography Scanning 

Computed tomography (CT) scanning remains 
the investigation of choice for the most accurate 
assessment of AAA disease. The procedure 
allows visualization of the upper extent of the 
aneurysm and its nature (i.e., noninflammatory 
or inflammatory), and provides reliable infor- 
mation regarding iliac artery involvement. 
Interobserver variation in the measurement of 
aneurysm size is superior to ultrasound, with 
over 90% of studies exhibiting a discrepancy 
of less than 5 mm. Computed tomography 
scanning with 3-mm cuts can also be used to 
determine if the patient is a candidate for 



endoluminal stent grafting. The imaging, 
however, is invasive, time-consuming, and 
expensive, and it entails substantial radiation 
exposure for the patient. It is therefore restricted 
for preoperative assessment in those suspected 
to be imminent candidates for surgical repair, 
follow-up of endovascular stent graft repairs, 
and for patients with suspected AAA rupture 
who are stable enough for deferment of 
laparotomy. 

The advent of spiral CT scanning in some 
centers has led to increased speed of assess- 
ment (a single breath hold) with the ability to 
reconstruct CT images in a three-dimensional 
manner, resulting in a more user-friendly and 
informative imaging. The use of CT scanning 
must be judicious in patients with a prior 
history of contrast reaction, high blood pres- 
sure, and preexisting renal impairment. As with 
ultrasound, CT scanning cannot detect visceral 
artery involvement or the presence of renal 
artery stenosis, and this must be remembered in 
assessment of the generated images. 

Magnetic Resonance Imaging 

Magnetic resonance imaging (MRI) for AAA 
offers comparable results to CT scanning in 
terms of assessment of aneurysm size, proximal 
extent, iliac extension, and etiology (e.g., 
inflammatory), and has the added advantage of 
being noninvasive and safe because no contrast 
administration is required. The procedure, 
however, is not well tolerated by claustrophobic 
patients, and its limited availability and expense 
have relegated its use to a second-line investi- 
gation for AAA, reserved for those patients for 
whom CT scanning is inappropriate (e.g., 
iodinated contrast allergy, chronic renal failure, 
or claustrophobia). 

Angiography 

Contrast arteriography offers little information 
regarding the possible diagnosis or sizing of an 
AAA, as it is only the vessel lumen (not the 
aneurysmal sac) that is delineated in this exam- 
ination. For these reasons, this investigation 
plays little role in the management of the 
routine infrarenal AAA. It does, however, offer 
detailed information with respect to possible 
visceral or renal artery disease, and allows ade- 
quate vessel anatomy analysis for planning prior 




> 



196 



VASCULAR SURGERY 



to surgical repair. Indications for arteriography, 
therefore, include aneurysms with possible 
suprarenal involvement or suspected visceral 
artery disease, renal stenosis, and in patients 
with coexistent peripheral vascular or nonaor- 
tic aneurysmal disease (e.g., popliteal). Again, 
the investigation is invasive, involves the use of 
substantial amounts of contrast agent, and 
carries with it the risks of embolic and renal 
complications. 

Screening for Abdominal 
Aortic Aneurysm 

There is currently much debate about the merits 
about introducing a national screening program 
for the detection of aneurysmal disease affect- 
ing the abdominal aorta (Table 17.1). The nature 
of AAA is such that the majority of patients have 
the condition silently, until they present at the 
hospital under the most extreme circumstances: 
aneurysm rupture. Because the prevalence of 
this disease is relatively high (5% of the over-60 
population), it has been suggested that a simple 
ultrasound-screening test at about this age 
would uncover the asymptomatic pathology at 
an early stage. Appropriate follow-up and elec- 
tive repair could then be planned, thus dimin- 
ishing the frequency of ruptured aneurysm 
presentation and its inherent cost implications. 
Recent studies in the U.K. have shown that a 
screening program would indeed fulfill these 
expectations, observing a reduction in the AAA 
rupture rate by 50%. Other groups suggest a 
screening program would have a greater diag- 
nostic yield and hence prove more cost-effective 
if only the specific, high-risk populations were 
targeted, such as elderly male smokers. What- 
ever the proposal, it would certainly appear that 
a timely ultrasound examination may be 
beneficial in the early management of AAA, but 
a large multicenter trial is needed to obtain 



Table 1 7.1 . Why screen for aneurysms? 

Abdominal aortic aneurysms (AAA) are asymptomatic 

Major complication of AAA is rupture 

Morbidity and mortality of repair of ruptured AAAs: 

50% to 70%; elective AAAs, 1% to 5% 
Patients who survive repair have a similar life 

expectancy as the general population 
Ultrasound is inexpensive and noninvasive 



adequate numbers of patients with the power 
to demonstrate the cost-effectiveness of this 
approach to AAA disease. 



Management of the Elective 
Abdominal Aortic Aneurysm 

The basic goal underpinning the management 
of the asymptomatic and symptomatic non- 
ruptured AAA is to avert the presentation of 
aneurysm rupture. As with all surgery, a surgeon 
must balance the risks of the required major 
operation in terms of mortality and morbidity 
against that of a possible rupture-related death 
with nonoperative "conservative" management. 
With appropriate case selection and surgical 
expertise, mortality following elective AAA 
repair should be between 2% and 5%. Important 
issues to be taken into account, which enable the 
surgeon to make the correct decision whether or 
not to operate, are discussed in the following 
subsections. 



Aneurysmal Size 

The maximum diameter of AAA is without 
doubt the most important factor in predicting 
the ruptured risk of an aneurysm. The 5-year 
rupture risk of an aneurysm less than 4 cm in 
diameter is approximately 15%, and this rises to 
nearly 95% if the aneurysm grows to 7 cm in 
diameter. Unfortunately, vascular surgery is not 
an exact science, however, and small aneurysms 
will continue to rupture and many people with 
large aneurysms will outlive the disease and die 
of an unrelated cause. 

The recently completed U.K. small aneurysm 
trial aimed to provide guidelines regarding 
when to offer elective surgery on the basis of 
aneurysm size (Lancet, 1998). Comparison of 
surgical repair of small aneurysms (4 to 5.5 cm) 
with nonoperative ultrasound surveillance sug- 
gested that there was no survival benefit to be 
gained from operative management of the AAA. 
The trial researchers recommended that 
aneurysms should be considered for surgical 
repair when the maximal diameter exceeded 
5.5 cm or the aneurysm became symptomatic or 
tender. It was at this stage the operative risk of 
repair was less than the risk of rupture associ- 
ated with nonoperative management. 



197 



ANEURYSMAL DISEASE 




Treatment protocols for small aneurysms 
remain less clear-cut, but as a general rule all 
AAAs greater than 30 mm in diameter should be 
kept under ultrasound surveillance. Aortas less 
than 40 mm in diameter should probably be 
monitored with annual ultrasound scans, 
whereas if there is growth to between 40 and 55 
mm the frequency of observations should 
increase to every 6 months. Other measures in 
management include encouragement to stop 
smoking, aggressive blood pressure control, 
and possibly the administration of propranolol 
unless contraindicated (see later). If an 
aneurysm under surveillance appears greater 
than 50 mm in diameter, a CT scan should be 
obtained to formally size and delineate aneurys- 
mal anatomy in preparation for operative repair. 
At some centers, it is at this stage that the patient 
may be considered for endovascular treatment 
as opposed to open repair of their aneurysm. 

Expansion Rate 

Aneurysmal expansion rate is considered an 
important factor as it enables the clinician to 
estimate the timing of elective surgery governed 
by the size of the AAA. Much evidence suggests 
that the larger the aneurysm, the quicker the 
expansion rate, so that aneurysmal growth 
can be considered as exponential function of 
aneurysm size. Small aneurysms between 40 
and 60 mm are expected to expand "normally" 
at an annual rate of around 10% (4 to 5 mm). 
Growth patterns that exceed this reflect unpre- 
dictable pathology and may warrant earlier sur- 
gical intervention. It is generally accepted that if 
the aneurysm expansion rate is greater than 10 
mm, this is an indication for operative repair, 
regardless of the aneurysm size. 

The exact reasons for the vast differences wit- 
nessed in aneurysm expansion remain unclear. 
Studies have shown that smoking, advancing 
age, cardiac disease, and hypertension are all 
independent predictors of an increased expan- 
sion rate, and that administration of certain 
beta-blocker drugs may reduce this phenome- 
non. Recently, much interest has been directed 
toward the medical management of small 
aneurysms with the drug propranolol, which is 
observed to reduce the rapid expansion of AAA 
by nearly 40%. The exact mechanism of this 
finding could simply be explained by improved 
blood pressure or heart rate control, but some 



groups suggest that since propranolol blocks 
circulating tissue plasminogen activator (tPA), 
there is a direct effect on the aortic wall by 
diminished plasmin-mediated MMP activation 
and therefore reduced expansion rate. Whatever 
the pharmacological explanation, it would cer- 
tainly appear that unless contraindicated (e.g., 
asthma, severe cardiac failure), a good argument 
can be made for the administration of propra- 
nolol while the aneurysm is under surveillance. 

Proximal Extent of Abdominal 
Aortic Aneurysm 

The relationship of the aneurysm to both the 
renal and visceral arteries must be elicited prior 
to surgical repair. Juxtarenal aneurysms com- 
mence immediately below the renal arteries 
and thus do not permit a clamp to be placed 
infrarenally, as is also the case with suprarenal 
aneurysms, which may or may not involve the 
superior mesenteric artery or coeliac axis. If the 
pathology commences higher than the coeliac 
trunk, the aneurysm is deemed to be thora- 
coabdominal. Repair of all these aneurysms 
carries significantly increased patient mortality 
or morbidity with requirement for higher levels 
of surgical expertise. Hence, referral to a terti- 
ary vascular center is mandatory and repair 
should not be attempted at a district general 
hospital. 

Patient Comorbidity 

Full patient assessment prior to proposed AAA 
surgery should aim to identify medical factors 
to be addressed in the preoperative workup. 
Aneurysmal surgery is obviously not appropri- 
ate in those patients with widespread advanced 
malignancy and limited life expectancy or those 
with neurological degenerative disease. Other 
conditions that do not preclude surgery should 
be appropriately treated in order to optimize the 
patient for the operating room and so minimize 
the risks of aneurysm repair. Specific preopera- 
tive conditions associated with poor outcome 
following elective AAA surgery include renal 
dysfunction, cardiac failure, myocardial 
ischemia, and respiratory impairment. The 
presence of one or more of these conditions may 
indicate the need for extensive preoperative 
investigation and appropriate referral to other 
medical specialties. 




» 



198 



VASCULAR SURGERY 



Individual Surgeon's Audited Results 

In the current climate of clinical governance 
and accountability, it is no surprise that the 
decision to operate in AAA does not entirely 
depend on the patient but also on the ability of 
the operating surgeons themselves. Advances in 
operative technique and vascular anesthesia in 
major centers have been reflected in a fall of 
operative mortality following elective AAA 
repair to around 2% to 5%. Unfortunately, the 
death rate at some district general hospitals 
after the same operation by personnel without 
specialist knowledge has been reported in 
excess of 10% and poses the question of whether 
this surgeon-specific discrepancy warrants the 
shift of all elective AAA repairs to be performed 
at recognized major vascular centers. 

Aneurysmal Symptomatology 

Symptomatic and suspected ruptured aortic 
aneurysms are usually in themselves an indica- 
tion for surgical intervention. The rationale for 
this decision is explained by the high mortality 
rate of a presumed imminent rupture, and pre- 
dicted morbidity following embolism from the 
aneurysmal sac. 

Preoperative Assessment of the 
Elective Abdominal Aortic Aneurysm 

One of the cornerstones of the successful man- 
agement of the elective AAA is adequate preop- 
erative preparation. An extensive patient history 
and full examination should be performed, 
followed by a set of basic investigations for all 
patients. These measures should aid the deci- 
sion of whether or not operative intervention 
is appropriate and may uncover any subclinical 
pathologies that can be corrected, or at least 
optimized prior to surgery. We consider the 
required analysis as of two distinct types, exam- 
ining both general health status with quality of 
life and also aneurysm-specific factors. 

General Health Status 

After initial assessment of an apparently med- 
ically fit patient, it is essential to gain an appre- 
ciation of that person's quality of life. This loose 
term is difficult to define but encapsulates all 
aspects of the patient's daily living, involving 



not only physical status but mental and social 
aspects also. Operation for AAA should both 
prolong and preserve quality of life. Due to the 
nature of the disease, it is unlikely that AAA 
repair will lead to any physical improvement in 
the preoperative condition with the risk of a 
drastic deterioration in health. On the other 
hand, for many, quality of life is still vastly 
improved by elimination of the cause of much 
psychological torment that was the "time bomb" 
ticking in their abdomen. There is little doubt 
that issues pertaining to a subject's quality of life 
can raise difficult and emotive questions. The 
astute clinician should enlist help with manage- 
ment decisions from not only the patient but 
also the family, nursing staff, and caregivers. 

Basic laboratory investigations to be per- 
formed on all patients considered for AAA 
surgery include a full blood count, erythrocyte 
sedimentation rate (ESR), blood glucose, serum 
urea, and electrolytes. Hematological disorders 
and anemia may well be uncovered at this stage, 
prompting further investigation before surgery. 
An elevated ESR might be the only indicator of 
an inflammatory AAA or even reflect ongoing 
chronic infection or inflammation elsewhere. 
Hyperglycemia may expose the unknown dia- 
betic and has implications for both peri- and 
postoperative patient care. Silent renal or 
endocrine dysfunction is suggested with abnor- 
mal urea and electrolyte values, and these would 
necessitate further investigation and optimiza- 
tion before surgery. 

In addition to these tests a 12-lead electro- 
cardiogram (ECG), chest x-ray, arterial blood 
gas analysis, and pulmonary function tests 
should be performed. These reflect the impor- 
tance of adequate cardiorespiratory reserve 
on the successful outcome of AAA surgery. 
Depending on the results obtained, it may well 
be the case that further investigations such as an 
exercise ECG, echocardiography, and even coro- 
nary angiography should be performed before 
surgery. The presence of significant cardiac 
disease may indeed require significant inter- 
vention such as angioplasty, stenting, or even 
bypass grafting before the patient is considered 
fit enough elective AAA repair. 

Aneurysm-Specific Factors 

It is most likely that by this stage an ultrasound 
scan will have been performed confirming the 



199 



ANEURYSMAL DISEASE 




presence of an AAA. Unless contraindicated, the 
aneurysm should now be imaged by means of 
CT scanning for accurate sizing, proximal and 
distal extension, relationships to other abdomi- 
nal vasculature, and possibly an assessment for 
endovascular stenting. In those patients where 
CT scanning is not an option, MRI offers an 
alternative method of gaining the required pre- 
operative information. 

In the majority of cases, this constitutes all 
that is required radiologically to appropriate 
plan operative intervention. However, in 
juxtarenal or suprarenal aneurysms, or patients 
with peripheral vascular disease or aneurysmal 
disease elsewhere, angiography should be 
considered. 

Operative Management 
of the Elective Abdominal 
Aortic Aneurysm 

Assuming the patient workup is completed and 
the patient is suitably optimized, elective AAA 
repair can now be attempted. Broadly speaking, 
there are two techniques used for aneurysm 
repair, and consideration will be given to each 
in turn. 

Open Repair of Elective Abdominal 
Aortic Aneurysm 

It is now over half a century since the first 
reported successful open repair of an AAA, and 
the technique remains well established today. 
Informed patient consent for the procedure is 
obtained and adequate (8 to 10 units) type- 
specific blood is cross-matched before surgery. 
Invasive monitoring is mandatory in AAA 
repair, requiring insertion of a urinary catheter 
and also arterial and central venous cannula- 
tion. A nasogastric tube is inserted for stomach 
decompression, and the anesthetist may decide 
to float a pulmonary artery catheter (Swan- 
Ganz) in those patients with significant cardiac 
disease. It is at this stage that an epidural 
catheter maybe inserted to minimize postoper- 
ative pain. A single bolus dose of a wide- 
spectrum antibiotic (e.g., cefuroxime 1.5 g) is 
usually given to reduce the risk of prosthetic 
graft infection and this dose maybe repeated in 
long operative procedures. The general anes- 
thetic is administered and the airway is secured 



by endotracheal intubation so that artificial ven- 
tilation can commence. Once the vascular anes- 
thetist is satisfied, the stable patient is then 
transferred into the main operating room. 

Elective AAA repair may proceed by either a 
transabdominal or retroperitoneal approach as 
long as adequate exposure to gain proximal and 
distal control is achieved. The patient is thus 
appropriately positioned, draped, and prepped. 
A relative indication for retroperitoneal access 
is the case of a "hostile" abdomen either due to 
adhesions from a previous laparotomy or the 
presence of an abdominal wall stoma. In the 
majority of elective repairs, however, it is a stan- 
dard transperitoneal approach that is preferred. 
Laparotomy is performed by standard full- 
length midline incision offering a generous 
exposure, although some surgeons advocate a 
transverse incision in cases with no iliac 
aneurysmal involvement. Advantages of the 
latter are a better cosmesis and a decreased inci- 
dence of respiratory complications postopera- 
tively as a result of reduced wound pain. 
Long-term wound disorders, however, are seen 
more frequently with a transverse incision. 

Following skin incision the peritoneum is 
divided from a point just left to the base of the 
small bowel mesentery extending distally as far 
as the iliac artery. A self-retaining retractor is 
then positioned so that the surgeon has ade- 
quate exposure of the aneurysm, with the small 
bowel usually "stored" in a sterile bag for pro- 
tection and facilitation of access. The neck of the 
aneurysm is dissected free followed by each of 
the iliac arteries prior to application of vascular 
clamps. Intravenous heparin is given just before 
aortic cross-clamping in order to reduce the risk 
of thrombotic complications, and this dose may 
be repeated in prolonged procedures. 

After allowing enough time for systemic anti- 
coagulation, the clamps are applied in a manner 
so that the possibility of distal complications are 
minimized. In practice this requires clamping of 
the least-diseased vessels first, taking care to 
avoid vessel damage and mural plaque disrup- 
tion. The proximal clamp should be as close to 
the renal arteries as possible so that the resid- 
ual native infrarenal aortic segment is minimal, 
therefore limiting the risk of subsequent 
aneurysmal recurrence. Once proximal and 
distal control has been achieved, the sac of the 
aneurysm is incised longitudinally along its 
anterior surface, taking care not to damage the 




> 



200 



VASCULAR SURGERY 



inferior mesenteric artery (IMA) that arises just 
to the left of midline from the aorta. Problem- 
atic bleeding may be encountered at this stage 
from patent lumbar arteries, and these are over- 
sewn by vascular sutures. Back-bleeding from 
the IMA should be temporarily controlled with 
a small vascular clamp (e.g., bulldog clamp) 
during aneurysm repair, and the need for pos- 
sible reimplantation should be assessed later in 
the operation. 

An appraisal can now be made regarding the 
type of graft to be used and appropriate sizing 
performed. In 60% of cases a straight "tube" 
synthetic inlay graft may be used, requiring 
aorto-aortic anastomosis. The remainder of 
cases require a bifurcated graft due to either 
iliac extension of aneurysmal disease or con- 
comitant occlusive pathology. In this situation 
the distal anastomosis should be performed into 
a normal vessel, distal to the affected artery. This 
usually requires iliac anastomosis, but if there 
is extensive iliac artery disease or technical 
difficulties, a femoral anastomosis must be con- 
sidered. It should be remembered in the latter 
scenario that the patient will be at an increased 
risk of infection and false aneurysm formation 
as a result of opening the groin. The graft is 
sutured with 2-0 or 3-0 nonabsorbable stitches, 
ensuring to take deep-enough bites in the prox- 
imal anastomosis for adequate strength. In cases 
of friable aortic tissue, the surgeon may wish to 
consider the use of pledgets in the suture line in 
order to reinforce the anastomosis. Integrity 
of the proximal anastomosis is confirmed by 
careful release of the proximal clamp against 
resistance offered by prior application of a 
clamp across the graft. Minimal bleeding sug- 
gests successful anastomosis, and the proximal 
clamp is reapplied to its original position so 
that attention can now be given to the run-off 
vessel(s). 

Distal anastomosis should be to the appro- 
priate vessel as outlined earlier, with the aim to 
preserve at least internal iliac artery for mainte- 
nance for colonic and neural blood supply. After 
completion of the first iliac (or distal aortic) 
anastomosis, blood flow into the supplied limb 
should be restored after confirmation of anas- 
tomotic integrity. Close lesion with the anes- 
thetic team is required at this stage, and release 
of the distal clamp should be very gradual. 
Sudden clamp release could lead to precipitous 
falls in blood pressure ("declamping hypoten- 



sion") as a result of rapid volume redistribution 
into the large dilated distal ischemic vascular 
beds. This phenomenon is further exaggerated 
by systemic release of accumulated vasoactive 
metabolites from the clamped limb, and there- 
fore timely intravenous fluid "loading" is neces- 
sary following surgical alert before clamp 
release. 

With the graft in situ and hemostasis 
ensured, attention returns to the IMA. In the 
majority of cases the IMA maybe simply ligated 
with a vascular suture. This practice, however, 
requires at least one patent internal iliac artery, 
a disease-free superior mesenteric artery, pink 
healthy large bowel, and finally good back- 
bleeding on release of the bulldog clamp. If 
these criteria are not met or colonic perfusion 
appears suboptimal at the end of the procedure, 
the IMA may have to be reimplanted into the 
graft by means of a Carrel patch. 

Assuming IMA reimplantation is not neces- 
sary, the native aneurysm sac is closed over the 
graft and loosely sutured, providing physical 
graft protection from other intraabdominal 
structures. The bowel is released from the tem- 
porary storage bag and a final examination is 
made to ensure adequate organ perfusion and 
also to allow correction of any iatrogenic injury. 
The patient's limbs are also assessed to ensure 
adequate distal perfusion and exclude immedi- 
ate graft thrombosis or embolism that would 
require prompt surgical correction. The self- 
retaining retractor is removed and the abdomen 
closed in routine fashion. 

Following the operation the patient should be 
transferred to an intensive care or high depend- 
ency unit for close monitoring. Return to the 
ward is usually possible the following day if 
there have been no complications. 

Discharge following elective AAA repair is 
variable but can be expected at between 7 and 
14 days depending on preoperative comorbidity, 
operative difficulty, patient determination, and 
postoperative complications. 

Complications Following Open 
Abdominal Aortic Aneurysm Repair 

Despite extensive preoperative workup and 
improvements in anesthesia and surgical tech- 
nique, complications following elective open 
repair of AAA still occur. All possible measures 



201 



ANEURYSMAL DISEASE 




must be taken to minimize them, as often mor- 
bidity following aneurysm repair can translate 
to mortality. An acceptable mortality rate of less 
than 5% justifies prophylactic AAA repair in 
appropriate aneurysms as compared to the risk 
of rupture and subsequent near- 100% mortality 
with a conservative approach. The nonfatal 
complications are considered as early or late. 

Early Complications 

Cardiorespiratory 

Cardiorespiratory complications account for 
25% to 30% of all morbidity following elective 
open AAA repair. The majority of cardiac 
ischemic events take place in the initial 48 hours 
postoperatively, myocardial infarcts accounting 
for a third of these. Measures that can be taken 
by the surgical team to reduce the incidence of 
these complications include appropriate fluid 
management to optimize preload and reduce 
myocardial stress, administration of oxygen 
therapy, and adequate postoperative pain relief. 
Chest infections complicate approximately 5% 
of AAA repairs and can be minimized by appro- 
priate analgesia without sedation, chest physio- 
therapy, and early mobilization. 



Hemorrhage 

Problematic hemorrhage intraoperatively is 
usually due to bleeding from the proximal 
suture line or inadvertent iatrogenic vessel 
injury. The renal and iliac veins are particularly 
susceptible to damage during initial exposure, 
and all attempts should be made to address this 
before proceeding to aneurysm repair. Difficult 
dissections in freeing the iliac arteries for 
clamping are probably best managed with intra- 
luminal balloon occlusion rather than poten- 
tially hazardous clamp application. 

Generalized postoperative hemorrhage is 
most often due to an acquired coagulopathy 
rather than a direct consequence of surgery. 
Massive transfusion and hypothermia lead to 
clotting factor deficiencies, low platelet counts, 
an impaired coagulation cascade, and altered 
platelet function. Appropriate blood product 
and platelet transfusion with rewarming is nec- 
essary to reverse these problems and should be 
guided by serial laboratory coagulation studies 
and hematology advice. 



Renal 

Impaired renal function post open AAA repair 
accounts for 5% to 12% of all early complica- 
tions, and of these nearly half require renal 
replacement therapy with dialysis. These figures 
reflect a substantial decrease in the incidence 
of renal morbidity and are attributed to the 
improvements made in its pre- and periopera- 
tive management. Advancing knowledge and 
understanding of radiological contrast-induced 
nephrotoxicity has led surgeons to defer surgery 
following such investigations in order to allow 
renal recovery. Current perioperative fluid 
management regimes that are guided by sensi- 
tive invasive indicators of circulating volume 
(e.g., Central Venous Pressure (CVP) lines) have 
also improved the perioperative maintenance of 
a normal cardiac output and renal blood flow. 
The exact cause of renal failure despite these 
measures after AAA repair remains unknown. It 
is highly probable that the damage is a conse- 
quence of debris embolizing from the diseased 
aortic wall that is dislodged during proximal 
aortic cross-clamping. Meticulous preoperative 
image analysis may highlight this potential 
complication before surgery and thus allowing 
modification of the surgical technique. 

Patients presenting for elective open AAA 
repair with known renal impairment are at the 
greatest risk of postoperative renal failure. 
Administration of renoprotective agents such as 
mannitol, frusemide, or dopexamine in these 
patients may limit this risk, but substantial evi- 
dence for this is lacking. 

Limb Ischemia 

Limb ischemia attributable to AAA surgery is 
seen in 1% to 4% of cases. This is due to 
either distal embolization of debris from the 
aneurysm sac or more rarely graft thrombosis. 
The microemboli released following AAA repair 
are usually too small for surgical intervention to 
be warranted, triggering a variety of limb symp- 
toms such as persistent pain, cold, and blue toes. 
Rarely, larger emboli may cause postoperative 
problems, and in this situation operative explo- 
ration is mandatory. 

Gastrointestinal 

A degree of intestinal dysmotility after laparo- 
tomy ("ileus") is very common and usually 




> 



202 



VASCULAR SURGERY 



settles with bowel rest and fluid management. 
Removal of the nasogastric tube should not 
be too premature, and reintroduction of a 
normal diet must not occur until there is objec- 
tive evidence of returning bowel function (e.g., 
flatus). 

The most severe gastrointestinal following 
open AAA surgery is colonic ischemia. This con- 
dition, fortunately, occurs much less often then 
ileus, in about 1% of open AAA repair. The 
cause of this potentially fatal complication is 
explained by inadequate colonic perfusion via 
the IMA or internal iliac artery. It may occur 
due to inappropriate IMA ligation (see earlier), 
inadvertent collateral vessel damage during 
IMA ligation distant from its origin, or internal 
iliac artery thrombosis or embolism. Sinister 
postoperative symptoms include bloody diar- 
rhea, left iliac fossa pain, and an explained high 
intravenous fluid requirement. 

In cases of colonic ischemia, urgent 
colonoscopy is indicated, and management is 
dependent on the findings. Patchy areas of 
partial bowel wall ischemia and sloughing may 
well settle with conservative bowel rest and 
intravenous antibiotics. Severe bowel ischemia, 
which is evident as a full-thickness infarction, 
requires urgent resection of the affected colon 
with end-colostomy 

Venous Thromboembolism 

The incidence of deep venous thrombosis 
(DVT) and pulmonary embolism following 
open AAA surgery is less than is seen with other 
general surgical operations. This is probably 
explained by the intraoperative heparin antico- 
agulation administered during surgery. Never- 
theless, DVT still occurs in 8% of patients, 
and that risk should be addressed with sub- 
cutaneous prophylactic low-molecular-weight 
heparin, compression stockings, and early 
patient mobilization where appropriate. 

Neurological 

The most devastating and feared complication 
after open AAA repair is that of paraplegia. For- 
tunately, this is a rare event in infrarenal surgery 
as compared to thoracoabdominal aneurysm 
repair. It results following ligation of an abnor- 
mally low accessory spinal artery (of 
Adamkiewicz), which is usually found in a 



descending thoracic or upper abdominal aorta. 
Interruption of spinal blood flow through col- 
lateral vessels by compromising internal iliac 
artery perfusion may also result in paraplegia, 
but this is more frequently seen in conjunction 
with systemic hypotension as experienced 
during ruptured AAA. 

Careless dissection may damage the impor- 
tant autonomic neural pathways found on the 
left side of the infrarenal aorta. The true inci- 
dence of impotence and retrograde ejaculation 
after elective AAA surgery is unknown, but may 
approach 20%. 

Late Complications 

Delayed complications arising from open AAA 
surgery are infrequent and can be classified as 
graft or wound related. 

Graft-related late complications include false 
aneurysms (0.2% to 1.2% at 3 years) due to 
anastomotic disruption, graft infection, and 
aortoenteric fistulas. Data suggest that the com- 
bined likelihood of infection or fistula at 10 
years is approximately 5% and is much more 
likely if a femoral anastomosis is used. 

Long-term wound disorders such as poor 
cosmesis requiring revision or hernia depend 
on both surgical technique and perioperative 
factors such as wound infection, nutritional 
status, diabetic management, etc. Surgical atten- 
tion is not required in the vast majority of cases, 
and each presentation should be assessed indi- 
vidually on its merits. 

Endovascular Repair of Abdominal 
Aortic Aneurysm 

The first successful treatment of an AAA by 
transfemoral endoluminal repair was reported 
over a decade ago. Since that time, advances in 
endovascular surgery coupled with a greater 
experience and understanding of this technique 
has led to possibly a viable alternative to open 
repair in aneurysm management (Woodburn et 
al., 1998). The basic principle of endovascular 
management is the exclusion of the AAA from 
the systemic circulation by means of a pre- 
operatively sized deployed stent graft, thus 
preventing further aneurysm expansion 
and elimination of the rupture risk. For this 
to be achieved, a self-expanding or balloon- 



203 



ANEURYSMAL DISEASE 




expandable device is advanced into the abdom- 
inal aorta to be released at a position so that 
the seal is generated both proximal and distal to 
the aneurysmal segment. Depending on the type 
of repair, the procedure may or may not require 
a combined surgical bypass for successful 
treatment. 

Suitability for endovascular intervention 
depends on extensive preoperative investigation 
of aneurysm morphology, and the limitations of 
currently available stent graft devices mean that 
this approach is not an option in all elective or 
ruptured cases. At present, about 50% to 60% of 
all elective AAAs could potentially be considered 
for stenting,but this figure is extremely variable 
as much debate continues regarding the exclu- 
sion criteria for the procedure. With current 
devices it is generally agreed that the most 
important morphological features in assessing 
aneurysms' suitability for stenting are as follows: 
an adequate length (>15mm) of infrarenal 
aneurysm neck with a maximal neck diameter 
of 30 mm to ensure a good proximal seal; limited 
vessel tortuosity, defined as vessel angulation 
(60 degrees at the neck of the aneurysm and 90 
degrees at the iliac arteries); and iliac arteries of 
sufficient caliber (>7 to 9mm) to tolerate the 
passage of the graft as it is admitted into the 
infrarenal aorta. Features that are severe as rel- 
ative contraindications to the procedure include 
short (<25mm) and wide (>14mm) common 
iliac arteries and the presence of mural throm- 
bus in the aneurysmal sac. Funnel-shaped necks 
and excessive calcification are absolute con- 
traindications to endovascular repair. 

The exciting advances in this arena offer a less 
invasive approach to AAA management so that 
some patients considered unfit for open surgery 
may well be candidates for endoluminal repair. 
Results of endovascular intervention as com- 
pared to open repair are currently being 
assessed with prospective trials such as the 
ongoing Endovascular Arterial Reconstruction 
(EVAR) project. At this stage it would appear 
that endovascular repair offers mortality and 
morbidity rates similar to those of open surgery, 
but stenting has the advantage of a markedly 
reduced length of total in-hospital stay and 
diminished requirement for intensive care and 
high dependency support. The financial advan- 
tage of this, however, is offset by the major draw- 
back of this approach, which is the cost of the 
stent graft itself. 



Type of Endovascular Procedure 

Three distinct endovascular prostheses are cur- 
rently available for AAA repair. All the manu- 
factured devices form the proximal seal in the 
infrarenal aortic segment, but differences exist 
in the location of their distal "landing site." The 
aorto-aortic straight graft resides entirely in the 
abdominal aorta but is only suitable in 5% of 
cases amenable to endovascular repair. Bifur- 
cated stents can be used in 30% of patients, with 
the distal seals formed in the iliac vessels. The 
remainder (65%) of suitable aneurysms may be 
repaired with aorto-uniiliac stent graft devices. 
This latter method requires a combined surgi- 
cal crossover procedure (usually femoro- 
femoral) for maintenance of contralateral lower 
limb blood supply following radiological 
embolization or surgical occlusion of the non- 
graft common iliac artery. 



Perioperative Management 

The patient should be assessed and prepared for 
endoluminal intervention as discussed earlier 
for open repair. Adequate appropriate radiol- 
ogical imaging should be obtained to enable 
calibration of both aneurysm and related 
vasculature for graft sizing. Informed consent 
should include the known specific complica- 
tions associated with endovascular repair and 
also mention the possibility of conversion to 
open surgery if necessary. The operating room 
should be equipped with a C-arm or equivalent 
for intraoperative imaging, and adjunctive input 
is required from both the resident surgical and 
interventional radiology team. 

After anesthesia induction, the patient is 
appropriately positioned, prepped, and draped. 
The procedure commences by surgical cut- 
down to the femoral artery to gain access to the 
arterial circulation. The device is admitted 
through the exposed artery and advanced prox- 
imally under radiological control until deploy- 
ment of the appropriate site for aneurysm 
exclusion. Correct graft position and the 
absence of endoleak is confirmed by on-table 
angiography, and the groin is closed in a routine 
fashion unless a surgical crossover graft is to be 
performed. At the end of the procedure, the 
lower limbs should be assessed for evidence of 
graft thrombosis or embolism before transfer to 
the recovery area. The patient can be mobilized 




> 



204 



VASCULAR SURGERY 



gently the day after surgery, and discharge can 
be expected at 5 to 7 days in uncomplicated 
cases with a satisfactory early postoperative CT 
scan. Follow-up guidelines currently advise 6- 
month CT and plain lumbar spine imaging in 
the first year and then annually thereafter. 

Complications of Endovascular Abdominal 
Aortic Aneurysm Repair 

Endovascular AAA repair unfortunately carries 
with it a distinct spectrum of specific complica- 
tions in addition to "routine" causes of postop- 
erative morbidity. 



Iatrogenic Vessel Injury 

The introduction of guidewires, large-bore 
catheters, and a stent graft poses the risk of arte- 
rial and proximal vessel injury such as rupture 
or dissection. Delayed manifestations of iatro- 
genic vessel damage may present later, with the 
development of a false aneurysm at the cannu- 
lation site, and requires prompt surgical repair. 



Endoleak 

Endoleak refers to the persistence of arterial 
flow of blood into the "excluded" aneurysm sac 
but outside the lumen of the deployed stent 
graft. It represents one or more of the following 
processes: incomplete sealing of the proximal or 
distal landing sites (type I); continued blood 
flow into the sac by collateral and lumbar vessels 
(type II); an incomplete seal at junctions of 
overlapping graft components or ruptured graft 
fabric (type III); or leakage of blood through a 
porous graft membrane (type IV). The inci- 
dence of endoleak is approximately 20% and 
may be further classified as immediate, early, 
or late. Without treatment, the aneurysm may 
continue to expand, thus carrying the risk of 
rupture, although most cases seal spontaneously 
with thrombosis. Bearing this in mind, the 
majority of vascular surgeons initially manage 
an endoleak with simple observation. If, after a 
prescribed time period, the endoleak persists or 
rapid aneurysm expansion is observed, further 
interventions such as endoluminal graft exten- 
sion deployment, band ligation of the aortic 
aneurysmal neck, or even open repair may be 
indicated. 



Embolization 

Endoluminal stent graft deployment carries 
the risk of distal embolization of debris from 
the aneurysmal sac. This is particularly true in 
cases of extensive mural thrombosis, and there- 
fore the benefits of endovascular repair should 
be questioned in these circumstances. Shower- 
ing of microemboli may result in renal failure 
and leg ischemia requiring amputation. 

Graft Migration, Dislocation, 
and Displacement 

Long-term follow-up of endoluminal stenting 
has shown that the stent devices may migrate, 
resulting in endoleak, dislocating, and jeopard- 
izing the seal and kink, with potential thrombo- 
sis. The advent of second-generation devices has 
attempted to address these problems with rein- 
forcement of graft struts and the introduction 
of suprarenal uncovered fixation. 

Postimplant Syndrome 

Well described in the literature, postimplant 
syndrome refers to the presence of early back 
pain and fever without leukocytosis following 
endoluminal AAA repair. It lasts for up to 1 week 
after insertion. The condition is self-limiting 
and is managed conservatively. 

Conversion to Open Repair 

Failure of endovascular management of AAA 
may require conversion to open repair, occur- 
ring in 2% of cases. Indications for such 
measures include aortic rupture during a 
primary stenting procedure, persistent endoleak 
with failed secondary intervention, and graft 
infection. 

Ruptured Abdominal 
Aortic Aneurysms 

Ruptured aortic aneurysms are uniformly fatal 
without operative intervention, explaining the 
emphasis of AAA management of early detec- 
tion and elective repair. Unfortunately, the vast 
improvements in outcome following elective 
AAA repair have not simultaneously been wit- 
nessed in the emergency situation, and opera- 



205 



ANEURYSMAL DISEASE 




tive mortality under these circumstances has 
remained static over the last 20 years at between 
40% and 70%. Even this estimation can be con- 
sidered conservative, as the figure does not take 
into account community prehospital deaths, 
and therefore an overall mortality following 
AAA rupture is probably in the region of 80% 
to 90%. 

The exact process that triggers rupture of 
an AAA remains unknown. Several factors 
(discussed earlier) are known to suggest an 
increased likelihood of rupture and the most 
important of these is aneurysm size. 

Clinical Presentation 

A high index of suspicion is paramount in 
clinching the diagnosis. Any patient with a 
known AAA who presents with sudden severe 
abdominal or back pain has a ruptured 
aneurysm until proven otherwise. The difficulty 
arises in atypical presentations of patients with 
unknown aneurysmal disease leading to detri- 
mental delays in surgical management. Rup- 
tured AAA should feature in the differential 
diagnosis of any patient who presents with 
unexplained hypertension, severe abdominal 
pain, or cardiac arrest with no prior history of 
myocardial disease or trauma. 

Clinical symptoms of rupture are varied, but 
classically comprise sudden back or abdominal 
pain with an episode of collapse, fainting, and 
maybe nausea. Examination reveals a pulsatile 
mass on abdominal palpation that may or may 
not be tender. The patient may be pale, irritable, 
clammy, tachycardic, and hypotensive. Mainte- 
nance of a normal blood pressure may indicate 
a contained rupture, affording the clinician time 
to confirm the diagnosis with appropriate 
imaging prior to surgery. This luxury is not 
offered in an uncontained ruptured AAA when 
the patient should be taken to the operating 
room for operative repair without delay. 

Operative Repair of a Ruptured Abdominal 
Aortic Aneurysm 

Perioperative management is aimed at stabiliz- 
ing the precarious hemodynamic status of the 
patient. After an initial brief history and exam- 
ination, venous access should be secured with 
at least two large-bore cannulae and blood 



samples withdrawn for biochemistry, hematol- 
ogy, and urgent crossmatch (at least 10 units) of 
packed red cells and blood products. 

Arguments persist regarding the optimal goal 
of fluid resuscitation in cases of ruptured AAA. 
It has been suggested that overzealous fluid 
management runs the risk of a sudden increase 
in blood pressure and hence converting a con- 
tained leak into a frank rupture. Management 
strategies should aim to keep a systolic blood 
pressure of 90 mm Hg by infusion of intra- 
venous fluids until operative repair can be per- 
formed. Patient response should be assessed 
with continuous hemodynamic monitoring and 
a urinary catheter inserted. Early involvement of 
both the surgical and anesthetic team is desir- 
able in order to optimize patient status and min- 
imize operative delay. 

In some stable presentations of suspected 
AAA rupture, radiological imaging may be per- 
formed to confirm the diagnosis and delineate 
aneurysmal anatomy. The investigation of 
choice remains CT scanning. Ultrasound is of 
little benefit due to its inability to reliably dis- 
tinguish a ruptured AAA. 

Operative management for leaking AAA is by 
open repair by midline laparotomy. Although 
some centers have reported encouraging results, 
endovascular management currently plays little 
role in the management of ruptured AAA. 
Surgery should be expedient, performed by the 
most senior available member of the vascular 
team. Following incision, brisk but careful dis- 
section should proceed with placement of the 
clamp in the infrarenal aorta, avoiding damage 
to the left renal vein. Temporary supraceliac 
clamping maybe performed to initially control 
problematic bleeding and facilitate dissection 
for later re-siting in the infrarenal aortic 
segment. In extreme circumstances, the sur- 
geon's fingers or an intra-aortic balloon maybe 
used to gain proximal control. Intravenous 
antibiotics should be administered to limit graft 
infection, but unlike in elective cases, intra- 
venous heparin is not routinely given. Once the 
aorta is cross-clamped, the operation is then 
performed as is described for elective AAA. 

Outcome Following Repair of a Ruptured 
Abdominal Aortic Aneurysm 

Significant postoperative morbidity following 
repair of ruptured AAA occurs in 50% to 70% 




> 



206 



VASCULAR SURGERY 



of cases (Heller et al., 2000). This figure repre- 
sents both suboptimal patient preparation for 
major surgery and the sequelae of prolonged 
hypotension experience pending operative 
repair. Cardiorespiratory disorders and renal 
impairment are the most commonly encoun- 
tered complications, closely followed by 
ischemic colitis. The latter condition carries a 
subsequent mortality of 25% and results from 
gut hypoperfusion secondary to aneurysm 
rupture. The clinician should anticipate its 
diagnosis. 

Assuming operative survival, patients with a 
ruptured AAA enjoy a long-term prognosis as 
governed by their other comorbidities. Repair 
has not been shown to diminish the patient's 
quality of life postoperatively, and no different 
exists in outcome should the aneurysm have 
been repaired electively 

Inflammatory Abdominal 
Aortic Aneurysms 

An important subdivision of infrarenal aortic 
aneurysmal disease is the inflammatory aortic 
aneurysm (Rasmussen and Hallett, 1997). 
Inflammatory AAAs were not classified per se 
until the early 1970s, and even today their 
nature in terms of being a distinct clinical and 
pathological entity is still disputed. 

Definition of the inflammatory AAA depends 
on the presence of a triad of factors: a thickened 
aneurysmal wall, marked perianeurysmal/ 
retroperitoneal fibrosis, and dense adhesions 
involving adjacent abdominal viscera to the 
aneurysm. These aneurysms comprise 5% to 
10% of all AAAs, and the typical patient is a 
male in his sixth decade. Females are rarely 
affected by the disease (male-to-female ratio of 
20 : 1). Interestingly, the average age at diagnosis 
is about 10 years younger than those patients 
presenting with noninflammatory aortic 
aneurysms. Postulated risk factors that are 
specific for the development of inflammatory 
AAA include cigarette smoking and a genetic 
predisposition. 

There is currently much debate regarding the 
etiology and pathogenesis of the inflammatory 
AAA. Until recently, it was generally upheld that 
these aneurysms were of a distinct exclusive 
nature and therefore to be considered different 
from their noninflammatory counterpart. This 



belief is now being challenged. An early hypoth- 
esis suggesting the periaortic fibrosis was sec- 
ondary to retroperitoneal blood leakage from 
tiny perforations in a previously noninflamma- 
tory aneurysm has been rejected. Similarly, 
other theories of an initial prodromal athero- 
sclerotic AAA developing into an aneurysm of 
inflammatory type remain unproven. The sug- 
gested mechanism for this transformation is by 
either lymphatic vessel compression by the 
expanding aneurysm resulting in stasis, edema 
and subsequent fibrosis, or as a direct conse- 
quence of an inflammatory reaction between 
blood in the aneurysmal sac and the aortic 
wall. Viral infection with herpes simplex or 
cytomegalovirus has also been proposed as a 
causal factor, with evidence of their presence 
in the aortic wall proven by DNA polymerase 
reactions. 

A more convincing theory regarding inflam- 
matory AAA development is that it is the result 
of a specific inflammatory process. This process 
is responsible for both inflammatory and 
noninflammatory AAAs, and thus we are not 
to consider the two pathologies as distinct 
pathological entities; rather, the former is an 
"inflammatory variant" of the latter. There 
seems to be good evidence for this theory. 
Initially, histological analysis revealed the 
presence of a chronic inflammatory infiltrate 
present in the walls of inflammatory and 
noninflammatory aortic aneurysms, with the 
only difference being an augmented level of 
inflammatory response in the putative inflam- 
matory AAA. This finding was then supported 
by serial observations of a progressive inflam- 
matory response from the initial atherosclerotic 
to the inflammatory AAA proven both under 
the microscope and radiologically It is sug- 
gested that there is a primary inflammatory 
reaction to an antigen present in the aortic wall 
whereby there is gradual infiltration of B lym- 
phocytes, T lymphocytes, and macrophages. 
Subsequent cytokine production from the 
infiltrate triggers a proteolytic cascade with 
increased turnover of MMPs, elastin, and colla- 
gen. With time the aortic wall begins to degrade, 
losing tensile strength, and the aneurysm 
begins to develop. It is proposed that this 
inflammatory response is accentuated further 
in smokers and those with a genetic pre- 
disposition, explaining an earlier presentation 
of aneurysmal disease. 



207 



ANEURYSMAL DISEASE 




Clinical Features 

The inflammatory AAA is typically sympto- 
matic with only 20% cases being discovered 
incidentally. Classically, the presence of abdom- 
inal or back pain, weight loss, and an elevated 
ESR in patients with a known AAA confers 
a diagnosis of inflammatory aneurysm until 
proven otherwise. Clinical examination may 
reveal a pulsatile mass on abdominal palpation 
and generalized evidence of weight loss. Specific 
blood tests to be taken if an inflammatory AAA 
is suspected are an ESR, which will be raised in 
70% cases signifying the acute phase response, 
and less reliably a white cell count. Serum elec- 
trolytes and creatinine should also be taken to 
look for evidence of obstructive uropathy and 
associated chronic renal failure (10% to 20% of 
patients). 

Diagnosis 

Despite symptomatology and available labora- 
tory investigations, the inflammatory AAA still 
tends to be a diagnosis that is made at opera- 
tion. Preoperative CT scanning can sensitively 
detect aneurysm wall thickening and peri- 
aneurysmal fibrosis but depends on the radiol- 
ogist's awareness to make the diagnosis. 
Although ultrasound remains useful in routine 
AAA surveillance, it is less helpful than CT in 
distinguishing the inflammatory variant. If 
there is any suspicion of ureteric involvement by 
the retroperitoneal fibrosis, an intravenous pyel- 
ogram should be requested. 

Management 

Although inflammatory aneurysms are usually 
larger than their atherosclerotic counterpart, it 
is generally accepted that the risk of rupture is 
lower. Presumably this is due to the paradoxical 
strengthening of the periaortic tissue by fibrotic 
change. Some clinicians advocate administra- 
tion of oral corticosteroids in an effort to atten- 
uate the inflammatory aneurysmal response, 
but there is no evidence to support this. In fact 
there is a body of opinion that suggest steroid 
therapy may actually increase the risk of 
inflammatory AAA rupture by a reduction in 
the protective fibrosis. Ultimately, surgery 
remains the treatment of choice for inflamma- 
tory AAA. 



Surgical Intervention for Inflammatory 
Aortic Aneurysms 

At laparotomy the inflammatory AAA is easily 
recognized with its thickened aortic wall and 
shiny, smooth white appearance. There is 
periaortic and retroperitoneal change with 
adhesions invariably involving the duodenum 
and less commonly the inferior vena cava and 
left renal vein. Disease affecting the ureters 
occurs in 20% to 40% of cases, which may neces- 
sitate a team approach with both the vascular 
surgeon and urologist. 

Aneurysm repair should proceed with as 
little dissection as possible. The propensity of 
these aneurysms to bleed compounded by the 
high risk of iatrogenic damage to neighboring 
structures poses potentially high surgical mor- 
bidity. Early inflammatory AAA surgery with 
futile attempts at periaortic adhesiolysis were 
complicated by needless duodenal enterotomies 
and inadvertent venous and ureteric injury, and 
this approach to repair has now been discarded. 
The method of surgical repair is otherwise 
as previously described for noninflammatory 
infrarenal AAAs, and operative mortality rates 
for elective inflammatory AAA repair should be 
comparable (3% to 4%).Ureterolysis at the time 
of AAA repair remains a controversial issue. 
Over half of inflammatory AAAs demonstrate 
ureteric involvement on preoperative CT scan- 
ning. We advise that such ureteric surgery be 
reserved for those exhibiting signs or symptoms 
of obstruction or proven incipient uropathy on 
preoperative investigation. Early consultation 
with the urology team is advisable in these 
circumstances. 

Thoracoabdominal 
Aortic Aneurysms 

Aneurysmal diseases affecting both the abdom- 
inal and thoracic aorta are known collectively as 
thoracoabdominal aortic aneurysms (TAAAs). 
In clinical practice these essentially refer to 
aneurysms arising above the celiac axis. Previ- 
ously considered as quite a rare condition, 
recent advances in clinical diagnostic capability 
by methods such as angiography, CT, and MRI 
suggest that these aneurysms in fact account for 
up to 10% of the aortic aneurysm population. 
The management of these patients is complex 




» 



208 



VASCULAR SURGERY 



and indeed may fall into the realm of both the 
vascular and cardiothoracic surgeon. It is there- 
fore suggested that these aneurysms are best 
dealt with at a tertiary referral center with 
appropriate expertise and not routinely at the 
district general hospital. 

Etiology 

Atherosclerosis is the most commonly impli- 
cated factor in the development of TAAA 
disease. The classical fusiform aneurysm that 
results is typically seen in men over the age of 
50 years, usually with evidence of arterial 
atherosclerotic pathology elsewhere. In fact, 
over half of TAAA presentations are subse- 
quently discovered to have coexistent aneurys- 
mal disease at an alternative site including the 
infrarenal aorta and femoral and popliteal 
arteries. Other causal factors to be excluded are 
thoracic dissection with secondary aneurysm 
formation, cystic medial necrosis (e.g., Marfan 
syndrome, Takayasu disease, trauma leading to 
false aneurysm), and infection. Previously the 
most common cause of this type of aneurysm, 
syphilitic aortitis leading to the classical Craw- 
ford type I (see later) saccular TAAA, has now 
thankfully declined and is rarely seen nowadays 
in non-Third World practice. 

Clinical Features 

Clinical presentation of TAAA can be catego- 
rized as asymptomatic, symptomatic but not 
ruptured, and ruptured. 

Asymptomatic Thoracoabdominal 
Aortic Aneurysm 

The vast majority of TAAAs are without 
symptoms. Unlike infrarenal and peripheral 
aneurysms, it is also improbable that routine 
clinical examination will yield the diagnosis. In 
these patients it is the review of the chest radi- 
ograph that initially arouses clinical suspicion 
of TAAA, for further investigations to confirm. 

Symptomatic Nonruptured 
Thoracoabdominal Aortic Aneurysm 

Symptomatic TAAAs usually result from local 
pressure effects, and this most commonly man- 



ifests as chest pain. The pain is central and radi- 
ates through to the upper back, depending on 
the neighboring anatomy (e.g., spinal nerves or 
vertebral column erosion). Irritation of the 
recurrent laryngeal nerve and bronchial struc- 
tures may result in voice changes, stridor, 
and dyspnea. Gradual unexplained orthopnea, 
paroxysmal nocturnal dyspnea, and ankle 
swelling all indicate a failing myocardium, and 
this may be due to a compromised pulmonary 
circulation by TAAA compression. Occasionally, 
if the abdominal component of the TAAA is 
large enough, the clinician or even the patient 
may discover a pulsatile mass. Occult symptoms 
of distal embolization should always arouse sus- 
picion of an ongoing proximal aneurysmal 
process, thus necessitating appropriate investi- 
gation for exclusion of TAAA. 

Ruptured Thoracoabdominal 
Aortic Aneurysm 

A TAAA rupture can be varied in its clinical 
presentation. There is usually a history of acute 
severe chest pain, and examination may reveal a 
hemodynamically unstable patient requiring 
vigorous resuscitation. The site of rupture 
and possible involvement of other thoracic 
structures govern other associated symptoms. 
Massive hematemesis may result if rupture 
occurs into the esophagus, whereas hemoptysis 
and dyspnea suggests the possibility of rupture 
into the tracheobronchial tree. Hemorrhage into 
the pleural cavity or retroperitoneal space can 
often result in a more delayed and profoundly 
hypovolemic presentation. 

Diagnosis 

Examination of a standard chest radiograph 
may reveal a widened mediastinum, loss of tho- 
racic aorta outline, or a "mass" extending into 
the left hemithorax. Calcification in the wall of 
the aorta is seen in about one third of patients. 
In cases of TAAA rupture into the pleural space, 
evidence of a hemothorax should be seen. 
Confirmation of the diagnosis is achieved by 
spiral CT scanning, which usually gives all the 
required preoperative information regarding 
aneurysm size, visceral artery relation, and 
proximal extent of aneurysm. Magnetic 
resonance imaging techniques are equally 



209 



ANEURYSMAL DISEASE 




instructive, but their availability and cost limits 
their use. Unlike investigation of infrarenal 
aneurysms, ultrasound for TAAAs offers unreli- 
able and poor quality results; hence, it plays no 
role in either radiographic diagnosis or surveil- 
lance. Angiography is indicated only if CT scan- 
ning does not give sufficient information 
regarding relationships to visceral (especially 
renal) arteries and for the evaluation of possi- 
ble visceral artery stenoses. 

Classification 

Classification of thoracoabdominal aneurysms 
is into one of four types as described by Craw- 
ford: 

Type I: Aneurysm involves the descend- 
ing thoracic aorta as far as visceral 
artery branches 

Type II: Aneurysm extends from the aortic 
arch, just distal to the left sub- 
clavian artery as far as the aortic 
bifurcation (involves visceral 
arteries) 

Type III: Aneurysm commences in the 
midthoracic aorta and extends to 
the aortic bifurcation 

Type IV: Aneurysm starts at or just below 
the diaphragm but above the renal 
arteries and extends as far as the 
aortic bifurcation 

Of the four types, surgical repair is carried out 
most often on type IV. 

Management of Thoracoabdominal 
Aortic Aneurysm 

As in all surgery a balance must be struck 
between the high morbidity and mortality of 
operative repair, with a reported 5-year survival 
of 20% in nonoperative management. Aneurys- 
mal size is important, and as general rule, 
asymptomatic aneurysms are not considered 
for intervention unless maximal diameter 
exceeds 6 cm. This threshold may be reduced in 
saccular pathology due to the increased propen- 
sity to rupture. 

Extensive preoperative preparation is man- 
datory in order both to exclude patients with 
insufficient physiological reserve to survive 
operation and to allow adequate planning of 



proposed surgery. Required investigations are as 
outlined in the workup for AAA repair, with an 
emphasis on rigorous optimization of car- 
diorespiratory and renal status. 

Open Repair of Thoracoabdominal 
Aortic Aneurysm 

Following appropriate patient positioning, 
draping, and skin preparation, a left-sided 
thoracoabdominal incision is performed. In 
high aneurysms (types I and II) some surgeons 
advocate the use of extracorporeal circulation 
with atriofemoral or femorofemoral cardiopul- 
monary bypass to reduce the increased after- 
load and left ventricular strain induced by 
thoracic aorta cross-clamping. If this maneuver 
is not performed, the requirement for expedient 
surgery is further augmented. The thoracic and 
abdominal aorta are dissected out to enable 
proximal and distal control through nona- 
neurysmal aortic (and possibly iliac) cross- 
clamping. Under normal circumstances heparin 
is not routinely given prior to clamp application 
to avoid unacceptable blood loss. An appropri- 
ately sized inlay prosthetic graft is introduced 
after opening the aneurysmal sac, and the prox- 
imal anastomosis is performed by continuous 
Prolene suture end-to-end with the thoracic 
aorta distal to the left subclavian artery. After 
confirmation of proximal anastomosis integrity, 
the visceral vessels (celiac axis, superior mesen- 
teric, and renal arteries) are then reimplanted 
into the graft with sequential clamping in order 
to minimize organ ischemic time. Most high 
intercostal arteries are oversewn, but those in 
the region of T10 to L2 must be reimplanted for 
preservation of spinal cord blood supply. After 
verification of successful vessel anastomosis 
with evidence of perfusion, the distal anasto- 
mosis is formed at the level of the aortic bifur- 
cation or iliac vessels. The aneurysm sac is 
closed over the graft and the patient closed in 
routine fashion. Postoperatively, the patient is 
transferred to the intensive care unit for close 
observation. 

The nature of type I and type IV thoracoab- 
dominal aneurysms may permit modifications 
to the outlined surgical technique in operative 
management. In certain low type IV aneurysms, 
a "rooftop" abdominal incision can be used for 
a total transabdominal access without the 
need for thoracotomy. The abdominal aorta is 




> 



210 



VASCULAR SURGERY 



approached retroperitoneally with mobilization 
of the spleen, left kidney, and bowel to the con- 
tralateral side. A vascular clamp can then be 
applied to the suprarenal aorta to allow an 
oblique proximal anastomosis to be performed, 
incorporating a tongue of native aorta from 
which the celiac axis and superior mesenteric 
and renal arteries emerge. Once achieved, the 
clamp is reapplied infrarenally to allow organ 
perfusion following minimal ischemic time. 
Certain type I aneurysms may be approached 
with the need for thoracotomy only. In a similar 
manner to type IV repair, the distal anastomo- 
sis can be obliquely fashioned to incorporate a 
reversed tongue of native vessel from which the 
visceral arteries originate and so limit organ 
ischemic time. 

Perioperative spinal fluid drainage is sug- 
gested as an adjunctive measure to lessen the 
risk of neurological complication. The tech- 
nique aims to optimize spinal cord perfusion by 
enabling regulation of the increases in spinal 
fluid pressure that result from aortic cross- 
clamping. A fine-bore intrathecal catheter con- 
nected to a pressure transducer is introduced 
between the fourth and fifth lumbar vertebrae. 
If a potentially deleterious rise in spinal fluid 
pressure are observed, fluid drainage by a tap is 
performed to correct it. The catheter is removed 
once normal neurological function is confirmed 
in the early postoperative period. 

Endovascular Repair of Thoracoabdominal 
Aortic Aneurysm 

Endoluminal stent grafting may treat suitable 
descending thoracic aneurysms without vis- 
ceral artery involvement. The principles of graft 
deployment were discussed earlier, with the 
exception that the aneurysmal neck is ideally 
situated at least 25 mm distal to the left subcla- 
vian artery. More proximal pathology may still 
be considered in the knowledge that compro- 
mised upper limb perfusion due to subclavian 
artery occlusion would require either operative 
reimplantation into the carotid artery or a 
carotid-subclavian bypass procedure. 

Outcome of Thoracoabdominal 
Aortic Aneurysm Repair 

As with AAA repair, cardiorespiratory morbid- 
ity remains the most frequently encountered 



complications postoperatively. Renal failure 
requiring replacement therapy with dialysis 
occurs in 10% and is more likely in those with 
preoperative renal dysfunction. 

Postoperative paraplegia and paraparesis 
remain the most feared legacy of thoracoab- 
dominal aneurysmal surgery. In spite of the out- 
lined operative measures to reduce this risk, 
type I and II repairs are associated with a 15% 
to 20% chance of paraplegia compared to 5% to 
10% occurring after types III and IV surgery. In 
general terms, higher thoracic aorta clamping 
equates to an increased risk of paraplegia, and 
this is supported by reported paraplegia rates 
of<l% for low type IV repairs performed by a 
total abdominal approach. Informed patient 
consent must include detailed discussion with 
respect to these risks prior to operative repair. 
If paraplegia does result from surgery, the clini- 
cian should make immediate referral to the local 
spinal rehabilitation team for early physiother- 
apy and patient support. 

Mortality rates following TAAA repair are 
variable. Assuming appropriate case selection 
and sufficient surgical expertise, acceptable 
operative mortality rates of 10% to 15% for 
types I and II aneurysms and 5% to 8% for types 
III and IV are observed. Attempted repair 
of ruptured TAAA transmits inferior results, 
and postoperative death rates are reported as 
between 40% and 60%. If operative survival is 
accomplished, the patients' long-term prognosis 
is good and nearly two thirds of operated 
patients are alive at 5 years. 

Peripheral Arterial Aneurysms 

Aneurysmal disease affecting the nonaortic 
arterial vasculature may occur in isolation or 
more commonly as part of a generalized 
systemic arterial dilatation. It is for this reason 
that a thorough examination of all susceptible 
vessels should be performed after the 
initial assessment of the presenting peripheral 
aneurysm. 

Popliteal Arterial Aneurysms 

Popliteal aneurysms are the commonest 
nonaortic aneurysms, accounting for 70% of all 
diagnosed peripheral aneurysms (Dawson and 
van Bockel, 1997). Defined as an external diam- 



211 



ANEURYSMAL DISEASE 




eter of one and a half to two times the normal 
proximal vessel, the true prevalence and inci- 
dence of the disease remains unknown. That 
said, it is estimated currently that approximately 
five patients present to a major vascular center 
on an annual basis with such lesions. Males in 
their sixth and seventh decades are most fre- 
quently affected, and the aneurysms are bilat- 
eral in around half of patients; 6% to 12% of 
presentations have a coexistent AAA. 

The majority of popliteal aneurysms are 
associated primarily with atherosclerosis but 
other etiologies include trauma, infection, and 
popliteal artery entrapment syndrome. These 
should always be considered, especially in the 
younger patient. The aneurysms occur above 
the knee joint and are either fusiform or saccu- 
lar in nature. The beaded fusiform popliteal 
aneurysms tend to be smaller (2- to 3-cm diam- 
eter) than the saccular variety with diameters 
reached of 4 to 6 cm. 

Clinical Features 

About one third of all popliteal aneurysms are 
asymptomatic; the diagnosis is made only after 
investigation of a lump in the popliteal fossa 
found on a routine clinical examination. The 
remaining two thirds are symptomatic and 
reflect the complications of these aneurysms: 
limb ischemia, rupture, or local pressure effects. 

Limb ischemia remains the commonest pres- 
entation and is due to either aneurysmal sac 
thrombosis or distal embolization of intraa- 
neurysmal clot. The emergence of ischemia 
(acute or chronic) is dependent on the presence 
of an established collateral circulation. In the 
more frequent case of acute thrombosis, such a 
circulation is usually poorly developed, and 
presentation is typically that of acute severe 
ischemia with threatened limb viability. Chronic 
thrombosis of popliteal aneurysms affords more 
time to the surgeon, resulting in one or more of 
symptoms of claudication, rest pain, blue-toe 
syndrome, or gangrene. 

Local pressure effects of popliteal aneurysms 
may cause edema by compression of the adja- 
cent popliteal vein and even local or referred 
pain by irritation of the neighboring sciatic 
nerve or its derivatives. Rupture of popliteal 
aneurysm is a rare presentation, necessitating 
immediate surgical attention in order to pre- 
serve the limb. 



Diagnosis 

The diagnosis of popliteal aneurysm should be 
suspected if an exaggerated pulsation is discov- 
ered on palpation behind the knee during clin- 
ical examination. A firm pulseless mass in the 
popliteal fossa may indicate a thrombosed 
aneurysm, especially if an aneurysm is found on 
examination of the contralateral limb. Further 
information regarding the lump should then be 
obtained by appropriate imaging. 

Arterial duplex scanning is cheap, easily 
available, and therefore ideal for diagnosis 
confirmation, sizing, and subsequent popliteal 
aneurysm surveillance. Arteriography is of less 
use in the estimation of aneurysm size, but is 
paramount in the assessment of the regional 
vasculature for determination of planned surgi- 
cal intervention. Computed tomograph and 
MRI can offer detailed assessment of the 
popliteal fossa contents and the nature of the 
intravascular thrombus content, but their 
expense and limited availability renders them of 
little use in the management of the routine 
popliteal aneurysm. 



Management of Symptomatic 
Popliteal Aneurysms 

Acute Ischemia and Rupture 

Presentation of a pale, pulseless, cold, and pares- 
thetic leg signifies an immediate threat to limb 
viability, and prompt intervention is needed to 
avert primary amputation. There are a variety 
of surgical procedures with adjuvant therapy 
(thrombolysis) for repair, depending on the 
aneurysm's size, embolic potential, and the 
status of proximal and distal vessels. 

Ideally, a preoperative angiogram should be 
obtained and the patient taken for arterial recon- 
struction without delay. The aim of surgery is 
limb preservation and popliteal aneurysm exclu- 
sion. After adequate exposure by either the 
medial or the posterior approach (see later), the 
aneurysm is excluded by means of ligation 
and bypass grafting, inlay grafting, or resection 
with primary anastomosis. There should be a 
low threshold for performing an additional fas- 
ciotomy as indicated by the individual clinical 
picture. 

Bypass grafting after proximal and distal 
aneurysm ligation remains well established and 




> 



212 



VASCULAR SURGERY 



is the usual technique employed. The conduit 
may be prosthetic graft or preferably autoge- 
nous saphenous vein if available. Improved 
distal run-off can be achieved by the intra- 
operative use of balloon-tipped embolectomy 
catheters, and enhanced perfusion following 
surgery should be confirmed by an on-table 
angiogram. The commoner complications of 
this surgery include graft thrombosis, infection, 
and deep vein thrombosis. Resection of 
popliteal aneurysms is indicated only in the 
larger, saccular type that involves a short 
segment of popliteal artery, so the resultant end- 
to-end anastomosis can be performed without 
tension. Great care must be taken in resection to 
avoid damage to the adjacent popliteal vein, and 
for this reason some surgeons avoid this method 
of aneurysm repair. 

Thrombolysis has an important role to play in 
the management of acute symptomatic popliteal 
aneurysmal disease. In thrombosed aneurysms 
with acute ischemia, a percutaneous trans- 
femoral arterial catheter can be inserted and 
lodged in the aneurysmal thrombus. Infusion of 
thrombolytic agent [recombinant tissue-type 
plasminogen activator (rtPA), streptokinase, or 
urokinase] is commenced at an appropriate 
dose, and check angiograms are performed at 
suitable intervals (every 4 to 6 hours) to confirm 
the clinical observations of improved blood 
supply. Angiographic demonstration of ade- 
quate run-off is an indication to stop throm- 
bolytic infusion and proceed to bypass grafting. 
Long periods of thrombolysis carry the risks of 
bleeding and embolization of released intraa- 
neurysmal thrombus. Strict observation for 
excessive hemorrhage and worsening foot 
ischemia must be adhered to during treatment, 
and they mandate cessation of the infusion if 
they occur. In advanced ischemia with loss of 
sensation and muscle power, attempts at surgi- 
cal revascularization should not be delayed, and 
thrombolysis should be restricted to intraoper- 
ative use in an effort to clear tibial and peroneal 
vessel run-off. 

Chronic Ischemia 

Symptoms of chronic ischemia in popliteal 
aneurysmal disease may be due to thrombosis 
or distal embolization. A conservative nonoper- 
ative approach maybe considered in the case of 
thrombosed popliteal aneurysm, as an acute 



deterioration is unlikely due to the established 
collateral circulation. Distal embolization, on 
the other hand, involves a persistent blood flow 
through the aneurysm (unlike thrombosis), 
putting the patient at risk of further embolic 
episodes and worsening limb ischemia unless 
the aneurysm is surgically excluded. 

Management of Asymptomatic 
Popliteal Aneurysms 

Discovery of these aneurysms is usually on 
examination following diagnosis of aneurysmal 
disease elsewhere such as the abdominal aorta 
or contralateral popliteal artery. Subsequent 
treatment remains a controversial issue with as 
yet no clear guidelines for surgical intervention. 
The recent trend of aggressive repair in the 
majority of asymptomatic popliteal aneurysms 
is currently under review in an effort to balance 
the inherent surgical morbidity of repair and 
the fact that for many patients their popliteal 
aneurysm will be associated with no long-term 
adversity. Review of the literature suggests that 
following factors favor elective surgery: ane- 
urysm size >2 cm, the presence of mural throm- 
bus (potential embolic source), autogenous 
conduit availability, absent ankle pulses (indi- 
cating possible "silent" embolization), and a 
long life expectancy. Classical popliteal ane- 
urysm exclusion and bypass as described earlier 
is the operation of choice in this situation. 
Endovascular procedures have been described 
in the treatment of popliteal aneurysms but the 
long-term patency rates and outcomes are at 
present unknown. 

Surgical Approach in Popliteal 
Artery Aneurysms 

Access to the popliteal artery in aneurysmal 
surgery is achieved by either the medial or pos- 
terior approach. 

In the medial approach the knee is partially 
flexed and the skin incision follows the course 
of the long saphenous vein, taking care to pre- 
serve the vein for later harvest. The adductor 
muscles are retracted, allowing entry to the 
popliteal space with decent exposure and better 
access to the superficial femoral and tibial arter- 
ies than with the posterior approach. A further 
advantage of this method is that the incision 



213 



ANEURYSMAL DISEASE 




may be easily extended without the need for 
patient repositioning. 

The posterior approach requires the patient 
to be prone, and the incision is made between 
the heads of the gastrocnemii extending proxi- 
mally Excellent exposure of the popliteal artery 
is achieved, but access to more proximal or 
distal vessels proves more problematic. Further- 
more, unless the short saphenous vein is to be 
used for bypass, this approach compels a second 
incision for vein harvest. 



Femoral Arterial Aneurysms 

Femoral arterial aneurysms can be divided 
simply into true or false aneurysms. Due to its 
ease of accessibility and the trend for more inva- 
sive medical investigation and treatment (e.g., 
cardiac catheterization, intra-aortic balloon 
pumps), iatrogenic injury of the femoral artery 
leading to false aneurysm is a relatively com- 
mon occurrence. This chapter discusses only 
true femoral aneurysms, and false aneurysms 
are considered in detail elsewhere. 

Although rare, true femoral aneurysms are 
the second commonest peripheral aneurysm 
after those affecting the popliteal artery. They 
occur in between 2% and 3% of patients with 
aortic aneurysms and tend to be a disease of 
elderly men (male-to-female ratio of 30:1). The 
condition is frequently bilateral and similar to 
popliteal aneurysms; a coexistent generalized 
aneurysmal process may be manifest in other 
anatomical sites such as the aortoiliac or 
popliteal arteries. Multiple factors are impli- 
cated in their development including athero- 
sclerosis, turbulent blood flow (at proximal 
major vessel bifurcations and infrainguinally), 
and repeated hip flexion. Other etiologies to 
exclude are infection, inflammation (e.g., sys- 
temic lupus erythematosus, Takayasu's disease), 
trauma, and connective tissue disorders (e.g., 
Marfan syndrome). 



Classification 

True femoral aneurysms are distinguished by 
the Cutler and Darling classification: 

Type 1: aneurysm involves common 
femoral artery (CFA) only, not the 
bifurcation 



Type 2: aneurysmal disease of the bifurca- 
tion, the commencement of the 
profunda femoris (PFA), or the 
superficial femoral artery (SFA) 

Clinical Features 

Femoral aneurysms may be symptomatic or 
asymptomatic. The latter group is usually dis- 
covered incidentally on clinical examination of 
a patient with an aneurysm elsewhere or suffer- 
ing symptoms of chronic limb ischemia. Pre- 
sentation of a symptomatic femoral aneurysm 
can include a pulsatile groin mass (which may 
or may not be painful), leg swelling (in large 
aneurysms due to femoral vein compression 
and deep vein thrombosis), or features associ- 
ated with chronic ischemia attributable to 
aneurysm thrombosis/embolization. 

Diagnosis 

Once clinical suspicion has been aroused, the 
diagnosis should be confirmed with a duplex 
scan. If surgery is being considered (e.g., in 
CFA thrombosis or embolization), preoperative 
angiography is desirable for further informa- 
tion regarding the proximal and distal vessels. 

Management of Femoral Aneurysms 

Symptomatic femoral aneurysms are them- 
selves an indication for surgical intervention. As 
with popliteal disease, the management of the 
asymptomatic case is less clear. It is generally 
accepted, however, that if aneurysms are >4cm 
or aortoiliac disease is present (with indicated 
surgery), then operative repair is warranted. 

Surgical Repair of Femoral Aneurysms 

The skin incision extends from a point approx- 
imately 4 cm proximal to the inguinal ligament, 
following the course of the artery (and thus 
aneurysm) to a point just distal to the CFA 
bifurcation. Once proximal and distal control is 
achieved, heparin and intravenous antibiotics 
are administered so that aneurysmal repair 
can be performed. In Type 1 aneurysms, the 
CFA-restricted disease, may be addressed by 
aneurysm resection and insertion of an inter- 
position graft [polytetrafluoroethylene (PTFE)/ 
Dacron]. Repair of type 2 aneurysms is by graft 




> 



214 



VASCULAR SURGERY 



(autogenous saphenous vein or prosthetic) 
interposition with an end-to-end technique 
between the nonaneurysmal CFA and the larger 
of the bifurcated vessels (SFA or PFA). The 
smaller vessel is then reimplanted into the side 
of the graft directly, or by the use of a second 
graft. Patency rates following repair are good, 
80% to 85% at 5 years with a low operative mor- 
tality and morbidity. 

Iliac Artery Aneurysms 

Isolated iliac aneurysms are rare with a reported 
incidence of only 1% to 2% of all aortoiliac 
aneurysmal disease. The aneurysms tend to be 
large (4 to 8 cm), with annual expansion rates 
comparable to that of AAAs. Males over the age 
of 70 years are most frequently affected, with the 
atherosclerotic process implicated in the vast 
majority of cases. Other reported etiologies 
include infection (TB, syphilis), trauma (pelvic 
fractures and iatrogenic during surgery), 
inflammation, and pregnancy. 

Clinical Features 

Iliac aneurysmal disease may be asymptomatic 
or symptomatic. Human pelvic anatomy 
accounts for the fact that even very large iliac 
aneurysms may remain "silent" until an inci- 
dental diagnosis. Any symptoms experienced 
are typically due to local effects on neighboring 
structures including the ureter, small bowel, 
iliac vein, and femoral or sciatic nerve. 

Abdominal examination may reveal the diag- 
nosis but is usually normal. Palpation of a pul- 
satile mass on rectal or vaginal examination 
should alert the clinician to the possibility of 
iliac aneurysmal disease. 



Diagnosis 

Duplex ultrasound scanning is cheap, readily 
available, and thus the preliminary investigation 
of choice in diagnosis confirmation. Conven- 
tional angiography is extremely informative, 
but due to contrast load and radiation exposure 
its use is restricted to preoperative instruction. 
The cost and availability of spiral CT and mag- 
netic resonance angiography negates their 
use routinely, but the quality of information 
obtained remains impressive. 



Management of Iliac Aneurysms 

Management strategies are outlined depending 
on whether the common iliac (CIA) or internal 
iliac (IIA) artery is involved. 



Common Iliac Aneurysms 

Small CIA aneurysms (<3cm) do not require 
surgery and can be managed conservatively. If 
the maximal aneurysm diameter lies between 3 
and 4 cm, patients should be kept under ultra- 
sound surveillance (every 6 months) and oper- 
ation offered if expansion to >4 cm. Isolated CIA 
aneurysms may be repaired by means of an 
interposition graft and oversewing of the IIA 
origin. Alternatively, the CIA aneurysm can be 
ligated (proximally and distally) with execution 
of an iliofemoral or femorofemoral crossover 
graft. 



Internal Iliac Aneurysms 

Unilateral IIA aneurysms can be addressed 
by either radiological or operative techniques. 
Coil embolization of the IIA successfully ex- 
cludes the aneurysm from the circulation with 
minimal long-term sequelae. Surgical inter- 
vention requires simple proximal and distal 
aneurysm ligation for treatment in these cases. 
Bilateral IIA aneurysms pose an interesting 
problem as bilateral aneurysm ligation or 
embolization is not a viable option to preserve 
adequate colorectal and perineal blood supply. 
In this scenario an attempt at surgical revascu- 
larization should be made. The bypass graft 
runs from the nonaneurysmal CIA to the distal 
IIA with proximal aneurysm ligation. Extensive 
hemorrhage can often be encountered in this 
difficult procedure for which the surgeon should 
be prepared. 



Visceral Artery Aneurysms 

Aneurysmal disease involving the visceral arter- 
ies is uncommon but an important cause of 
vascular admission. As imaging techniques con- 
tinue to improve, there will no doubt be a cor- 
responding increase in the diagnostic yield of 
these aneurysms, so it is hoped that manage- 
ment strategies can be applied to the elective 
rather than the emergency presentation. 



215 



ANEURYSMAL DISEASE 




Splenic Artery Aneurysms 

The splenic artery is the most common visceral 
artery to be affected by aneurysmal disease, 
accounting for 60% of all visceral aneurysms. 
They are the second most common abdominal 
aneurysm after the aorta, with an estimated 
population incidence of 1%. Vessel dilatation is 
usually minimal (<2 cm) and about one fifth of 
these aneurysms are multiple. Contrary to 
aneurysmal disease elsewhere, it is multiparous 
women who are classically affected. The etiol- 
ogy of splenic artery aneurysm is unknown, but 
theories include the vascular sequelae of multi- 
ple pregnancies, altered flow dynamics in portal 
hypertension, and multifactorial vessel wall 
degradation. 

Eighty percent of splenic artery aneurysms 
are asymptomatic. The remaining proportion 
may present with a nonspecific pain in the left 
hypochondrium or epigastrium that can radiate 
to the left flank or be referred to the left shoul- 
der by diaphragmatic irritation. As aneurysmal 
progression continues, the symptoms worsen 
and are further aggravated by excessive move- 
ment. Abdominal examination is usually 
normal, although splenomegaly is detected in 
20% of patients. Rupture occurs in 5% to 10% of 
splenic artery aneurysms, and in these cases the 
patient presents with severe upper abdominal 
pain, hypotension (volume loss), and frank peri- 
tonism. Nearly all ruptures occur during preg- 
nancy, and associated mortality rates are high 
(75% mother, 95% fetus). 

Diagnosis of splenic artery aneurysms is 
usually incidental on imaging modalities such 
as plain abdominal radiographs (calcification), 
angiogram, spiral CT, or MRI scanning. Alter- 
natively, these lesions may be discovered during 
laparotomy for other pathology. Management 
planning in splenic aneurysms tends to be 
dependent on whether symptoms exist. As a 
general rule, the majority of asymptomatic 
aneurysms maybe treated conservatively, but in 
those experiencing symptoms, surgical inter- 
vention is indicated at an early stage. Operative 
procedure is governed by the anatomical loca- 
tion of disease within the splenic artery. Proxi- 
mal aneurysms (in the first third of the splenic 
artery) are treated by either ligation or excision 
and grafting after an approach through the 
lesser sac. Ligation is indicated in aneurysms 
affecting the middle third of the artery, whereas 



either splenectomy or aneurysmal excision (for 
splenic conservation) is advocated for distal 
disease. Coil embolization performed by a 
member of the resident interventional radiol- 
ogy team may be successfully used to treat 
appropriate splenic aneurysms in high-risk 
patients. 

Hepatic Artery Aneurysms 

Aneurysmal disease involving the hepatic artery 
accounts for approximately one fifth of visceral 
aneurysms. They occur more often in the male 
population with an average age at presentation 
of 40 years, somewhat lower than is observed in 
other aneurysmal processes. Site of aneurysm 
formation is either intrahepatic (25%) or extra- 
hepatic (75%), the latter aneurysms being 
usually solitary affecting the common hepatic 
artery (60%). Isolated involvement of the right 
and left hepatic arteries is less frequent, but if 
present, a predilection for the right hepatic 
artery is seen (seven times more common). The 
cause of extrahepatic aneurysm is normally ath- 
erosclerosis, but factors such as infection, 
trauma, and cystic medial degeneration must be 
considered. Conversely, intrahepatic disease 
tends to be false aneurysms resulting from 
trauma. 

Nonruptured hepatic aneurysms are usually 
asymptomatic but may present with discomfort 
in the right hypochondrium or epigastrium. As 
its size increases, the pain radiates into the back. 
Sudden development of acute abdominal pain, 
hypotension, and peritonism indicates rupture 
of the aneurysm, occurring in 20% of cases. 
Unless the aneurysm has indeed ruptured, clin- 
ical examination is invariably normal, although 
detection of a pulsatile mass and bruits have 
been reported. 

Prerupture clinical diagnosis of hepatic 
aneurysm is problematic. Elevated serum 
amylase, bilirubin, and white cell counts may be 
observed, but more often than not diagnosis 
depends on ultrasound and CT findings. Once 
suspected, the investigation of choice for 
hepatic aneurysm is selective celiac and supe- 
rior mesenteric angiography. 

Due to high rates of rupture-related mor- 
tality of approximately 40%, proven hepatic 
aneurysms merit aggressive management. The 
exact surgical procedure is again guided by 
anatomical location of aneurysms: for extra- 




> 



216 



VASCULAR SURGERY 



hepatic aneurysms proximal to the gastroduo- 
denal artery, simple aneurysm ligation should 
be performed (hepatic blood supply maintained 
by gastroduodenal and right gastric arteries); if 
the aneurysm location is distal to the com- 
mencement of the gastroduodenal artery, then 
an excision and graft procedure should be 
attempted. Intrahepatic aneurysm repair is 
more difficult. Both embolization and limited 
liver resections have been performed, but not 
all aneurysms are suitable for coil deployment, 
and operative mortality in these cases remains 
high. 

Renal Artery Aneurysms 

Renal artery aneurysms are about as common 
as hepatic aneurysms, accounting for one fifth 
of all visceral aneurysms. The true incidence of 
these aneurysms lies between 0.3% and 1.3%, 
with less than 10% of those affected exhibiting 
symptoms. They occur in both sexes, with 
women of childbearing age considered to be of 
highest rupture risk. Etiologies of renal artery 
aneurysms are varied, but the majority (80% 
to 85%) are caused by the degradation of the 
internal elastic lamina at the renal arteriolar 
divisions leading initially to microaneurysm 
formation that then progresses into frank 
aneurysmal disease. Other factors to consider in 
aneurysm development include previous renal 
trauma and generalized arteritis. As already 
mentioned, the vast majority of these 
aneurysms are clinically silent, but if present, 
symptoms include flank pain on the affected 
side, hematuria, and systemic hypertension. 
Clinical examination may reveal a bruit on aus- 
cultation. Rupture is manifest by acute localized 
pain and hypovolemia due to either retroperi- 
toneal hemorrhage or rupture into the neigh- 
boring renal vein, resulting in a high-output 
arteriovenous fistula. 

Diagnosis is usually made at laparotomy for 
a different abdominal pathology or incidentally 
on radiographic imaging such as CT scanning, 
angiography, or MRI. Management of asympto- 
matic, low-risk renal artery aneurysms should 
be nonoperative, as the procedure carries a 
significant (5%) risk of nephrectomy. On the 
other hand, "high risk" asymptomatic popula- 
tions (defined as women of fertile age) and 
symptomatic patients should proceed to 
aneurysm repair irrespective of aneurysmal 



size. Operative procedures include excision with 
direct end-to-end arterial anastomosis, graft 
interposition (prosthetic or autogenous vein), 
and nephrectomy. Suitable aneurysms may be 
treated with embolization, but these techniques 
carry a significant risk of parenchymal damage. 



Superior Mesenteric Artery Aneurysms 

These aneurysms are rare, representing approx- 
imately 8% of all visceral aneurysms. Both sexes 
are equally affected, with infection implicated as 
the causal factor in two thirds of cases. Other 
etiologies include atherosclerosis and trauma. 
Age at presentation is variable, mycotic 
aneurysms being more common before 50 years 
of age, whereas atherosclerotic aneurysms 
usually present later (over 60 years of age). 
Superior mesenteric artery aneurysm man- 
agement tends to be aggressive, as there is a 
high rate of rupture whatever the size of the 
aneurysm. Surgery should attempt to ligate the 
aneurysm without reconstruction or excision 
due to the high risk of damaging adjacent struc- 
tures. Again, embolization may be considered, 
but it carries the potentially fatal complication 
of intestinal ischemia. 



Celiac Artery Aneurysms 

Celiac aneurysms are even less common than 
those affecting the superior mesenteric artery, 
representing 3% to 4% of all visceral 
aneurysms. There is an equal sex distribution, 
and most are due to atherosclerosis and medial 
degeneration. The majority are without symp- 
toms, but if present they include epigastric pain, 
nausea, and vomiting. A bruit may be heard on 
clinical examination. Diagnosis is confirmed by 
CT scanning or abdominal ultrasound, and sub- 
sequent management involves surgical excision 
(for small aneurysms) or reconstruction. 



Gastroduodenal, Pancreaticoduodenal, and 
Pancreatic Artery Aneurysms 

Aneurysms of these arteries are extremely rare, 
collectively accounting for 3% of visceral 
aneurysms. The patient is usually a man in his 
sixth decade with a prior history of pancreati- 
tis. Symptoms, if present, are of epigastric pain 
radiating through to the back, and angiography 



217 



ANEURYSMAL DISEASE 




or CT scanning reveals the diagnosis. Rupture- 
related death rates are high (about 50%); there- 
fore, a low threshold should exist for surgical 
repair. Operation is by aneurysm ligation, or 
in suitable cases coil embolization may be 
appropriate. 

Jejunal, Ileal, and Colic Artery Aneurysms 

Together these aneurysms constitute 2% to 
3% of all visceral aneurysms. Sex distribution 
appears to be equal, with an average age at pres- 
entation of 75 years. These aneurysms tend to 
be small, isolated, and asymptomatic until 
rupture. Diagnosis is invariably made at sub- 
sequent laparotomy, where repair is by either 
aneurysm ligation or excision. Concomitant 
limited bowel resection may have to be per- 
formed if the vascular repair threatens an 
inadequate intestinal blood supply. 

Axillary and Subclavian 
Artery Aneurysms 

Aneurysmal disease affecting the upper extrem- 
ity is rare but remains of importance due to the 
severity of possible sequelae associated with the 
disease. Ideal management, therefore, aims at 
early diagnosis and repair in order to minimize 
this potential morbidity. Pathology in this 
region is divided anatomically into one of three 
types, each with a distinct etiology. 

Subclavian Artery Aneurysms 

Subclavian aneurysms (SCAs) are arbitrarily 
classified as proximal or distal in reference to 
the location of diseased arterial segment. Prox- 
imal aneurysms are totally confined to the 
parent artery and are usually caused by degen- 
erative disease, trauma, or infection. They occur 
more commonly in men and over the age of 60 
years. In contrast, distal subclavian aneurysms 
tend to occur in the younger female population. 
Clinical features associated with SCA can be 
considered as local or distant. The patient may 
complain of a painless pulsatile mass in the 
lower neck, whereas acute aneurysm expansion 
or rupture causes pain experienced in the chest, 
neck, or shoulder. Local symptoms due to com- 
pression are common and include neuropathic 
pain (which may be referred) due to brachial 



plexus irritation, an ipsilateral Horner syn- 
drome from sympathetic chain compression, 
hoarseness due to recurrent laryngeal nerve 
pressure, stridor due to partial extrinsic tra- 
cheal obstruction, and upper limb edema 
caused by venous outflow obstruction. Pul- 
monary erosion by the aneurysm explains any 
hemoptysis, whereas distant manifestations of 
the condition can include acute or chronic limb 
ischemia from thromboembolism and Raynaud 
syndrome. Examination may reveal a supraclav- 
icular pulsatile mass with an audible bruit. An 
assessment of full upper limb neurovascular 
status is mandatory, which classically reveals 
normal pulses in the evidential face of prior 
microembolism and altered neurology in the 
corresponding segment of brachial plexus or 
sympathetic chain. 

Duplex or CT scanning confirms the diagno- 
sis of SCA, but angiography is also required to 
plan appropriate intervention. Proximal SCA 
management is by resection with end-to-end 
anastomosis for (small aneurysms) or by surgi- 
cal resection with interposition arterial graft. 
Proximal vascular control can be difficult, 
and occasionally sternotomy (right SCA) or 
extended thoracotomy (left SCA) is required. 
Surgical correction of the distal SCA adheres to 
the same principles as for proximal pathology, 
with the advantage that adequate exposure is 
achieved by simple supraclavicular incision. 

Axillosubclavian Artery Aneurysms 

These aneurysms involve the junction between 
the terminal subclavian and proximal axillary 
arteries at the border of the first rib. They are 
invariably poststenotic aneurysms from a tho- 
racic outlet syndrome caused by either a cervi- 
cal rib or fibrous band. The disease is more 
common in younger females, probably because 
it is this population that has the highest inci- 
dence of cervical rib. 

Symptoms are similar to those outlined for 
isolated subclavian aneurysms, but it is worth 
noting that there is an increased frequency of 
embolic phenomena. Investigation of axillosub- 
clavian disease is initially by duplex ultrasonog- 
raphy, but arteriography is advised for all cases 
to be considered for surgery. Exact management 
depends on both aneurysmal size and the pres- 
ence of distal thromboembolic complications. 
Large (defined as more than twice the arterial 




> 



218 



VASCULAR SURGERY 



diameter) or symptomatic aneurysms require 
arterial repair by means of excision and graft 
interposition with synchronous removal of the 
antecedent cause (e.g., excision of cervical rib or 
division of fibrous band). Small, asymptomatic 
aneurysms may well recede once the thoracic 
outlet syndrome has been surgically corrected 
so that arterial reconstruction is often not 
required. 

Axillary Artery Aneurysms 

Isolated axillary artery aneurysms are very rare 
but typically affect young males as a conse- 
quence of trauma. Repeated blunt trauma to 
the axillary artery, classically due to long-term 
crutch-use, may result in aneurysm develop- 
ment and the subsequent complicating symp- 
toms of acute or chronic upper limb ischemia. 
Penetrating trauma tends to result in false 
aneurysm formation. A rich collateral blood 
supply in this region may lead to delayed and 
atypical presentations, so a high index of suspi- 
cion is needed to make the diagnosis. 

Investigation is by duplex ultrasound fol- 
lowed by arteriography, and appropriate 
surgery is aneurysm resection and arterial 
reconstruction by interposition autogenous 
vein graft (saphenous vein). Prosthetic grafts 
have been successfully used but are associated 
with inferior long-term patency rates. Recently, 
successful management with axillary endovas- 
cular stent grafts has been reported, but the 
long-term results of this intervention are 
presently unknown. 

Carotid Artery Aneurysms 

Extracranial carotid aneurysmal disease requir- 
ing vascular surgical expertise is extremely rare. 

True carotid aneurysms are located either in 
the carotid bifurcation, internal carotid system 
or external carotid, in decreasing frequency of 
that order. Etiological factors are varied, but the 
most common include atherosclerosis, trauma, 
previous carotid surgery (e.g., endarterectomy), 
and carotid artery dissection. Historically, 
infection used to be the major culprit causing 
disease, but widespread antibiotic availability 
has rendered this mechanism responsible in 
only a minority of cases. 

The patient may present with an awareness of 
a pulsatile mass in the neck that may or may not 



be painful. Other symptoms are attributed to 
local compression and include dysphagia as a 
result of pharyngeal proximity, deafness or 
facial pain due to cranial nerve compression, 
and hoarseness caused by vagus nerve impinge- 
ment. Distal central nervous neurological symp- 
toms such as stroke, amaurosis fugax, or 
transient ischemic attack (TIA) indicate throm- 
boembolic complications from the carotid 
aneurysm and are in fact the most common pre- 
senting feature of the disease. Large aneurysms 
have also been implicated in posture-related 
transient neurology as a result of limitation and 
even occlusion of blood flow through the neigh- 
boring internal carotid artery. Ruptured carotid 
aneurysms are fortunately rare but are usually 
fatal if they occur. 

It is interesting that despite the paucity of 
the disease, carotid aneurysms remain over- 
diagnosed. True aneurysms must be distin- 
guished from the much commoner reality of 
tortuous or coiled carotid vessels, and other 
diagnoses such as cervical lymphadenopathy, 
carotid body tumors, and branchial cysts. In the 
majority of cases history and clinical examina- 
tion alone should be sufficient to expose the 
bona fide aneurysm, but confirmation can be 
obtained with duplex or CT scanning. If inter- 
vention is being contemplated, an angiogram 
must also be obtained. 

The aim of management is ultimately pre- 
vention of the neurological complications 
associated with carotid aneurysmal disease. 
Current opinion regarding preferred treatment 
is divided between surgical and endovascular 
methods. In the former, the aneurysm is 
resected, arterial reconstruction is performed, 
and cerebral blood flow is maintained during 
carotid clamping with an operative shunt. The 
adequacy of this maneuver is assessed by some 
surgeons with the use of intraoperative tran- 
scranial Doppler or electroencephalography in 
order to confirm distal perfusion. Surgery is 
notoriously perilous in proximal internal 
carotid disease and in larger aneurysms where 
"distal" control can be difficult to secure. The 
most feared complication of carotid aneurysm 
surgery is disabling stroke, occurring in 6% to 
10% of cases, which may result from cerebral 
hypoperfusion or intraoperative dislodgment of 
debris from the vessel wall. Another specific risk 
of surgery is cranial nerve palsy, resulting from 
local tissue handling or careless dissection. The 



219 



ANEURYSMAL DISEASE 




condition affects 20% of patients and is usually 
temporary, but must nonetheless be mentioned 
to the patient preoperatively while obtaining 
informed consent. 

Endovascular approaches to carotid 
aneurysm management may soon render 
surgery as second-line treatment for extracra- 
nial carotid aneurysmal disease. Graft stenting 
and coil embolization of such lesions is reported 
to be as effective as surgical repair, without 
the need for operation. These techniques may 
be especially appropriate in large aneurysms, 
revisional surgery, or the case of high carotid 
aneurysm close to the base of skull so that 
access is problematic. The risk of neurological 
complications following the procedures re- 
mains, but these cerebrovascular events appear 
less common in this approach to treatment. Still 
in its infancy, these initial encouraging results 
may be tempered as we await the longer term 
results of endoluminal intervention. 



Controversial Issues 

• Should there be a national screening 
program for the detection of AAA? 

• Are all elective AAA repairs best per- 
formed at tertiary referral centers? 



Is open repair preferable to endovascular 
therapy in elective AAA repair? 
What is the optimal management of type I 
and type III endoleaks? 
Does routine uterolysis have a role in the 
management of the inflammatory AAA? 
Do all high (Crawford types I and II) tho- 
racoabdominal aneurysm repairs mandate 
the use of cardiopulmonary bypass and 
spinal fluid drainage? 
What is the best treatment for the asymp- 
tomatic popliteal aneurysm? 
When should carotid aneurysms be 
repaired with endovascular methods? 



References 



Dawson I, Sie RB, van Bockel JH. (1997) Br J Surg 84: 

293-9. 
Hallin A, Bergqvist D, Holmberg L. (2001) Eur J Vase 

Endovasc Surg 22:197-204. 
Heller JA, Weinberg A, Arons R, et al. (2000) J Vase Surg 

32:1091-100. 
Rasmussen TE, Hallett JW Jr. (1997) Ann Surg 225:155-64. 
Wassef M, Baxter BT, Chisholm RL, et al. (2001) J Vase Surg 

34:730-8. 
Wilmink AB, Quick CR. (1998) Br J Surg 85:155-62. 
Woodburn KR, May J, White GH. (1998) Br J Surg 85: 

435-43. 



18 



Renovascular Hypertension and 
Ischemic Nephropathy 

Sherry D. Scovell 




Renovascular hypertension is a relatively 
uncommon cause of hypertension and is only 
seen in 5% to 10% of the hypertensive popula- 
tion. However, this translates to at least 600,000 
people in the United States alone when consid- 
ering that nearly 60 million people in the United 
States have some degree of hypertension. Renal 
artery stenosis (RAS) often produces an unclear 
clinical picture. Patients maybe asymptomatic. 
However, they may also present with severe, 
uncontrolled hypertension referred to as reno- 
vascular hypertension or with evidence of renal 
insufficiency, otherwise known as ischemic 
nephropathy. This chapter focuses on the clini- 
cal characteristics that may be helpful in iden- 
tifying those patients who may be at risk for 
RAS, how to accurately diagnose RAS, and how 
to correlate RAS with the symptoms of uncon- 
trolled hypertension or ischemic nephropathy. 
It also outlines the options available for treat- 
ment, including medical management, endo- 
vascular correction of RAS via angioplasty 
with or without stenting, and open surgical 
revascularization. 



Characteristics 

With renovascular disease being responsible 
for only 5% to 10% of the hypertensive popu- 
lation, it would be helpful to define certain 
clinical characteristics that are prevalent in 
this population to aid in screening patients. 
Typically, renovascular hypertension produces 



severe diastolic hypertension, which is defined 
as diastolic blood pressure greater than 115mm 
Hg. In a blood pressure screening program 137 
patients with new-onset hypertension were 
diagnosed in a shopping center. Further workup 
consisted of angiography followed by renal 
vein renin measurements and split renal func- 
tion tests in patients with evidence of critical 
renal artery stenosis. None of the 102 patients 
with a diastolic blood pressure between 90 
and 115mmHg had evidence of renovascular 
hypertension. However, nine of the 35 patients 
(26%) with a diastolic blood pressure of 115 mm 
Hg or higher did have evidence of renovascular 
hypertension. With respect to race, none of 
the African-American patients had renovascu- 
lar hypertension, whereas nine of the 22 (41%) 
Caucasian patients did have renovascular 
hypertension. 

Renovascular hypertension has also been 
described as having a bimodal age distribution, 
with patients at the two extremes of age carry- 
ing the highest incidence of having renovascu- 
lar disease responsible for their hypertension. 
It is typically seen in children younger than 5 
years old and in patients over 60 years of age. 

Based on observations such as these, addi- 
tional characteristics thought to be associated 
with renovascular hypertension were postulated 
to include recent onset of hypertension, young 
age, lack of family history of hypertension, and 
the presence of an abdominal bruit. The most 
comprehensive study used to compare these 
characteristics in hypertensive patients both 



221 




> 



222 



VASCULAR SURGERY 



with and without RAS was the Cooperative 
Study of Renovascular Hypertension (Simon, 
1972). One of the ultimate conclusions of this 
study was that neither the above four criteria 
nor any other clinical criteria could accurately 
predict the presence of renovascular hyperten- 
sion. Indeed, the majority of patients with ren- 
ovascular hypertension do not present with 
what were once thought to be the "classic" char- 
acteristics delineated above. As a result of these 
studies, it is necessary to maintain a high index 
of suspicion in patients whose hypertension is 
difficult to control with medical management 
and to rely on screening examinations in 
patients with severe diastolic hypertension. 



Etiology 



The etiology of RAS is primarily atherosclerotic 
in approximately 90% of patients. The second 
most common etiology is fibromuscular dyspla- 
sia (FMD). Other less common causes of RAS 
include aortic dissection, vasculitis, emboli, 
radiation, and extrinsic compression from 
masses or tumors. 

Atherosclerotic renal artery stenosis is seen 
predominantly in males. The majority of these 
lesions are believed to be ostial encroachment 
of an aortic plaque on the orifice of the renal 
artery or arteries. As a result, 50% of the time 
these lesions are bilateral. As well, these lesions 
are typically eccentric and irregular luminal 
stenoses. It is well known that atherosclerotic 
disease is progressive and the renal artery in not 
an exception. RAS will be progressive in 30% to 
70% of cases. Approximately 11% of the time, 
patients with a greater than 60% stenosis of the 
renal artery will progress to total occlusion 
within a 2-year period (Zierler, 1994). 

The stenoses caused by FMD are primarily 
nonostial and often extend into the branches 
of the renal arteries. Fibromuscular dysplasia 
may be divided into three types: medial fibro- 
dysplasia, perimedial dysplasia, and intimal 
fibroplasia. 

Medial fibro dysplasia is the most common 
type, accounting for 85% of all FMD lesions. 
Women are predominantly affected, usually 
between 25 and 45 years of age. In 55% of 
patients, the disease is bilateral. If it is unilateral, 
it is more likely to affect the right renal artery 
when compared to the left renal artery. The 



angiographic appearance is that of a string of 
beads. There are two distinct types of medial 
fibrodysplasia — diffuse and peripheral. 

Perimedial dysplasia is seen in only about 
10% of patients with FMD. This subtype is again 
seen most often in women in the fourth or fifth 
decades of life. This type may be more progres- 
sive when compared to medial fibrodysplasia. 

Finally, intimal fibroplasia is seen in 5% of 
patients with FMD. These lesions are long, irreg- 
ular, tubular regions of stenosis. They tend to 
progress more rapidly than those caused by 
medial fibrodysplasia. 



Pathophysiology 



Any comprehensive review of renovascular 
hypertension must cite Goldblatt's (1934) classic 
experiments. He described both the two-kidney, 
one-clip model of unilateral RAS in the setting 
of a normal contralateral kidney, as well as the 
one-kidney, one-clip model that represents uni- 
lateral RAS in a patient with only one kidney. 

In the two-kidney, one-clip model, one renal 
artery is clamped while the other one is left 
open. In this model, there is decreased renal 
blood flow seen by the ipsilateral juxtaglomeru- 
lar apparatus. This leads to increased secretion 
of renin by the clipped kidney. Through the 
renin-angiotensin-aldosterone system, there is 
mild sodium retention as a result of increased 
aldosterone and increased blood pressure sec- 
ondary to angiotensin II-related vasoconstric- 
tion. The contralateral kidney then suppresses 
its release of renin to compensate. When an 
angiotensin-converting enzyme (ACE) inhibitor 
is administered to a patient in the two-kidney, 
one-clip model acutely, there is a dramatic 
reduction in blood pressure. This demonstrates 
that the elevation in blood pressure is primarily 
angiotensin II-dependent. 

In the one-kidney, one-clip model of reno- 
vascular disease, a clip is applied to the renal 
artery supplying a single kidney. In this case, 
there is an initial elevation in renin. This initial 
rise in the renin leads to activation of the renin- 
angiotensin-aldosterone system, and there is 
peripheral vasoconstriction as well as an 
increase in sodium and water retention. Blood 
pressure rises as a result of both of these mech- 
anisms. This model is both an angiotensin- 
dependent system as well as a volume-driven 



223 



RENOVASCULAR HYPERTENSION AND ISCHEMIC NEPHROPATHY 




system and produces a more persistent form of 
hypertension. 

Diagnostic Evaluation 

The patients who are well served by renal revas- 
cularization are those with severe RAS in the 
setting of either uncontrolled hypertension or 
ischemic nephropathy. Patients without an asso- 
ciation between the anatomical stenosis and 
these symptoms are not well served by correc- 
tion of the RAS. Therefore, it is necessary to 
establish a correlation between the anatomical 
RAS and the uncontrolled hypertension or 
ischemic nephropathy to determine who will 
benefit from an attempt at revascularization. 
This is done primarily through the combination 
of both anatomical screening tests as well as 
studies that document associated physiological 
consequences. Anatomical tests are those that 
delineate RAS and document associated hemo- 
dynamic data. Physiological tests attempt to 
establish an association between the anatomical 
stenosis and the alterations in the renin- 
angiotensin-aldosterone axis. 

Anatomical Tests 

Duplex Ultrasound 

Duplex examination of the kidneys and the 
renal arteries is an extremely useful screening 
modality as it is a noninvasive examination. The 
only preparation necessary is an overnight 
fast, and it is not necessary to discontinue anti- 
hypertensive medications for reliable results. 
Renal artery duplex examination is able to iden- 
tify hemo dynamically significant renal artery 
lesions. It offers information on renal length 
as well as on the renal artery blood flow. The 
resistive index (RI) indicates the degree of 
renal artery resistance and should be calculated. 
It is defined as the peak systolic shift minus 
the minimum diastolic shift over the peak sys- 
tolic shift. A normal RI is less than 0.70. The RI 
may be predictive of outcome with respect 
to renal revascularization (Radermacher et al., 
2001). However, caution should be taken in 
the interpretation of RI as it may also be in- 
creased in patients with intrinsic renal disease, 
decreased cardiac output, or perinephric fluid 
collections. 



The degree of RAS is typically calculated 
based on the renal-to-aortic ratio (RAR) as well 
as the peak systolic velocities in the renal artery. 
The RAR is the ratio of the peak systolic veloc- 
ity of the renal artery to the aortic peak systolic 
velocity. If the RAR is less than 3.5 with a peak 
systolic velocity of greater than 180 cm/sec, 
there is a less than 60% stenosis. If the RAR is 
greater than 3.5 and the peak systolic velocity 
is greater than 180 cm/sec, this is diagnostic 
of a greater than 60% stenosis. If there is no 
detectable renal artery signal with a kidney 
length of less than 9 cm, the renal artery is con- 
sidered occluded. 

Although this is an excellent screening test 
to detect the presence of renal artery stenosis, 
duplex examination is unable to accurately 
predict the clinical response of either the hyper- 
tension or renal insufficiency following correc- 
tion of the RAS. It is also unable to accurately 
identify all accessory branches of the renal 
artery and their contribution to the clinical 
picture. This is, however, an appropriate nonin- 
vasive screening test to evaluate for RAS and is 
useful for following the patient with serial 
examinations after intervention. 

Angiography/Digital 
Subtraction Angiography 

The gold standard for the identification of RAS 
still remains angiography, although it is an inva- 
sive test and not an optimal screening tool. It 
serves to define the presence, severity, and loca- 
tion of intraluminal anatomical defects in the 
renal artery. It facilitates a full evaluation of 
the abdominal aorta as well as the renal artery 
orifices and the accessory branches of the renal 
artery. Multiple accessory renal arteries exist in 
up to 25% of patients and are well defined with 
angiography. It is able to define a nephrogram. 
It is still controversial whether angiography 
should be utilized as a screening test for reno- 
vascular disease. It is likely best used as an 
adjunct when duplex examination is unable to 
visualize the renal arteries, or smaller accessory 
branches are suspected of contributing to severe 
renovascular hypertension. 

Technically, the renal arteries usually arise 
posterolaterally from the aorta and require 
oblique views for adequate visualization of their 
orifices. Peak systolic pressure gradients across 
the lesion of >20mmHg or mean gradients of 




> 



224 



VASCULAR SURGERY 



10 mm Hg have been used as objective criteria to 
define a significant reduction in blood flow. 

It has been well documented that angiogra- 
phy may exacerbate renal failure in patients 
with baseline renal insufficiency. Nonionic, low- 
osmolar contrast material is recommended 
for the evaluation of the renal arteries and is 
associated with a lower incidence of contrast- 
induced nephropathy (Barrett and Carlisle, 
1993). In patients with an elevated creatinine, 
C0 2 or gadolinium may be substituted without 
an increase in contrast-induced nephropathy. As 
well, preintervention hydration is critical as is 
acetylcysteine in select patient groups. 

The calculation of R AS is determined by com- 
parison of the narrowest segment of the renal 
artery to the normal diameter of the renal artery 
either proximal to the stenosis or distal to any 
region of poststenotic dilatation. Poststenotic 
dilatation is common in many cases of long- 
standing arterial stenosis. 

Patient Selection 

Renal artery stenosis alone, in the absence of 
symptoms, does not mandate repair. The coex- 
istence of RAS and hypertension does not 
establish a causal relationship. For this reason, 
it is essential to determine the functional 
significance of the anatomical stenosis. 

Patients with RAS and symptoms attributable 
to that stenosis are considered for treatment and 
correction of the anatomical lesion. Symptoms 
of hypertension, renal insufficiency, or pul- 
monary edema in the setting of a greater than 
60% RAS serve as criteria for repair. Clinical cri- 
teria for revascularization have been defined 
recently in the Guidelines for Reporting on 
Renal Artery Revascularization in Clinical Trials 
(Rundback, 2002). Based on these guidelines, 
hypertension was classified as either accelerated 
with sudden worsening in the setting of previ- 
ous control, refractory hypertension resistant to 
medical management with three or more anti- 
hypertensive medications including a diuretic, 
or malignant hypertension with evidence of 
severe end-organ damage as a result. The pres- 
ence of hypertension in the setting of a unilat- 
erally shrunken kidney was also used as an 
indication for intervention. With respect to 
renal insufficiency, the clinical criteria for inter- 
vention were defined as unexplained worsening 
of renal function or a decrease in renal function 



with the addition of an ACE inhibitor, as well as 
renal dysfunction not attributable to another 
obvious etiology. Finally, recurrent episodes 
of "flash" pulmonary edema not able to be 
explained on the basis of severe left ventricular 
dysfunction in the setting of RAS was also con- 
sidered an indication for revascularization. The 
value of prophylactic renal artery revasculariza- 
tion in asymptomatic patients is unproven at 
this point, and that is why physiological tests are 
significant in these patients. 

Physiological Tests 

When a unilateral RAS is defined by anatomical 
studies, the functional significance must be 
determined. In the past, many different tests 
were attempted in order to define this correla- 
tion including intravenous urography, nuclear 
medicine studies, conventional angiography, 
computed tomography, and magnetic resonance 
imaging. More recently, this is accomplished 
through the use of renal vein renin assays or by 
isotope renography. The value of these tests is 
not as great in patients with severe bilateral 
renal disease or RAS in a solitary kidney when 
compared to unilateral renal disease and a func- 
tioning contralateral kidney. 

Physiologically, there is a functional increase 
in plasma levels of renin from the affected 
kidney, which is characteristic in the setting of 
RAS. Originally, systemic levels of renin were 
obtained in an attempt to establish a diagnosis. 
However, utilizing this method, the rate of false- 
negative and false-positive results were high, 
43% and 34%, respectively, in patients with 
confirmed renovascular hypertension. As well, 
roughly 20% of patients with essential hyper- 
tension had an elevated plasma renin level. The 
value of selective renal vein sampling of renin, 
although an invasive procedure, has been found 
to be more sensitive and specific. This test com- 
pares the renin levels between the ipsilateral 
and contralateral kidneys. If the renal vein/renin 
ratio is greater than 1.5, there is lateralization of 
renin secretion. This, as well, seems to be pre- 
dictive of improvement following renal revascu- 
larization. However, the failure to demonstrate 
lateralization does not reliably predict failure of 
revascularization procedures. In addition, there 
are unfortunately strict prerequisite guidelines 
required prior to performance of this test. Anti- 
hypertensive medications must be held for up to 



225 



RENOVASCULAR HYPERTENSION AND ISCHEMIC NEPHROPATHY 




3 weeks prior to the test, which is not without 
clinical consequences. 

For these reasons, other physiological tests 
became more favorable. Initially, isotope renog- 
raphy was developed as a screening procedure 
for renovascular hypertension. The theory was 
that the underperfused kidney would have 
delayed uptake and excretion of solute when 
compared to the normal, well-perfused con- 
tralateral kidney. Various nucleotides were uti- 
lized; however, this test still yielded a high rate 
of false-negative results. More recently, the 
results of this functional test have improved 
through the use of the ACE inhibitor captopril. 
Angiotensin II is elevated in patients with RAS, 
and this elevation leads to selective vasocon- 
striction of the efferent arterioles in an attempt 
to maintain glomerular filtration. The use of 
captopril blocks angiotensin II production, and 
thus filtration drops precipitously. The finding 
of a decrease in the uptake and excretion of the 
tracer is more pronounced in patients after cap- 
topril in the setting of clinically significant RAS. 
Through the use of captopril, this study has 
most recently demonstrated a 92% sensitivity 
and a 94% specificity. 



Therapeutic Options 

The options for the treatment of symptomatic 
RAS include medical management utilizing 
multiple antihypertensive medications, the 
endovascular approach including angioplasty 
with or without the placement of a stent, or open 
surgical management. 

Medical Management of 
Symptomatic Renal Artery Stenosis 

The medical management of symptomatic RAS 
is accomplished using a variety of antihyper- 
tensive medications in an attempt to control the 
hypertension. Control of the hypertension in 
itself is critical, as uncontrolled hypertension is 
a major risk factor for myocardial infarction, 
cerebrovascular accident, and chronic renal 
insufficiency. Thus, the goals of medical man- 
agement of symptomatic RAS are to reduce the 
blood pressure, to control cardiovascular risk 
factors, and ultimately to prevent end-organ 
damage from sustained hypertension. 



However, the natural history of uncorrected 
RAS is not benign. It has previously been docu- 
mented that 11% of patients with a greater than 
60% stenosis of the renal artery progress to 
occlusion within a 2-year period. As well, over 
40% of patients with a critical stenosis of the 
renal artery will have a decrease in glomerular 
filtration by greater than 25% with progressive 
deterioration in renal function being common 
in this group of patients. 

As a result of these findings, there have been 
numerous studies designed to evaluate medical 
management for symptomatic RAS compared to 
both open surgical repair as well as endovascu- 
lar management. 

Hunt and Strong (1973) orchestrated a com- 
parative analysis examining drug therapy com- 
pared to operative therapy to treat symptomatic 
RAS; 114 patients were in the medical treatment 
arm and 100 patients were in the surgical treat- 
ment arm. The follow-up was over the course of 
7 to 14 years. Overall, 84% of patients were alive 
in the surgical group compared to 66% alive in 
the group treated medically. Of those alive in the 
surgical group, 93% were cured or significantly 
improved, whereas of those patients in the 
medical treatment group, 21% subsequently 
required surgical intervention for uncontrolled 
hypertension. Another seven patients from that 
group continued to have uncontrolled hyper- 
tension but were not taken to surgery. In addi- 
tion, death was twice as common in the 
medically treated group. These differences were 
statistically significant (p < .01) for both ather- 
osclerotic and FMD lesions. This study demon- 
strated that in patients with renovascular 
hypertension, there is a need to treat sympto- 
matic lesions surgically. Indeed, in this series, 
surgical intervention offered better sympto- 
matic relief of hypertension, which translated to 
a lower incidence of end-organ damage. 

Webster and colleagues (1998) performed the 
first prospective randomized trial comparing 
medical management with renal artery angio- 
plasty in 55 patients. All patients were hyper- 
tensive and showed evidence of >50% RAS on 
angiography. They found that only patients with 
bilateral RAS demonstrated a significant 
decrease in blood pressure with angioplasty. 
There was no significant improvement in renal 
excretory function. Otherwise, there was no sta- 
tistical difference between the two groups. This 
study suggested that there might be a significant 




> 



226 



VASCULAR SURGERY 



benefit to be derived from the endovascular 
management of symptomatic RAS over medical 
management of these patients, which ultimately 
led to the validation of this therapeutic option. 
Based on the results of the above studies as 
well as other smaller studies, it became clear 
that intervention, either endovascular or open 
surgical, should play a significant role in the 
treatment of symptomatic RAS. 

Endovascular Repair of Renal 
Artery Stenosis 

In addition to open surgical repair for sympto- 
matic RAS, renal artery angioplasty with or 
without stenting offers a less invasive method to 
correct the anatomical stenosis. Although still a 
matter of great debate, it is clear that angio- 
plasty with or without stenting may offer at least 
short-term improvement in blood pressure 
and, in certain circumstances, renal function. 
Although this option is especially appealing for 
patients, it is certainly necessary to further eval- 
uate and better define what the role of endovas- 
cular management of symptomatic RAS should 
be through a multi-institutional randomized, 
prospective trial. 

There is much debate regarding the need to 
place a stent in the renal artery compared to 
angioplasty alone. Typically, renal artery angio- 
plasty alone is utilized for fibrodysplastic 
lesions and lesions that do not involve the origin 
of the renal artery. However, as mentioned pre- 
viously, RAS that is atherosclerotic in etiology is 
often an extension of aortic plaque. For this 
reason, angioplasty alone is not as durable. It 
has been demonstrated that atherosclerotic 
lesions of the renal artery seem to have lower 
rates of re-stenosis following angioplasty and 
placement of a stent for this reason. In addition, 
it is clear that if there is a residual stenosis fol- 
lowing initial angioplasty or evidence of dissec- 
tion, a stent should be placed at that time. 

There has been some concern that renal 
artery angioplasty and stenting may make oper- 
ative intervention difficult or impossible sec- 
ondary to postangioplasty periarterial fibrosis. 
This is especially true for transaortic renal 
endarterectomy. In the case of a previously 
placed renal stent placement, renal artery 
bypass grafting to a site on the renal artery 
distal to the stent may be the best option. 



However, in certain cases, endarterectomy may 
still be feasible (Pak, 2002). 

Although it is widely practiced at most insti- 
tutions, renal artery angioplasty and stenting is 
a topic that is still the subject of modest debate. 
There are only a few prospective randomized 
controlled trials that compare medical therapy, 
endovascular management, and open surgical 
repair of symptomatic RAS. 

Weibull and associates (1993) prospectively 
and randomly assigned 58 patients with symp- 
tomatic RAS to either renal angioplasty or sur- 
gical renal revascularization. Percutaneous 
transluminal angioplasty (PTA) was initially 
technically successful 83% of the time, whereas 
surgical management was successful 97% of the 
time. The primary patency rate after 2 years was 
75% in the PTA group compared to 96% in the 
surgical group. Secondary patency was 90% 
versus 97%, respectively, in the PTA and surgi- 
cal patients. Hypertension was improved in 90% 
after PTA and 86% following open surgical renal 
revascularization, and improvement in renal 
function was seen in 83% and 72%, respectively. 
The authors concluded that PTA is a reasonable 
option when compared to surgical revascular- 
ization. Indeed, the primary patency was lower 
in the endovascular group, but the secondary 
patency was comparable. Patients treated with 
angioplasty of RAS clearly require close surveil- 
lance and often subsequent procedures to main- 
tain patency. 

Martin and colleagues (2003), as well, have 
demonstrated a 30% incidence of re-stenosis 
following initially technically successful renal 
artery angioplasty. This high incidence of re- 
stenosis with respect to renal artery ostial 
lesions is likely due to the fact that these lesions 
represent extension of an aortic plaque. The 
idea of placing stents in the renal arteries as a 
method of preventing re-stenosis was intro- 
duced by Palmaz in 1976, initially in animal 
models and subsequently in humans. Currently, 
the indications for the placement of a stent in a 
renal artery following renal artery angioplasty 
include substantial elastic recoil, resistance of 
the plaque, or dissection. 

Bush and colleagues (2001) retrospectively 
examined the results of renal artery stenting in 
73 consecutive patients over a 7-year period. In 
this study, the majority of stents were placed 
for residual stenosis following angioplasty or 
dissection. The technical success rate was 



227 



RENOVASCULAR HYPERTENSION AND ISCHEMIC NEPHROPATHY 




89%, with one patient requiring thrombolytic 
therapy for intrastent thrombus. The complica- 
tion rate was 9.1% with renal artery thrombo- 
sis, extravasation, and hematoma. In this study, 
the authors noted a significant decrease in both 
systolic and diastolic blood pressures (p < .001) 
as well as a decrease in the number of medica- 
tions required to treat the hypertension (p < 
.01). Over the course of an average of 20 months' 
follow-up, serum creatinine levels decreased 
by more than 20% in 22% of patients. It 
remained unchanged in 48% of patients and 
deteriorated in 25% of patients. Of the 25% of 
patients who had an increase in serum creati- 
nine, 12% eventually required hemodialysis. 
Five of these patients had a preoperative creati- 
nine level over 4.0mg/dL. Over the course of 
approximately 11 months' follow-up, 10 patients 
had evidence of re-stenosis in a total of 14 renal 
arteries. Overall, this retrospective study estab- 
lished the safety of endovascular management 
with respect to these lesions as an alternative to 
open surgical repair. 

Attempts to validate the endovascular man- 
agement of symptomatic RAS seem to be 
beneficial, at least in the short term. This 
method of treatment, although less invasive 
than open surgical repair, appears not to be as 
durable over the long term. This group of 
patients needs close post-angioplasty and stent 
surveillance to detect re-stenosis and subse- 
quent intervention to have results that are com- 
parable to open surgical repair. However, this 
still remains an option that is feasible for the 
treatment of these lesions and attractive for the 
patients. It is hoped that through the use of a 
well-constructed trial, more data will be avail- 
able to formulate more solid conclusions on 
angioplasty and stenting of symptomatic renal 
artery stenosis. 

Open Surgical Renal 
Revascularization 

Open surgical repair or reconstruction of symp- 
tomatic renal artery lesions still remains the 
gold standard in treatment. Options for direct 
open surgical repair include aortorenal bypass, 
reimplantation of the renal artery, and throm- 
boendarterectomy Indirect renal revasculariza- 
tion may be accomplished via splenorenal or 
hepatorenal artery bypass. This option is espe- 



cially useful because aortic cross-clamping may 
be avoided. The conduits available for use 
include autologous vein grafts, synthetic polyte- 
trafluoro ethylene (PTFE) and Dacron pros- 
thetic grafts, and autologous hypogastric artery. 
Nephrectomy, as well, may be considered in 
patients with unreconstructable renal disease 
with a nonfunctioning ipsilateral kidney. 

The operative approach may be through a 
midline xiphoid to pubis incision, a supraum- 
bilical transverse incision, an extended flank 
incision, or a subcostal incision. The midline 
approach is useful for atherosclerotic lesions as 
well as lesions combined with aortic proce- 
dures, whereas the subcostal or flank incisions 
are efficacious for fibrodysplastic lesions or 
splanchnorenal bypass. When combined with 
mesenteric artery bypass grafting, the extended 
flank incision may be useful. Thromboen- 
darterectomy may be easily accomplished via a 
retroperitoneal trapdoor approach. 

With respect to exposure of the distal renal 
artery, the left renal artery lies posterior to the 
left renal vein. The vein must be sufficiently 
mobilized with division of the gonadal and 
adrenal veins. The renal vein may be retracted 
either cephalad or caudad to expose the renal 
artery. There is also a lumbar vein that enters the 
posterior aspect of the renal vein that may be 
easily avulsed if not carefully identified and 
ligated. Distal exposure of the renal artery is 
essential to clearly identify the extent of the 
atheroma, and the renal artery should be dis- 
sected free distal to the atherosclerotic plaque. 
Silastic loops may be used for distal arterial 
control, as they are less traumatic compared to 
metal clamps. 

The aortic anastomosis is typically per- 
formed utilizing an aortotomy that is two to 
three times the diameter of the conduit. The 
anastomosis is usually placed on the anterolat- 
eral aspect of the aorta. It is essential to prevent 
the graft from kinking. For right-sided revascu- 
larizations, the aortorenal graft is typically 
placed in a retrocaval position, although this 
must be individualized. With respect to left- 
sided revascularizations, the graft is usually 
placed beneath the left renal vein. 

Subsequently, the renal anastomosis is com- 
pleted. This is typically anastomosed in an end- 
to-end fashion with spatulation of both the graft 
and renal artery. Spatulated anastomoses are 
ovoid and less prone to the development of 




> 



228 



VASCULAR SURGERY 



strictures. Intraoperative evaluation of blood 
flow is typically accomplished using a direc- 
tional Doppler device. Intraoperative angiogra- 
phy is usually unnecessary; however, many 
surgeons advocate baseline angiography prior 
to discharge to establish a baseline and to 
confirm the adequacy of repair. 

Hansen and colleagues (1992) retrospectively 
reviewed the data on 200 patients who under- 
went open surgical renal revascularization over 
a 54-month period. There was a mortality rate 
of 2.5% and only a 1.4% primary failure rate at 
30 days. In this series, the hypertension was 
cured in 21% of patients and improved in 
another 70%. Of the patients with ischemic 
nephropathy, 49% demonstrated an improve- 
ment in glomerular filtration; 36% of these 
patients remained stable and only 15% wors- 
ened. This study clearly demonstrates that open 
surgical renal revascularization is a safe option 
that is successful in treating the underlying clin- 
ical manifestations of the RAS in the majority of 
patients. 

Recently, Cherr and associates retrospectively 
reviewed the clinical outcome of 500 consecu- 
tive patients with renovascular hypertension. 
The perioperative mortality was 4.6% at 30 days. 
The hypertension was cured in 12%, improved 
in 73%, and unchanged in only 15%. With 
respect to ischemic nephropathy, 43% had an 
improvement in renal function, 47% were 
unchanged, and 10% became worse. Interest- 
ingly, of the 43% of patients who demonstrated 
an improvement in renal function, 28 patients 
were removed from dialysis dependence. These 
data confirm the findings of Hansen and asso- 
ciates and once again demonstrate that open 
surgical repair of severe, flow-limiting, sympto- 
matic RAS is efficacious in improving hyper- 
tension and ischemic nephropathy in the 
majority of patients. It adds the point, however, 
that if there was a blood pressure cure or an 
improvement in renal function, there was an 
association with dialysis-free survival. This has 
a significant impact on patient lifestyle as well 
as on health care dollars. 

At this time, there is significant controversy 
regarding the optimal treatment in patients 
with symptomatic RAS. It is important that a 
multicenter, prospective, randomized clinical 
trial be organized in an effort to further define 
the best possible treatment options for these 
patients. 



Conclusions 



Renovascular hypertension is an uncommon 
cause of hypertension in the general population. 
However, it may cause considerable morbidity 
with respect to end-organ damage from uncon- 
trolled hypertension as well as progression of 
ischemic nephropathy. It appears as if duplex 
ultrasound is at least a reasonable screening tool 
in patients suspected of having hypertension 
that is renal in origin. However, there clearly 
needs to be correlation between the anatomical 
stenosis and the physiological effect. For this 
purpose, there is not a clearly optimal test, 
although captopril renography seems to offer 
the best results at this time. 

Once diagnosed and found to be functionally 
significant, there are several options for the 
treatment of RAS, including medical manage- 
ment, endovascular angioplasty with or without 
stent placement, or open surgical revasculariza- 
tion. From the data that are available, it is 
clear that there is a patient population that 
does benefit from revascularization, either 
endovascular or surgical. Surgical revascular- 
ization is more durable, and long-term success 
has been previously demonstrated. Angioplasty 
with or without stent placement is indeed less 
durable over the long-term; however, with close 
surveillance and secondary intervention, it may 
offer results comparable with open surgical 
repair. 

Although there have been a multitude of 
attempts to define which patients with sympto- 
matic RAS would benefit from renal revascular- 
ization, both surgical and endovascular, there is 
still no clear consensus at the present time. 
Recently, the American Heart Association 
released the guidelines for the reporting of renal 
artery revascularization in clinical trials. This 
document serves to clearly outline criteria and 
definitions for randomized, controlled clinical 
trials, which are needed to further dictate the 
optimal management of symptomatic RAS. At 
this time, it is clear that certain patients do 
benefit from restoration of blood flow to the 
kidney, but a multicenter, prospective, random- 
ized, controlled study will be better able to 
define which patients benefit from surgical 
repair compared to endovascular management 
and which patients, if any, should merely be 
treated medically. 



229 



RENOVASCULAR HYPERTENSION AND ISCHEMIC NEPHROPATHY 




References 



Barrett BJ, Carlisle EJ. (1993) Radiology 188:171-8. 

Bush RL, Najibi S, MacDonald MI, et al. (2001) J Vase Surg 

33:1041-9. 
Goldblatt H. (1934) J Exp Med 59:347. 
Hunt JC, Strong CG. (1973) Am J Cardiol 32:562-74. 
Martin LG, Rundback JH, Sacks D, et al. (2003) J Vase Interv 

Radiol 14:5297-310. 
Pak LK, Kerlan RK, Mully TW, Messina LM. (2002) J Vase 

Surg 35:808-10. 
Radermacher J, Chavan A, Bleck J, et al. (2001) N Engl J Med 

344:410-17. 



Rundback JH, Sacks D, Kent KC, et al. (2002) J Vase Intervent 
Radiol 13:959-74. 

Simon N, Franklin SS, Bleifer KH, Maxwell MH. (1972) 
JAMA 220:1209-18. 

Webster J, Marshall F, Abdalla M, et al. (1998) J Hum Hyper- 
tens 12:329-35. 

Weibull H, Bergqvist D, Bergentz SE, Jonsson K, Hulthen L, 
Manhem P. (1993) J Vase Surg 18:841-50; discussion 
850-2. 

Zierler RE, Bergelin RO, Isaacson JA, Strandness DE Jr. 
(1994) J Vase Surg 19:250-7; discussion 257-8. 



19 



Visceral Ischemic Syndromes 

George Geroulakos, Peter A. Robless, and 
William L. Smead 




The disorders of the visceral circulation are 
infrequent mainly because of the very extensive 
and efficient collateral system connecting the 
celiac, superior mesenteric, and inferior mesen- 
teric arteries. However, there is a lot of interest 
in the optimal management of these conditions, 
because of their catastrophic outcomes that are 
usually associated with a high morbidity and 
mortality. Over the last two decades there has 
been a greater awareness of these conditions, 
which was followed by the introduction of new 
diagnostic and therapeutic techniques such as 
duplex ultrasound, computed tomography (CT), 
magnetic resonance angiography (MRA), and 
angioplasty/stenting. 

Visceral ischemic syndromes can be classified 
as acute or chronic. Acute ischemic syndromes 
are the result of embolic or thrombotic occlu- 
sion of a visceral branch of the infradiaphrag- 
matic aorta. In addition, it could be the result of 
mesenteric venous thrombosis. Chronic visceral 
ischemia is usually caused by stenotic or occlu- 
sive atherosclerotic lesions involving two or 
more visceral vessels. 

Acute Mesenteric Ischemia 

Acute mesenteric ischemia (AMI) is a life- 
threatening condition seen in 1 per 1000 hospi- 
tal admissions with mortality rates ranging 
between 60% and 100% despite advances in 
operative technique and perioperative manage- 
ment (Bradbury et al., 1995). Early diagnosis 



and aggressive management are essential. 
However, the relative rarity of this condition, 
along with the nonspecific physical findings 
makes early diagnosis difficult and is often 
delayed. Furthermore, patients with AMI are 
often elderly, malnourished, and have signi- 
ficant comorbidity that increases the risk of 
major surgical intervention. 

The most common causes of AMI are supe- 
rior mesenteric artery embolization (50%) or 
thrombosis (25%), nonocclusive mesenteric 
ischemia (20%), and acute mesenteric venous 
thrombosis (5%). Rarer causes include vas- 
culitis, fibromuscular dysplasia, dissection, 
trauma, and mesenteric aneurysm rupture or 
thrombosis. 

Pathophysiology 

Two thirds of mesenteric blood flow supplies 
the gut mucosa. Splanchnic autoregulation fails 
when perfusion pressure falls below 40mmHg, 
and prolonged gut ischemia results in anaerobic 
metabolism. The extent of injury is related to 
the duration and the anatomical extent of 
mesenteric ischemia. At the cellular level, 
adenosine triphosphate (ATP) is depleted with 
a buildup of catabolic products and lactate. 
Mucosal and vascular permeability increases 
and tissue injury occurs. Hemorrhagic necrosis 
follows with mucosal sloughing, bowel wall 
edema, and intestinal hemorrhage (Table 19.1). 
The bowel wall becomes permeable to gut bac- 
teria once the mucosa is shed. Peritonitis results 



231 




232 



VASCULAR SURGERY 



Table 19.1. Pathophysiology of mesenteric ischemia 

Mucosa — villous sloughing 
Increased capillary permeability — Edema 
Submucosal hemorrhage 
Transmural necrosis 



from transudation of microflora across the 
intestinal wall. Septicemia develops as the 
organisms enter the portal circulation. Massive 
fluid shifts into the bowel wall and peritoneum 
follow, resulting in hemoconcentration, oliguria, 
and hypotension. Serum levels of lactate dehy- 
drogenase (LDH), serum glutamic oxaloacetic 
transaminase (SGOT), and creatine kinase (CK) 
become markedly elevated with the death of 
intestinal cells. 

Ischemia reperfusion injury due to free 
radical production through the xanthine 
oxidase pathway may result from subsequent 
reperfusion of the acutely ischemic gut. There is 
a high mortality rate from the ensuing multior- 
gan dysfunction syndrome. 

Acute Mesenteric Embolism 

The main presenting symptom is usually the 
sudden onset of severe central abdominal pain. 
The severity of the pain is usually out of pro- 
portion to the physical findings. This may be 
accompanied by vomiting or diarrhea. 

Mesenteric emboli can originate from the 
heart or the supradiaphragmatic aorta and most 
frequently occur in patients with cardiac arry- 
thmias, valvular disease, or following myocar- 
dial infarction. If the embolus disintegrates and 
travels distally, the resulting ischemia may be 
patchy, typically affecting the duodenum, prox- 
imal jejunum, and colon. The majority of emboli 
lodge in the superior mesenteric artery (SMA) 
distal to the origin of the middle colic artery, 
often sparing the proximal jejunum and ascend- 
ing/transverse colon. Owing to the lack of ade- 
quate collaterals, this may lead to reactive 
vasoconstriction, thereby reducing existing col- 
lateral blood flow and increasing the ischemic 
injury. 

Mesenteric Artery Thrombosis 

Thrombosis of the SMA or celiac axis occurs 
as a result of underlying mesenteric atheroscle- 



rotic stenosis progressing to occlusion. This 
usually occurs at the origin of the vessel. Other 
causes include systemic vasculitic and pro- 
thrombotic syndromes. Less common causes 
include aortic and visceral artery aneurysms 
or dissection. Some patients may describe pro- 
dromal symptoms compatible with chronic 
mesenteric ischemia. These patients often have 
coexisting multilevel atherosclerotic disease. 
Unlike acute mesenteric embolism, the onset of 
pain is often gradual, with nonspecific central 
abdominal pain. The extent of infarction usually 
involves the duodenum to the transverse colon 
and is typically more extensive than that seen 
with acute embolism. 

Nonocclusive Mesenteric Ischemia 

Nonocclusive mesenteric ischemia (NOMI) 
develops in patients with low-cardiac-output 
states, especially in the presence of digoxin or 
vasoconstrictors. Secondary mesenteric vaso- 
constriction results in segmental vasospasm of 
the secondary and tertiary branches of the SMA. 
It is not caused by underlying atherosclerosis or 
venous obstruction. Causes of low-flow states 
include cardiac failure, shock, and hypovolemia. 
The use of vasoconstricting agents that affect 
the splanchnic circulation such as digoxin, 
cathecholamines, angiotensin II, vasopressin, 
beta-blockers, and cocaine has been associated 
with NOMI. 

Diagnosis 

Acute mesenteric ischemia presents classically 
with acute onset of abdominal pain out of pro- 
portion to the physical findings. Central abdom- 
inal pain occurs as a result of mid-gut ischemia 
and spasm. Gastrointestinal emptying, with 
emesis and bloody diarrhea may occur. Labora- 
tory findings including leukocytosis, acidosis, 
hyperkalemia, raised hematocrit, LDH, SGOT, 
and CK occur later. However, no single labora- 
tory investigation or combination of tests has 
proved to be sensitive or specific enough to 
enable the early diagnosis of AMI. 

Early recognition of AMI is crucial, as bowel 
necrosis develops in many patients by the time 
of surgery and investigations should not pro- 
duce unnecessary delays in revascularization. 

Plain abdominal x-rays may show nonspecific 
findings of intestinal dilatation, gasless abdo- 



233 



VISCERAL ISCHEMIC SYNDROMES 




men, or other signs of ileus. Occasionally mural 
"thumb printing" is caused by submucosal 
edema or hemorrhage. Pneumoperitoneum or 
portal vein pneumatosis may be seen in 
advanced cases with transmural infarction. 
Ultrasonography in patients with AMI may 
reveal a thickening of the bowel wall, signs of 
ileus with distended bowel loops, intraperi- 
toneal fluid, or air in the portal vein. Ultra- 
sonography is also helpful in excluding other 
causes of an acute abdomen. 

Ultrasound and plain x-rays are not very 
specific in diagnosing AMI and other imaging 
modalities such as CT, MRA, and contrast 
angiography play a major role in the diagnosis. 
Computed tomography angiography is sen- 
sitive for the diagnosis of mesenteric occlusion 
or bowel ischemia. Computed tomography 
scanning also facilitates the identification of 
nonvascular causes of acute abdominal pain. 
Computed tomography angiography with three- 
dimensional reconstruction may facilitate iden- 
tification of vascular anatomy and pathology 
with good enough detail for diagnosis and oper- 
ative planning (Fock et al., 1994). 

Differentiation of the three forms of mesen- 
teric arterial occlusion can be done with aor- 
tography and it facilitates planning of 
treatment. Selective mesenteric angiography 
remains the most reliable and definitive diag- 
nostic tool for AMI. Endovascular interven- 
tions or catheter-directed vasodilator therapy 
can be started immediately after angiography 
(Park et al., 2002). 

Thrombotic occlusion of the SMA produces a 
sudden cut-off at the vessel origin or within 1 to 
2 cm of the SMA trunk. Extensive collaterals in 
the distal SMA indicate a chronic occlusion. 
Mesenteric arterial emboli produce a sharp, 
rounded filling defect with a typical meniscus 
sign on angiography. Vasospasm maybe present 
with mesenteric thrombosis or embolism. 
Angiographic criteria for NOMI include a 
diffuse narrowing of the SMA and its branches, 
alternating areas of narrowing and dilatation of 
the SMA branches (string of sausages sign), 
spasm of the peripheral vascular arcades, 
impaired filling of the intramural vessels, and a 
sluggish flow with reflux of contrast during 
selective SMA injection. Increased vessel diam- 
eter following papaverine infusion and the 
absence of atherosclerotic disease on angiogra- 
phy supports the diagnosis of AMI. 



Treatment 

Regardless of the etiology of the AMI, the goals 
of surgical treatment are to reestablish good 
pulsatile flow to the SMA, to restore adequate 
blood flow to the ischemic but viable gut, and to 
resect the necrotic bowel. Adequate volume 
resuscitation and correction of acid base and 
electrolyte imbalance must also be undertaken. 
Cardiac output has to be optimized, and any 
arrhythmias treated. Broad-spectrum antibi- 
otics are given if signs of peritonitis are present. 

The single factor in improving the results 
of surgical treatment of acute mesenteric 
insufficiency has been the addition of tran- 
scatheter intraarterial vasodilator infusion peri- 
operatively (Boley et al., 1981). 

The decision to undertake second-look 
laparotomy is made at the time of the initial 
laparotomy. It allows the reassessment of bowel 
viability and to decide if further bowel resection 
is required. Bowel viability can be assessed by 
physical examination, handheld Doppler scan 
examination, and intravenous injection of 
fluorescein (Ballard et al., 1993). 



Acute Mesenteric Artery Embolism 

Surgical revascularization in the presence of an 
embolism is performed with balloon embolec- 
tomy usually with p atch angioplasty of the SMA. 
Patients with chronic proximal occlusion or 
stenosis undergo revascularization with bypass 
grafting. Autogenous vein is the graft material 
of choice if resection of necrotic bowel is nec- 
essary. Resection of the infarcted bowel is per- 
formed following revascularization. 



Acute Mesenteric Artery Thrombosis 

The thrombotic process occurs in a severely ath- 
erosclerotic proximal SMA. Therefore, these 
patients require placement of a bypass graft to 
the SMA distal to the occlusive segment. Ante- 
grade or retrograde bypass may be performed 
depending on anatomical considerations or 
according to surgeon preference. 

Thrombolytic therapy is a potential consider- 
ation in patients with acute thrombosis and no 
clinical signs of peritonitis. Successful lysis 
returns the mesenteric circulation to its chronic, 
stable state. Subsequent operative revasculari- 




> 



234 



VASCULAR SURGERY 



zation or balloon angioplasty of the stenotic 
vessel can be undertaken electively. How- 
ever, this does not allow inspection for bowel 
viability following reperfusion and may delay 
operative revascularization if thrombolysis is 
unsuccessful. Thrombolysis in AMI therefore 
should be reserved for selected patients (Park 
et al, 2002). 



Nonocclusive Mesenteric Ischemia 

The treatment of NOMI is primarily nonsurgi- 
cal. A metabolic cause of the problem should be 
identified and corrected. The SMA is selectively 
catheterized, and vasodilating agents such as 
papaverine (or tolazoline hydrochloride) are 
administered. Resection of nonviable bowel 
may be required (Park et al., 2002). 



Chronic Visceral Ischemia 

Clinical Presentation 

The most common pattern of symptoms is 
chronic abdominal pain that is associated with 
involuntary weight loss. The pain is usually epi- 
gastric, dull, or colic. The patient experiences 
the pain 15 to 30 minutes after eating, and it 
lasts for 1 to 3 hours before disappearing. The 
pain becomes so severe that soon the patient 
develops fear of food and limits the oral intake. 
This results in a pronounced weight loss. 
Absence of weight loss may put in doubt the 
diagnosis of chronic visceral ischemia. Other 
gastrointestinal complaints may include diar- 
rhea, nausea or vomiting, and constipation. The 
most typical feature of the clinical presentation 
is that the symptoms are atypical. We and others 
have shown that the majority of the patients 
affected are women in their sixth decade of life. 
The reasons for this sex predilection remain 
undetermined. On examination, the patient 
looks emaciated, mimicking a patient with 
advanced malignant disease. These is often an 
epigasric bruit present. 

Untreated patients with symptoms of chronic 
visceral ischemia are at an increased risk of 
death from complications of acute visceral 
ischemia. In a large series of patients presenting 
with acute mesenteric ischemia over a period of 
10 years, 43% had prior chronic symptoms. 



Investigations 

Gastrointestinal contrast studies, endoscopy, 
and computed tomography are not essential to 
the diagnosis but are important in eliminating 
other sources of abdominal discomfort. 
Microulceration of the gastric mucosa and atyp- 
ical colonic mucosa ulceration are uncommon 
endoscopic findings. 

Duplex ultrasound examination of the celiac 
and the superior mesenteric artery origin can 
be successfully obtained in 80% to 95% of the 
cases, and in these it is a reliable screening test 
for chronic visceral ischemia. To obtain optimal 
visualization of the celiac and superior mesen- 
teric arteries, patients should be on a clear 
liquid diet on the day before the examination 
and should refrain from oral intake for 6 hours 
before the study. Lateral views of biplane aor- 
tography remains the primary diagnostic 
modality in demonstrating visceral occlusive 
lesions compatible with the diagnosis. In a 
typical patient there is involvement of two or all 
three visceral arteries. Aortography also reveals 
the meandering mesenteric artery (arch of 
Riolan or arch of Treves), a large collateral tor- 
tuous vessel of uniform caliber residing in the 
left upper quadrant of the abdomen. It connects 
the middle colic artery with the patent trunk of 
the inferior mesenteric artery. 

Treatment 

Angioplasty 

Mesenteric percutaneous transluminal angio- 
plasty (PTA) is a valuable treatment option in 
patients with chronic visceral ischemia who are 
considered high operative risk. The initial tech- 
nical success rate is good, with the majority of 
patients having symptomatic improvement and 
continuous relief of symptoms at short-term 
follow-up. However, long-term patency results 
are unknown. Allen et al. (1996) presented a 
series of 19 patients treated by PTA. The only 
technical failure resulted in mesenteric artery 
dissection, thrombosis, bowel infarction, and a 
fatal outcome. Complete symptomatic relief was 
attained in 15 patients. Recurrent symptoms 
developed in three patients (20%) at a mean 
interval of 28 months. Percutaneous translumi- 
nal angioplasty may also be used for the man- 
agement of recurrent symptoms following the 



235 



VISCERAL ISCHEMIC SYNDROMES 




development of a stenosis in the aortomesen- 
teric bypass. 

Surgery 

A variety of techniques have been developed 
to correct atherosclerotic obstruction of the 
major visceral branches of the thoracic aorta. 
Techniques of revascularization include tran- 
section of the mesenteric artery and reimplan- 
tation on the aorta, bypass grafting, and 
thromboendarterectomy There is no consensus 
regarding the best surgical approach for the 
treatment of chronic visceral ischemia. Bypass 
grafting is the most common type of visceral 
revascularization performed today. It provides 
immediate restoration of flow and may origi- 
nate from several different locations that 
include the suprarenal aorta (antegrade recon- 
struction) or the infrarenal aorta and the 
common iliac arteries (retrograde reconstruc- 
tion). The distal thoracic aorta is usually free of 
atherosclerosis and is an excellent origin of a 
short bypass placed in the direction of normal 
flow. This design eliminates the possibility of 
kinking and thrombosis by compression or 
traction from the overlying mesentery, which 
may be observed with retrograde grafts. The 
distal thoracic aorta may be approached from 
the abdomen or via a thoracoabdominal inci- 
sion. The retrograde bypass has the advantage 
that its origin is in a more familiar territory, 
but it is more commonly affected by atheroscle- 
rotic or aneurysmal disease. Park et al. (2002), 
in a large series of 98 reconstructions for 
chronic visceral ischemia, reported a 5-year 
symptom-free survival of 92%. The rate of 
recurrence was unaffected by the number of 
vessels revascularized or the orientation of the 
bypass. In contrast, others have reported that 
revascularization of as many visceral vessels 
as possible is important in reducing the risk 
of recurrent intestinal ischemic symptoms 
(Rheudasil et al., 1988). Thus the number of 
vessels that need to be revascularized remains a 
controversial issue. 

Thromboendarterectomy has been used by a 
small number of groups with excellent results. 
The aorta is approached via medial visceral 
rotation. For the majority of ostial stenoses, a 
"trapdoor" arteriotomy is made around the 
celiac and superior mesenteric arteries as they 
emerge from the aortic wall. A retrograde 



endarterectomy facilitates direct removal of the 
aortic plaques. Duplex scanning should be 
routinely used at the end of the procedure to 
document the technical result at the distal end 
point of the endarterectomy in the mesenteric/ 
celiac artery. A comparison of antegrade bypass 
(n =004 26) and transaortic endarterectomy 
(n = 48) showed the same incidence of periop- 
erative mortality and 5-year recurrence-free 
survival for both groups (Cunningham et al., 
1991). 



Mesenteric Venous 
Thrombosis 

Mesenteric venous thrombosis is a rare but 
potentially lethal form of intestinal ischemia 
that accounts for 5% to 15% of all cases of 
mesenteric vascular events. Improved survival 
depends on early recognition and appropriate 
treatment of this entity. 



Etiology 



Low-flow states in the mesenteric venous circu- 
lation that may be produced by liver cirrhosis, 
portal hypertension, and congestive heart 
failure could lead to mesenteric venous throm- 
bosis. Intraabdominal infections from organs 
that drain to the mesenteric venous system such 
as appendicitis, diverticulitis, pelvic abscess, 
and pancreatitis can be associated with mesen- 
teric venous thrombosis. Other conditions that 
may predispose to mesenteric venous thrombo- 
sis include hypercoagulable states associated 
with malignancy, oral contraceptives, and recent 
operations, particularly splenectomy followed 
by thrombocytosis. Thrombophilia is common 
in this group of patients. In a series of 31 
patients, 13 (42%) were diagnosed to have a 
hypercoagulable state. Twelve patients were 
found to have protein C, protein S, or antithrom- 
bin III deficiency. A single patient had activated 
protein C resistance. Six patients had prior 
thrombotic episodes, including deep venous 
thrombosis (« = 5) or arterial thrombosis (n = 
l).Four patients had a history of cancer, and five 
patients had previously undergone a splenec- 
tomy (mean platelet count 487,000) (Morasch 
et al.,2001). 




> 



236 



VASCULAR SURGERY 



Clinical Presentation 

The most common presenting symptom is 
abdominal pain. The pain is constant, colicky, 
and poorly localized. Often the complaints of 
abdominal pain are out of proportion to the 
clinical findings. Other symptoms include vom- 
iting, diarrhea, and constipation. In a series of 
patients with mesenteric venous thrombosis, 
upper and lower gastrointestinal bleeding 
occurred in 10% and 19% of the patients, 
respectively. Physical findings in patients who 
had gradual onset of abdominal symptoms may 
reveal low-grade pyrexia, abdominal distention, 
increased bowel sounds, and generalized 
abdominal tenderness. When the pain is local- 
ized, it may be located in either the upper or 
lower quadrants of the abdomen. 

Peritoneal signs such as guarding and 
rebound are present when infarction of the 
bowel has occurred. 

Investigations 

Plain abdominal films are abnormal but not 
specific in the majority of the patients. Edema- 
tous and dilated small bowel loops are the most 
common finding. 

Contrast-enhanced CT scanning is the most 
sensitive tool in detecting acute mesenteric 
venous thrombosis. The presence of a mesen- 
teric venous filling defect is diagnostic of this 
condition. Other CT findings include free fluid; 
an enlarged, dilated superior mesenteric vein; 
mesenteric edema or stranding; bowel wall 
thickening or edema; and dilated small bowel 
loops. Selective mesenteric angiography may 
show intense spasm of the arterial branches 
supplying the involved part of the bowel, pro- 
longed opacification of the thickened bowel 
wall, contrast extravasation into the lumen of 
the bowel, visualization of the venous thrombus, 
or nonvisualization of the venous phase. Cur- 
rently, angiography is not recommended as the 
initial investigation. 

Treatment 

Patients with mesenteric venous thrombosis 
and no signs of peritonitis are treated with fluid 
resuscitation, prophylactic antibiotic therapy, 



and intravenous heparin. Close clinical and 
hemodynamic monitoring is indicated, prefer- 
ably in an intensive care unit. If peritoneal signs 
are present, a laparotomy should be performed 
and the infarcted bowel should be resected. An 
ileostomy should be considered if there is exten- 
sion of the macroscopic involvement at the 
resection margins. The macroscopic features of 
the affected part of the bowel are striking, and 
consist of a dark red coloration and thickening 
of the bowel. It is often difficult to distinguish 
viable from nonviable bowel. In a recent series 
82% of patients who underwent bowel resection 
(about 100 cm length) did not have transmural 
necrosis. Bowel viability assessment by direct 
inspection may be improved by Doppler ultra- 
sound techniques or fluorescein examination 
under a Wood's lamp. Several case reports have 
been published indicating that thrombolysis 
may be a useful treatment option for patients 
with no signs of peritonitis. Antegrade transar- 
terial thrombolysis via the superior mesenteric 
artery has been effective in a small number of 
cases in lysing the thrombus and providing 
good symptomatic relief (Antoch, 2001). Post- 
operatively, patients should be anticoagulated to 
prevent recurrent episodes of thrombosis. The 
duration of anticoagulation is determined by 
the expected duration of the predisposing 
factors. 



References 



Allen RC, Martin GH, Rees CR, et al. (1996) J Vase Surg 

24:415-21; discussion 421-3. 
Antoch G, Taleb N, Hansen D, Stock W. (200 1 ) Vase Endovasc 

Surg 20:471-2. 
Ballard JL, Stone WM, Hallett JW, Pairolero PC, Cherry KJ. 

(1993) Am Surg 59:309-11. 
Boley SJ, Feinstein FR, Sammartano R, Brandt LJ, Sprayre- 

gen S. (1981) Surg Gynecol 153:561-9. 
Bradbury AW, Brittenden J, McBride K, Ruckley CV. (1995) 

Br J Surg 82:1446-59. 
Cunningham CG, Reilly LM, Rapp JH, Schneider PA, Stoney 

RJ. (1991) Ann Surg 214:276-87; discussion 287-8. 
Fock CM, Kullnig P, Ranner G, Beaufort-Spontin F, Schmidt 

F. (1994) Eur J Radiol 18:12-14. 
Morasch MD, Ebaugh JL, Chiou AC, Matsumura JS, Pearce 

WH, Yao JS. (2001) J Vase Surg 34:680-4. 
Park WM, Gloviczki P, Cherry KJ Jr, et al. (2002) J Vase Surg 

35:445-52. 
Rheudasil JM, Stewart MT, Schellack JV, Smith RB 3rd, Salam 

AA, Perdue GD. (1988) J Vase Surg 8:495-500. 



20 



Endovascular Approaches and Techniques 

Steven M. Thomas, Kong T. Tan, and Mark F. Fillinger 




This last chapter describes the equipment com- 
monly used during endovascular procedures; 
some of the commonly used endovascular tech- 
niques, beginning with arterial access tech- 
nique; and the endovascular interventions for 
arterial occlusive disease and aneurysmal 
disease. Embolotherapy is also discussed. 

Equipment for Access, 
Angiography, and 
Vascular Intervention 

Needles 

In general terms, there are two types of needles 
available for obtaining vascular access: one-part 
and two-part. The one-part needle has a sharp 
cutting bevel, whereas the two-part needle has 
a sharp inner stylet with a blunt outer needle. 
The two-part needle is preferred by many for 
vascular access at the femoral artery, because it 
reduces the risk of vascular damage at the access 
point, either due to movement of the needle tip 
or passage of the guidewire into the subintimal 
plane (see Access Technique, below). The most 
commonly used needle calibers are 18 and 19 
Stubs needle gauge (18 being the larger and 
equivalent to about 1.25 mm in outer diameter). 
For most purposes the 19-gauge needle suffices, 
though it does not accommodate an 0.038-inch- 
diameter guidewire if required. A newer variant 
of the one-part needle designed to reduce arte- 



rial injury is the 20- or 22-gauge needle typically 
used as part of a "micropuncture" set. The 
micropuncture technique uses a 0.014- or 0.018- 
inch guidewire and standard Seldinger tech- 
nique to introduce a short 4-French sheath, 
which is then used to place the typical 0.035- 
inch guidewire. 

Wires 

Guidewires are integral to successful catheteri- 
zation; they support the catheter and allow the 
catheter to be steered to its intended location. 
Guidewires come in many diameters and 
lengths, and degrees of stiffness, slipperiness, 
and steerability All wires have a softer, more 
flexible, curved, or "floppy" leading end to min- 
imize damage to the vessel as it is advanced, 
though the length of the flexible segment may 
vary. This is because a long flexible segment may 
be useful if a vessel is extremely tortuous, or a 
very short flexible segment may be needed to 
allow maximum support for an advancing 
catheter or sheath. 

Diameter 

Most standard guidewires are 0.035 inch in 
diameter, though some very stiff wires are 0.038 
inch in diameter. It is important to ensure that 
the wire used is compatible with the lumen 
diameter of the catheter to be placed over it. If 
small-caliber catheters are used, small-diameter 
wires such as 0.014 or 0.018 inch are required. 



237 




> 



238 



VASCULAR SURGERY 



However, many of these wires do not have the 
same stiffness as larger diameter wires, and 
hence may kink or result in poor tracking of the 
catheter. If a kink is produced, it is usually very 
difficult to pass a catheter over the wire, and a 
new wire is required. The hydrophilic Terumo 
(Terumo, Leuven, Belgium) guidewire has a 
nitinol core and is difficult to kink. 

Length 

The usual length of guidewire required is about 
150 cm, and this is adequate for most diagnostic 
imaging. However, in some circumstances 
longer wires are required, usually ranging from 
200 to 300 cm. This is particularly the case in a 
peripheral location, where the catheter has to be 
changed, or a guiding catheter, angioplasty 
balloon, or stent has to be introduced. When 
using monorail balloon catheters or stents for 
intervention, this is usually less of a problem 
compared to systems that are fully over the wire. 
This is because in a monorail system the wire 
passes through the wire guide for only about 30 
cm from the tip of the catheter or device, rather 
than through the whole length of the catheter or 
device as occurs in a fully over-the-wire system. 
Aortic stent-graft interventions also tend to 
require long (260 cm) guidewires due to the 
length of the delivery system for most aortic 
stent grafts. However, systems tend to be manu- 
facturer-specific, as some newer aortic stent- 
graft systems require only 180-cm guidewires. 

Stiffness 

The degree of guidewire stiffness varies consid- 
erably (Table 20.1). It is generally easier to track 
a catheter or device over a stiffer guidewire; 
however, a very stiff guidewire may be more 
difficult to manipulate into a vessel origin or 
through a diseased vessel. Exchanging wires is 
often necessary for different parts of a proce- 
dure, but a stiff wire should always be placed 
through a catheter that has been positioned 
using a more conventional wire to prevent 
damage to the vessel during advancement of the 
stiff wire. 

Slipperiness 

Some wires (e.g., Terumo Glidewire or Cook 
Nimble guidewire), have a hydrophilic coating 
that, once lubricated, becomes very slippery and 



Table 20.1. Variation in guidewire stiffness 

Floppy 

Bentson 

Movable core 

Standard J or straight 

Hydrophilic 
More supportive 

Heavy duty J or straight 

Stiff hydrophilic 

Wholey 
Stiff 

Amplatz super stiff 

Amplatz extra stiff 

TAD 

Flexfinder 
Very stiff 

Meier 

Lunderquist 



nearly frictionless. They are very useful for 
negotiating tortuous vessels and crossing 
stenoses and occlusions. They can be difficult to 
handle because when slippery, the wire may slip 
back through the operators hands during 
catheter exchanges, and if allowed to become 
dry, can stick to the operator's gloves and are 
then easily pulled out inadvertently. These prob- 
lems can be minimized by keeping hydrophilic 
wires moist, handling the wires with a damp 
gauze to allow slightly more friction than a 
glove, using a "torque device" when manipulat- 
ing the wire, and using caution when moving 
the wire in or out of the patient. 

Steerability 

A completely straight wire and a J wire have 
little or no inherent steerability. A straight wire, 
to which a curve has been introduced, can be 
directed using the curve, and this is enhanced if 
a directional catheter is also used. Many wires 
have tips that can be deformed into a curve or 
even into a J if required. Some hydrophilic wires 
have a preformed curve or angle, and if the core 
is made of nitinol (an elastic nickel-titanium 
alloy with "memory"), this cannot be changed. 
This gives more torque control and makes the 
wire very steerable. 

Catheters 

There are a plethora of catheters available, to be 
placed over the guidewire to facilitate arteriog- 



239 



ENDOVASCULAR APPROACHES AND TECHNIQUES 




raphy, selective entry to branch vessels, device 
delivery, and so on. The size of catheter is 
denoted using the French (F) system, which 
describes the outer circumference (external 
diameter of 3F = 1mm external diameter). 
Smaller caliber catheters have the advantage 
that the access puncture site can be kept small, 
although the turning ability or torque control is 
often compromised. For nonselective angio- 
grams this is not usually a problem, and 3F or 
4F catheters can be used, but if a selective 
catheterization is required, then larger caliber 
catheters may be required. 

It must be remembered that catheters are 
thrombogenic to some extent, and should 
always be regularly flushed with heparinized 
saline while in the patient. In the case of guide 
catheters, this is often best achieved using a bag 
of heparinized saline (using a pressure bag) 
connected via a side arm. 

Most catheters are reasonably radiopaque, 
though hydrophilic-coated "glide catheters" 
and smaller caliber catheters can be relatively 
radiolucent. Some catheters have radiopaque 
material added to them toward the tip, or a 
radiopaque band at the tip to enhance visibility. 

Catheter Types 

Most catheters can be divided into two main 
groups: nonselective and selective. 

Nonselective Catheters 

These are used to inject large volumes of con- 
trast at high rates into large vessels. They have 
multiple side-holes to allow high injection rates 
and reduce the risk of subintimal injection by 
the end-hole jet of contrast. The most com- 
monly used nonselective catheter is the pigtail 
catheter, the workhorse of diagnostic angiogra- 
phy. If the vessel is too small to accommodate 
the pigtail loop, a straight multi-side-hole 
catheter can usually be used instead. 

Selective Catheters 

These are shaped catheters that have directional 
properties that aid selective catheterization of 
side branch vessels. Some, such as the Multi- 
purpose, Cobra, Berenstein, and Sidewinder 
catheters, can be used for a variety of selective 
examinations, whereas others such as the inter- 



nal mammary or renal double-curve catheters 
are designed to aid catheterization of particular 
vessels. In practice, most angiographers prefer 
to have a range of catheters available and some- 
times many may be used to achieve a difficult 
selective arterial position. Probably the six 
most commonly used selective catheters are 
Berenstein (or similar Kumpe), Cobra, 
Sidewinder, Headhunter, Sos Omni, and Multi- 
purpose. Catheters such as the Sidewinder and 
Cobra come in a range of sizes designated 1 to 
3 dependent on the degree of catheter curve or 
loop. These are useful for varying the degree of 
angulation at the catheter tip, and the ease of 
manipulation as the diameter and tortuosity 
varies substantially in large vessels. When using 
a Sidewinder catheter in the aorta, if the loop is 
too large, the catheter tip will be held away from 
the aortic wall and will not engage branch 
vessels. This may be modified by introducing a 
guidewire into the apex of the catheter curve, 
but this often means that a smaller Sidewinder 
or a Sos Omni catheter should be used instead. 

Most of these catheters can be used as soon 
as the wire is removed from them, and will 
engage side branches if advanced or retracted. 
The Sidewinder and smaller Sos-Omni catheters 
need to be re-formed so that the reverse curve 
of the catheter can be used to engage side 
branch vessels. The Sos Omni, being of a smaller 
curve than the Sidewinder catheters, can usually 
be re-formed in the aorta. The Sidewinder 
catheter usually has to be re-formed using a 
variety of techniques. The simplest and safest is 
to place a guidewire over the iliac bifurcation 
using a pigtail catheter and re-form the 
sidewinder in the lower aorta. If this is not pos- 
sible, it can usually be easily reformed in the 
aortic arch by twisting and advancing the 
unformed catheter. Once formed, the catheter is 
pulled back to engage the origins of arterial side 
branches. 

When choosing a catheter, it is important first 
to select the appropriate gantry angle to view 
the origin of the vessel and its "true" degree of 
angulation relative to the parent vessel. The 
angle of origin of the vessel being catheterized 
often dictates which catheter is most likely to 
successfully engage the arterial origin, so, for 
example, vessels that are angled back toward the 
site of access are often best approached with a 
Sidewinder or similar catheter. There are also 
catheters that have hydrophilic coatings, and 




> 



240 



VASCULAR SURGERY 



these are often very useful for tracking a 
catheter to peripheral locations, particularly for 
embolization. 

Microcatheters 

These are coaxial catheters that are only 2F to 
3F in diameter that will pass through the 0.035- 
or 0.038-inch lumen of the selective catheters 
mentioned above. They were originally devel- 
oped for cerebral catheterization and inter- 
vention, but can be useful for superselec- 
tive catheterization of visceral or peripheral 
vascular beds, particularly for embolization 
procedures. 

Sheaths 

Sheaths are used to establish a secure pathway 
from the skin to the arterial lumen. This allows 
catheters and devices, such as balloons and 
stents, to be exchanged in an atraumatic 
manner. The sheath is a plastic tube with a 
hemostatic valve and an attached side arm to 
allow flushing. The size designation of the 
sheath refers to the size of catheter that will pass 
through it (i.e., the internal diameter). Hence, a 
7F sheath allows the passage of a 7F catheter or 
device. As a result the external diameter of any 
sheath is approximately IF larger than its desig- 
nated size for smaller sheath sizes, and 2F to 3F 
larger when the sheath size is in the 12F to 25F 
range. The thickness of the sheath is also some- 
what dependent on its length and intended 
application, generally thicker for longer sheaths 
intended to pass stents or stent grafts through 
tortuous arteries. If the puncture is at a steep 
angle or in obese patients, the smaller sheaths 
may kink, particularly with an antegrade punc- 
ture. There are reinforced sheaths that are resist- 
ant to kinking, but they have an external 
diameter IF to 2F greater than standard sheaths. 
Most sheaths are short, but longer sheaths 
and guiding catheters can be useful during 
interventional procedures to provide stability to 
aid selective catheterization and delivery of bal- 
loons or stents. Guiding catheters are larger 
than standard catheters, often 6F to 7F, because 
they are designed to work with a standard 
catheter within. Because they are catheters, it is 
important to remember that their size is desig- 
nated by the external diameter, unlike a sheath, 
despite similarities in appearance. Guiding 



catheters come in a variety of lengths and 
shapes to aid introduction into the vessel 
ostium, allowing conventional catheters to be 
introduced through them for more distal 
catheterization. Guiding catheters can perform 
a similar role to sheaths, but they do not usually 
have an integral hemostatic valve, and their 
internal lumen is smaller. Thus, in some cases a 
guiding catheter may be placed within a short 
sheath with a hemostatic valve, or used in com- 
bination with a Y adapter and a Tuohy-Borst 
valve for hemostasis. 

Commonly Used Medications 

When performing vascular interventions, a 
number of medications are frequently required. 
Examples in a number of broad groups are pro- 
vided, but this is not a comprehensive descrip- 
tion. Practioners should be familiar with local 
guidelines for medication use, and if in doubt 
should consult standard sources to check 
dosages and interactions. 

Local Anesthetics 

Local anesthetic is used for nearly all diagnos- 
tic and interventional procedures. The most 
commonly used agent is lignocaine hydrochlo- 
ride (lidocaine in the United States). It has a low 
incidence of side effects when used as a local 
anesthetic, but a total dose of 200 mg (20 mL of 
1% solution) should not be exceeded, as over- 
dosage may lead to central nervous and cardio- 
vascular system toxicity. Alternative agents 
are prilocaine hydrochloride or bupivacaine 
hydro chloride, which are longer acting and may 
be useful in certain circumstances. Depending 
on the agent, some adjustments may be advised 
for renal impairment, primarily related to 
metabolites that are renally excreted, or hepatic 
impairment. For any of these agents, direct 
intraarterial or intravenous injection should be 
avoided, due to risk of seizure and other poten- 
tial immediate systemic effects. 

Heparin 

Heparin is added to saline flushes and used rou- 
tinely in higher doses during interventional 
procedures. In both cases it helps prevent peri- 
catheter or vessel thrombosis during the pro- 



241 



ENDOVASCULAR APPROACHES AND TECHNIQUES 




cedure. For flushes, it is used in a dose of 
about 5000 IU per liter of normal saline. For 
interventional procedures, doses of between 
3000 and 5000 IU are given depending on the 
size of the patient and the proposed length of 
the procedure. For aortic stent-graft procedures, 
the sheaths may be totally occlusive, and sys- 
temic heparinization is commonly used. Sys- 
temic heparinization is also common in carotid 
stent procedures, due to the risk of emboli and 
to prevent thrombosis related to distal protec- 
tion devices. As the half-life of heparin is 45 to 
60 minutes, further doses should be considered 
if a procedure is prolonged. During longer 
cases or for systemic heparinization, dosing 
may be based on the activated clotting time 
(ACT), with target levels in the 250 to 300 s range 
depending on the procedure, the degree of vas- 
cular occlusion by devices, and the throm- 
boembolic risk. 

Vasodilators 

Vasodilators are used for prophylaxis or treat- 
ment of vascular spasm, and during intraarter- 
ial pressure measurements, to augment distal 
flow and give a better assessment of the 
significance of a pressure drop across a lesion. 
Commonly used intraarterial agents are glyc- 
eryl trinitrate (GTN, 100 jj,g) (nitroglycerin in 
the U.S., 50 to 100|J,g) and papaverine (30 mg). 
Of course, vasodilators may produce significant 
hypotension, so attention should be paid to 
patient hydration status and coexisting cardiac 
disease. Vasodilators should not be used in 
patients with significant aortic stenosis, as 
they may produce severe hypotension in this 
circumstance. Nifedipine (10 mg) can be given 
orally prior to a procedure if significant spasm 
is likely, such as embolization of the testicular 
vein. 

Atropine/Glycopyrronium Bromide 

These agents are given routinely during carotid 
stenting to reduce the risk of bradycardia and 
hypotension during balloon inflation in the 
carotid bulb. Glycopyrronium bromide (600 (J,g) 
is preferred in patients with a history of cardiac 
disease because it has a better side effect profile 
(U.S. equivalent is glycopyrrolate, adult dosage 
100 jj,g IV repeated every 2 to 3 minutes as nec- 
essary with a reasonable upper limit of 600 (J,g). 



Sedation/Analgesia 

Sedation is rarely required for vascular proce- 
dures, short of aortic stent grafts. Patients with 
limb ischemia should have their pain adequately 
controlled prior to investigation, as patients in 
severe pain can rarely lie still for prolonged 
periods. If sedation is required, small doses of 
short-acting benzodiazepines such as midazo- 
lam (2 to 10 mg IV) are preferred, with the aim 
being to relax, not anesthetize, an agitated 
patient. If anesthesia is produced, an anesthetist 
must be in control of it, but if oversedation does 
occur, the effects of the benzodiazepine can be 
reversed (temporarily) with flumazenil (200 ng 
over 15 s and then 100 |J,g every 60 s up to a total 
dose of lmg). If analgesia does become neces- 
sary, opiates such as morphine or pethidine (25 
to 100 mg) can be given IV, or an appropriate 
oral opiate can be given, though the onset of 
action may be considerably delayed. In all cases 
with analgesia and sedation, IV administration 
is preferred, as IM absorption is unpredictable 
and cumulative effects can result in sudden 
overdosage. 

Antibiotics 

When and which antibiotics are used varies 
according to local policy. Common indications 
are when prosthetic graft material is likely to be 
punctured during a procedure, when prosthetic 
material is to be placed (especially aortic stent- 
grafts), or for some embolization procedures. 

Antiplatelet Agents 

All patients with peripheral vascular disease 
should be advised to take antiplatelet agents, 
predominantly for their cardioprotective effect. 
Dosages of aspirin likely need be no higher than 
75 mg daily, although the precise optimal dosage 
has not been definitively determined. If the 
patient is intolerant of aspirin then dipyri- 
damole (25 to 50 mg) or clopidogrel (75 mg) are 
alternative agents. In our practice, patients are 
given clopidogrel 75 mg at least 3 days prior to 
and for 30 days following carotid stent inser- 
tion. If the patient has not taken a dose prior 
to the day of the procedure, a loading dose of 
300 mg is given on the day of procedure. In the 
U.S., most centers use both aspirin and clopido- 
grel for carotid stents. 




> 



242 



VASCULAR SURGERY 



Drugs for Treating 
Contrast Reactions 

It is imp erative to b e aware of the risk of a severe 
contrast reaction and its treatment. The drugs 
to do this should be readily available on the 
resuscitation trolley in the angiography suite. 
All centers should have a protocol for treatment 
of severe contrast reactions. This should be 
prominently displayed and usually involves 
the use of some or all of the following: chlor- 
pheniramine (10 to 20 mg IV); adrenaline or 
epinephrine (0.5 mL of 1:1000 solution subcu- 
taneous or 0.1 mL of 1:10,000 solution IV), 
usually given if there is severe anaphylactic 
shock; hydrocortisone (200 mg IV), though its 
effects are not immediate; cimetidine (300 mg 
IV), may be useful as a second-line drug follow- 
ing adrenaline; atropine (500 (J,g to 1 mg IV) can 
be used for severe bradyarrhythmia. 



Devices for Vascular 
Intervention 

Various devices may be required to perform 
vascular interventional procedures, such as 
balloon catheters and stents for treating arterial 
occlusive disease, or coils for embolization. The 
specifics of these devices are described during 
discussion on their use elsewhere in this 
chapter. 



Closure Devices 

Arterial puncture site closure devices aim to 
eliminate or reduce the need for manual com- 
pression of the puncture site, reduce the time 
to patient mobilization, and reduce the risk 
of significant bleeding, especially when using 
larger sheath sizes. For most centers it is the 
increased throughput of patients and the ability 
to perform intervention as a day case that 
justifies their use. 

There are a number of devices available, 
divided into two main groups: "stitchers" and 
"pluggers." The Closer (Perclose, CA) device is 
the original "stitcher." It percutaneously places a 
suture through the artery at the puncture site, 



and the patient can be fully mobilized immedi- 
ately. It can be used for closure of up to a 10F 
sheath size. Another suture-based device, 
Prostar XL (Perclose), has been used success- 
fully for closure of 24F femoral artery access. 
One of the authors has personally used these 
devices with good success for totally percuta- 
neous endovascular aortic aneurysm repair, 
both abdominal and thoracic. The primary 
issues are vessel size and calcification, which can 
affect the ability of the device to capture the 
sutures properly. Obesity and prior surgery or 
dense scar can also affect these devices. The 
vessel can be immediately reaccessed if neces- 
sary and other than the suture, there is no other 
residual material to cause problems if surgery is 
required at the access site. There are a number 
of plugging devices, such as the Angioseal (St. 
Jude Medical, St. Paul, MN), the Duett (Vascular 
Solutions Inc., Minneapolis, MN), and the 
Vasoseal (Datascope Corp., Montvale, NJ). 
The authors are also quite familiar with the 
angioseal. This is quick and simple to use and 
comes as a 6F or 8F device. An absorbable shoe 
remains inside the artery and a collagen plug is 
deployed over the puncture site outside the 
artery. The device now uses a knot mechanism 
to hold the collagen plug in position, and the 
patient can mobilize in 2 to 4 hours. The 
primary issues with this device are vessel size 
and the size of the arterial access device used for 
the procedure. 



Access Technique 



Before any diagnostic or interventional proce- 
dure can begin, vascular access has to be estab- 
lished. The basic technique remains the same as 
that described by Seldinger: 

• Vessel puncture 

• Guidewire introduction 

• Placement of the catheter over the 
guidewire 

Vessel Puncture 

For the purposes of this chapter, only arterial 
access is discussed; venous access is discussed 
in Chapter 13. In general, which vessel to punc- 
ture is dictated by the site of disease to be inves- 



243 



ENDOVASCULAR APPROACHES AND TECHNIQUES 




Table 20.2. Arterial access sites 




Access site 


Main indications 


Ipsilateral common femoral artery (CFA) 


All accessible stenotic/occlusive lesion 


Contralateral CFA 


Occluded ipsilateral CFA 

Lesion to be treated close to ipsilateral groin 

Scarred groin 


Low superficial femoral artery (SFA) 


Percutaneous transluminal angioplasty (PTA) of ipsilateral CFA (rare), SFA 
origin or proximal graft anastomosis but contralateral femoral puncture 
not possible 


Profunda femoris (PF) artery 


PTA of PF origin 

PTA of CFA (rare) if SFA occluded and contralateral approach not possible 


Direct graft puncture 


Arteriography in patients with graft crossing both groins 

Intervention at graft anastomoses 

Occluded graft, to allow accelerated thrombolysis 


Popliteal artery 


PTA of occlusions involving the origin of the superficial femoral artery 
Failure of guidewire passage antegrade, often due to steep, large collaterals 
at the proximal site of an occlusion 


Brachial artery 


Bilateral aortoiliac occlusions 

PTA of a supraaortic branch occlusion or stenosis 

Severe aortoiliac disease and vascular tortuosity 


Axillary artery (brachial approach preferred) 


As above 


Radial artery 


Arteriography as above; limited intervention possible 



tigated or treated, and the strength of the arte- 
rial pulses at various sites. The commonly used 
arterial puncture sites are shown in Table 20.2. 
For most purposes, the common femoral 
artery is used, as it is a relatively easy artery to 
puncture, the artery is readily compressed over 
the femoral head, and most vascular beds are 
easily within reach. The vessel is punctured at 
the point of maximal pulsation, not by means 
of external anatomical landmarks. At this point 
the vessel is over the femoral head. The site is 
first cleaned and draped, and local anesthetic 
inserted. A two-part needle is preferred for most 
access as this avoids the risk of damaging the 
vessel if the tip of the needle has to be reposi- 
tioned. There is also a reduced risk of the wire 
passing into a subintimal position. This can 
occur because the long bevel of a one-part 
needle may imply the needle tip is in the center 
of the lumen of the vessel, when it is only par- 
tially through the wall of the artery. The needle 
is guided at about 45 degrees to the skin surface 
toward the pulse, and the vessel is speared. The 
central stylet is then removed and the needle 



gently withdrawn until the tip is felt to flick into 
the lumen of the artery, and this should be 
accompanied by the free pulsatile backflow of 
arterial blood. 

Bony landmarks for the femoral artery on 
fluoroscopy can also be used, namely the medial 
aspect of the femoral head, but direct palpation 
is preferable. If there are difficulties gaining 
access or if the pulse is weak or absent, then gen- 
erally the best approach is to use ultrasound to 
directly visualize the arterial target. This is par- 
ticularly useful for antegrade femoral artery 
punctures, as it often avoids the need to use 
fluoroscopy to guide a wire into the superficial 
femoral artery (SFA) for distal intervention, and 
hence reduces the risk of irradiating the hands 
of the operator. In cases of difficult access, a 
micropuncture set is commonly used to reduce 
the risk of arterial injury, and in most of these 
sets the guidewire is specifically designed with 
an echogenic tip to aid visualization of the small 
0.018-inch wire. Once arterial access is obtained 
with a short 4F introducer, an exchange is made 
for a larger guidewire and 5F sheath. 




> 



244 



VASCULAR SURGERY 



Guidewire Introduction 

Once the needle is in position, a guidewire is 
introduced through it. For most purposes a 
standard 3-mm J wire is used to secure access 
and to allow initial introduction of catheters 
and sheaths. The wire should pass smoothly and 
easily into the artery. If any resistance is felt, 
fluoroscopy should be used to ensure the wire is 
following an appropriate course and is running 
freely. If the wire does not pass beyond a certain 
point, then a limited angiogram may be 
required to identify the problem, but this 
requires that at least some wire is within the 
arterial lumen to allow the introduction of a 
sheath or dilator. If the wire does not pass out 
of the needle, then there is probably an arterial 
plaque at the puncture site. The needle tip can 
often be repositioned without losing an intraar- 
terial position, but sometimes re-puncture is 
necessary, perhaps using ultrasound to guide 
the optimal site for puncture. 

Catheter Placement 

Once the wire has been advanced into a suitable 
position, a catheter or sheath can be introduced 
over it. The wire is held under slight tension as 
the catheter is introduced, as this helps to avoid 
the wire kinking in the soft tissues and makes 
tracking the catheter easier. Once the catheter 
has been advanced into position, the guidewire 
can be removed, the catheter flushed, and the 
intraluminal position verified with a test injec- 
tion of contrast. 

If the site of insertion has dense scar from a 
prior procedure, then insertion of even a small 
sheath or dilator may be difficult. If this is the 
case, then it is often prudent to use a stiffer 
guidewire initially or to change to one through 
a 4F dilator and then place a sheath for use 
during the rest of the procedure. Current 
micropuncture sets also have stiff versions for 
these cases. Although not needed frequently, 
these stiffer versions can be quite helpful. 

Endovascular Techniques for 
Arterial Occlusive Disease 

The mainstay of vascular intervention for arte- 
rial occlusive disease remains balloon angio- 
plasty, though arterial stenting also has an 



important role to play. Some techniques, such as 
laser treatments, have come and gone. Atherec- 
tomy has a limited role for occlusive disease, 
predominantly for graft or stent re stenosis, and 
stent grafts are being evaluated for occlusive 
disease to see if the expense can be justified over 
simple angioplasty or stenting. 

Angioplasty 

The technique of balloon angioplasty is used 
extensively to treat arterial stenoses and occlu- 
sions, and the principles and equipment are 
largely the same regardless of where they are 
applied. The basic principle is to place a 
guidewire through an arterial narrowing and 
over this to position an appropriately sized 
angioplasty balloon. The balloon is then inflated 
to high pressure, disrupting the stenotic arterial 
plaque. The luminal area, therefore, is increased 
and blood flow improved. 

As with all vascular intervention, planning is 
important and consideration should be given to 
the following: 

Access site: Access should usually be as near 
to the site to be treated as possible. This 
optimizes tactile control of wires and 
catheters, minimizes a tortuous approach 
to a lesion, and may help avoid an awkward 
approach to a vessel origin. For distal lower 
limb intervention, this usually means an 
antegrade femoral artery puncture. 

Crossing the lesion: The guidewire has to 
lie in an intraluminal position above and 
below the site to be treated (even in cases 
of subintimal angioplasty). For simple 
stenoses, the lesion is crossed intralumi- 
nally, often using a directional catheter, 
such as a Cobra and a curved wire, with or 
without a hydrophilic coating. Particularly 
when treating occlusions, the wire may 
pass (intentionally or otherwise) into the 
subintimal space within the diseased 
section. The wire usually buckles on itself 
when this happens, and if a hydrophilic 
wire is then used, a loop is formed with it. 
The wire must then reenter in an intralu- 
minal position in the more normal distal 
vessel before balloon dilatation is per- 
formed. This often occurs by advancing the 
wire loop in the subintimal space, with 
reentry occurring where a collateral arises 



245 



ENDOVASCULAR APPROACHES AND TECHNIQUES 




or at a vessel bifurcation — the basis of 
subintimal angioplasty. A Berenstein or 
other directional catheter can also be used 
to direct the wire back into the true lumen, 
and as with other steps in subintimal 
angioplasty, this should be performed with 
caution to avoid vessel perforation. New 
intravascular ultrasound catheters with 
wire guides and over-the-wire "spreading" 
devices have recently been produced to 
help cross-total occlusions, but are typi- 
cally needed only in difficult cases. 
Dilatation of the lesion: Angioplasty balloons 
fall into two main groups — compliant 
and noncompliant. Noncompliant balloons 
inflate to only one size, regardless of pres- 
sure, up to their burst rating. Compliant 
balloons vary in size, dependent on the 
pressure/volume of fluid within them. Most 
interventionists prefer noncompliant bal- 
loons because they often can be inflated 
to higher pressure and the risk of over- 
sizing of the balloon for the vessel is 
smaller. The size of balloon chosen is 
dependent on the size of the adjacent 
normal vessel, though most operators use 
an approximate guide based on experience 
and scale the size up or down for individ- 
ual patients (e.g., petite elderly women 
have small, fragile vessels). Inflation of the 
balloon can be performed by hand or using 
an inflation device. The latter has the 
advantage of a pressure gauge, allowing 
more control of the pressure and less like- 
lihood of exceeding the maximum balloon 
inflation pressure. Inflation is performed 
with a contrast/saline mix and the balloon 
should fully inflate with no "waist." Some 
operators inflate for a short period, others 
for 2 to 3 minutes, and some initially at 
high pressure, reducing to a prolonged low- 
pressure inflation. The basis for this is to 
reduce the risk of distal embolization and 
significant dissection. However, the evi- 
dence that these strategies achieve these 
aims is lacking. 

Following angioplasty a completion angio- 
gram should be performed to assess the appear- 
ance of the treated segment and the "runoff." 

Of primary importance is exclusion of vessel 
rupture with contrast extravasation, particu- 
larly in the iliac arteries. If there is arterial 



rupture, reinsertion of the balloon and inflation 
under low pressure proximal to the site of 
rupture should stop or minimize further bleed- 
ing. This may suffice for a small hole. It is 
prudent to inform a vascular surgeon of the 
problem. An angiogram should be performed 
after a trial of low-pressure balloon inflation. 
If extravasation continues, then the balloon 
should be immediately reinflated, and, if avail- 
able, an appropriately sized stent graft inserted. 
If this is not possible, then surgical repair of the 
arterial tear is required. 

If there is no injury and the treated segment 
remains significantly stenosed, then it may be 
that the balloon was undersized, in which case 
increasing the balloon size is often all that is 
required. In the iliac arteries, a pullback pres- 
sure can be performed through the lesion to 
assess if there is a significant pressure gradient 
at the site treated. This is usually taken to be 
a peak systolic pressure drop of lOmmHg 
across the lesion following administration of a 
distal vasodilator, such as GTN or papaverine. 
At other sites, for example, in the SFA, pres- 
sures are often not useful, as the catheter has to 
cross the treated segment for a pressure read- 
ing to be obtained distally, and this may in 
itself produce a pressure drop if the lumen is 
small. A normal distal pressure, however, is reas- 
suring that the stenosis has been treated ade- 
quately. If there is still a significant pressure 
drop in the iliac arteries after apparently ade- 
quate balloon inflation, then secondary stenting 
is warranted. If elastic recoil seems to be occur- 
ring on balloon deflation, again, stenting should 
be considered, depending on the site of the 
lesion. 

Often following angioplasty, a local dissection 
is visualized; however, most of these will 
remodel and do not require further treatment. 
If there is evidence of flow limitation, then there 
is a risk the segment will acutely occlude, and 
further investigation/treatment is warranted. 
Again a pressure measurement can be per- 
formed. For further treatment a long low-pres- 
sure inflation may tack back the dissection, 
but if this is unsuccessful, stenting may be 
required. 

Imaging the runoff vessels ensures that there 
has been no distal embolization. If this has 
occurred and the embolus is in an important 
vessel or producing poor flow/limb ischemia, 
then thrombo-aspiration should be performed 




> 



246 



VASCULAR SURGERY 



using an appropriate-size catheter for the 
occluded vessel and a sheath with a removable 
hemostatic valve. 

Stenting 

Intravascular stenting places a metallic scaffold 
inside the vessel to hold open the lumen. Stents 
were developed as an adjunct to balloon angio- 
plasty in recognition that angioplasty alone was 
occasionally unsuccessful. In this circumstance, 
the role of stenting is secondary to the primary 
treatment of angioplasty and is used to salvage 
a failed angioplasty. However, there are occa- 
sions when stenting is the initial treatment, so- 
called primary stenting. Examples include the 
treatment of ostial lesions, such as ostial renal 
artery stenosis (Fig. 20.1); most iliac occlusions; 
and carotid stenosis. The basis for stenting is to 
reduce problems caused by elastic recoil or 
distal embolization. Stents are generally avoided 
below the inguinal ligament, as there is no evi- 
dence they are superior to balloon angioplasty 
alone in this location. In these lower flow 
vessels, stents may result in early thrombosis 
and thus are reserved for bailout situations after 
angioplasty. 

There are a huge number of types of stents 
available for intravascular use, and choice of 
stent is usually at the discretion of the practi- 
tioner. The common types of stent are described 



here, with some examples given of those with 
which the authors are most familiar. 



Types of Stent 

Stents fall into two main groups: balloon- 
mounted or self-expanding. Balloon-mounted 
stents are usually made of stainless steel and 
are positioned on an angioplasty balloon for 
expansion and deployment. There is usually 
little shortening with expansion, and balloon- 
mounted stents are particularly useful where 
positioning of the stent is critical, enhanced 
by their radiopacity Larger balloon-mounted 
stents, for example, unmounted Palmaz stents, 
shorten considerably when expanded on a 
large-diameter balloon, making their place- 
ment more difficult. As these stents are quite 
inflexible, they may be difficult to position 
through tortuous vessels. Their rigidity, how- 
ever, gives them good radial strength, but if 
deformed once placed, for example, in superficial 
locations, or near joints, they will remain de- 
formed and may themselves cause vascular 
obstruction. Most of these stents now come pre- 
mounted on an angioplasty balloon ready for 
deployment, for example, Bridge stent (Medtonic 
AVE, Santa Rosa, CA), Genesis, or premounted 
Palmaz (Cordis, Miami Lakes, FL). Others have 
to be manually crimped to an appropriate 
balloon prior to use, for example, SAXX (C.R. 




Figure 20.1. Renal artery stenting. Gadolinium-enhanced magnetic resonance imaging (A) showing a tight stenosis in the solitary 
kidney, confirmed by catheter angiography (B) and successfully stented with a balloon-mounted stent (C). Note the left renal artery 
is occluded (arrow). 



247 



ENDOVASCULAR APPROACHES AND TECHNIQUES 




Bard, Murry Hill, NJ, USA) and the unmounted 
Palmaz (Cordis). The premounted stents are less 
prone to dislodgment from the balloon. This can 
occur as the stent is passed through the sheath 
and into its position for use, or during balloon 
inflation if a "dumbbell" is not formed around 
the stent as the balloon is initially inflated, in 
which case the stent may be squeezed off the 
deploying balloon. The latter problem is caused 
by movement of the stent from the center of the 
balloon or poor initial positioning so that one 
end of the balloon inflates first. Care is therefore 
required when using these stents, and if the stent 
has moved on the angioplasty balloon prior to 
deployment, it should not be used. 

Self-expanding stents all have an inherent 
ability to expand to a predetermined length 
and/or diameter. They are compressed onto the 
delivery catheter and are typically constrained 
by a covering sheath. As this sheath is with- 
drawn, the stent expands and is deployed. Being 
self-expanding, the stent continues to exert an 
expansile force on the vessel wall once deployed, 
and will show some flexibility and recovery to 
their expanded shape if compressed or kinked. 
Stents such as the Wallstent (Boston Scientific, 
Boston, MA) are made of surgical grade stain- 
less steel and rely on their design to be self- 
expanding. Newer stents tend to be made from 
nitinol, for example, the Memotherm Luminex 
(C.R. Bard), or the SMART stent (Cordis). 
Nitinol is a highly elastic nickel-titanium alloy 
that displays the property of thermal memory, 
returning to its preformed shape when released 
in the body. The "memory" properties of nitinol 
can be made to be more or less sensitive to tem- 
perature, which can be an advantage in certain 
situations. 

The Wallstent shortens considerably as it 
expands, with its final length governed by the 
diameter achieved in the vessel. This can make 
accurate placement difficult. It is very flexible, 
but has a relatively low expansile force once in 
place. Nitinol stents show considerably less 
shortening during expansion, and though 
nitinol is poorly radiopaque, many stents now 
have gold or platinum markers on their proxi- 
mal and distal struts to aid accurate placement. 

All stents come in a range of diameters and 
lengths to suit their use in different-sized vessels 
and different lesion lengths. Placement of a stent 
usually requires a larger sheath to allow entry of 
the stent into the vascular system, though many 



self-expanding and balloon-mounted stents can 
now be introduced through 5F to 6F sheaths. 

The basic principles of stenting are analogous 
to those of angioplasty, in the approach to the 
lesion and use of guidewires and catheters to 
cross the lesion. If there are difficulties tracking 
the stent to the lesion, a stiffer guidewire maybe 
required than for balloon angioplasty alone. The 
size and length of stent are selected for the size 
of vessel and the length of the lesion. For self- 
expanding stents the diameter of the stent is 
usually oversized by 1 to 2 mm, compared to the 
vessel diameter. Use of fluoroscopic road- 
mapping techniques help ensure that the stent 
is appropriately positioned, both prior to and 
during deployment, but this may require the use 
of a guiding catheter or catheter positioned 
from another access site to perform angiogra- 
phy. Many self-expanding stents, as they deploy, 
move away from the operator. Gentle back trac- 
tion on the delivery system and continuous 
screening during deployment should be used to 
maintain the position of the stent markers. Fol- 
lowing deployment of self-expanding stents, 
balloon dilatation can be used to ensure ade- 
quate expansion and apposition to the vessel 
wall. In some circumstances, for example, small 
external iliac arteries, ballooning to the desired 
vessel diameter may not be possible because the 
patient may experience pain, and there is a risk 
of arterial rupture. In these circumstances it is 
prudent to use a self-expanding stent, and in 
most cases the stent will expand adequately over 
the next few days with no adverse outcome. As 
with angioplasty, check angiography with 
runoff views should be performed. 

Atherectomy 

As with many new interventions, there was 
initially much enthusiasm for atherectomy. 
However, the lack of evidence that the technique 
was superior to simple balloon treatment for 
atheromatous disease has meant there is now 
limited use for this technique. Pending data 
from newer devices and techniques, the role of 
atherectomy is mainly limited to removal of 
neointimal hyperplasia from graft anastomoses 
or from within stents. Such lesions tend to be 
elastic and resist balloon angioplasty alone. 
Stenting is often counterproductive as it may 
exacerbate the hyperplastic process (at least 
pending data using new drug-eluting stents in 




> 



248 



VASCULAR SURGERY 



this role). However, the obstructing material can 
often be removed percutaneously using atherec- 
tomy devices. 

Stent Grafting 

Stent grafts are predominately used in the 
domain of aneurysmal disease. They have been 
advocated as an alternative to angioplasty 
and/or stenting for occlusive disease, often as a 
form of endovascular bypass procedure. As yet 
the technique has not shown superior effec- 
tiveness compared to angioplasty or conven- 
tional stenting in the iliac arteries, and studies 
on their use for long occlusions elsewhere are 
ongoing. 

Thrombolysis/Thrombectomy 

Thrombolysis may be considered if there is 
acute critical limb ischemia due to thrombosis. 
Thrombolytic agents have the potential to break 
down the clot, restore perfusion, and reveal the 
underlying abnormality that led to thrombosis 
in the first instance. The thrombolytic agents 
with which there is most experience for periph- 
eral use for limb ischemia are recombinant 
tissue-type plasminogen activator (rtPA), strep- 
tokinase, and urokinase. Urokinase was with- 
drawn for some time, but has recently been 
returned to commercial availability. 

Thrombolysis is indicated only if there is crit- 
ical limb ischemia, and only then if the limb is 
salvageable, there is sufficient time for lysis to be 
instigated, and there are no contraindications to 
giving lytic agents. Its use is usually limited to 
the following clinical situations: 

• Graft thromboses 

• Native vessel thromboses 

• Thrombosed popliteal aneurysm with 
occluded runoff 

In general terms the fresher the thrombus, the 
more likely the thrombolysis is to be successful. 
Many clinicians do not consider thrombolysis if 
the thrombus is more than 2 weeks old, based 
partly on the STILE trial data that suggested 
that surgery is associated with a better outcome 
for more chronic ischemia (Weaver et al. 1996). 
When considering thrombolysis, access 
should be as close as possible to the thrombosed 



segment. This may mean performing a prelimi- 
nary ultrasound to identify the upper extent of 
clot if a distal graft or the SFA is thrombosed, to 
ensure ipsilateral access is possible. This allows 
easy access to the thrombus and aids adjunctive 
procedures such as angioplasty or thrombo- 
aspiration if necessary. Use of a single arterial 
wall puncture technique is preferred, and once 
access is achieved the guidewire traversal test 
should be performed. If the wire does not 
pass through the thrombus, the success rate 
for thrombolysis is reduced, as the thrombus 
is likely to be organized. A catheter and/or 
infusion-type wire should be placed through 
the thrombus and lytic agents delivered into the 
thrombus. If the catheter is positioned above the 
thrombosed segment, lytic agents are usually 
carried away by collaterals and the only lytic 
effect will be systemic. A number of infusion 
techniques are available; the most commonly 
used is bolus lacing of the thrombus (e.g., 5mg 
of rtPA or 100,000 to 250,000 units of urokinase) 
followed by a low-dose (e.g., 0.5 mg/h of rtPA, or 
10,000 units/h urokinase) infusion of lytic agent 
with the catheter embedded in the proximal 
extent of the thrombosed segment. There is 
no evidence that a high-dose technique (e.g., 
5 mg/h of rtPA) or pulse spray techniques 
improve outcomes. However, it may be neces- 
sary to use these techniques if the degree of 
ischemia does not allow time for a low-dose 
approach, because although they are labor 
intensive, they do achieve lysis more quickly. 

The catheter and sheath should be secured 
using a suture to minimize the risk of displace- 
ment. Loss of the sheath at the groin may result 
in hemorrhage, and if the catheter is pulled 
back, the effectiveness of the lytic infusion will 
be lost. Patients undergoing lysis are in danger 
of a number of complications, predominantly 
those of hemorrhage and the effects of reperfu- 
sion injury, and should be monitored in a high- 
dependency area. Analgesia should be given 
IV and the amount required reviewed regularly, 
as reperfusion injury may also cause pain. The 
effect of the lytic regime is checked angio- 
graphically after an appropriate period, and if 
necessary the catheter position changed and 
lysis continued. If there has been no effect after 
a reasonable period, then lysis is deemed to have 
failed and alternative surgical strategies may 
be considered. Complications may lead to the 
procedure being abandoned. If the thrombus 



249 



ENDOVASCULAR APPROACHES AND TECHNIQUES 




clears, partially or completely, then adjunctive 
endovascular or surgical treatment may still be 
necessary. 

There is usually an underlying cause for 
graft or vessel thrombosis, and to maintain 
patency these should be treated immediately 
after they are revealed, usually with angio- 
plasty and/or stenting. If clot clearance is only 
partial, it may be appropriate to attempt percu- 
taneous thrombectomy. This can be simple 
aspiration of clot using a large-caliber catheter 
through a removable hemostatic valve, or using 
specialized mechanical thrombectomy devices 
such as the Amplatz Thrombectomy Device 
(Microvena, White Bear Lake, MN) or the 
Hydrolyser (Cordis). Mechanical thrombectomy 
devices macerate the thrombus, with the residue 
either aspirated or small enough to pass through 
the capillary circulation. Surgical intervention 
may be required even if lysis is apparently suc- 
cessful, for example, fasciotomy for reperfusion 
effects, though more commonly it is needed if 
lysis is only partially successful, with treatments 
such as graft revision or surgical thrombectomy. 

Endovascular Techniques for 
Aneurysmal Disease 

The aim of stent grafting is to line the aneurysm 
and adjacent normal arterial segments with 
graft material, therefore excluding the aneurys- 
mal segment. There are now many commercially 
produced devices, manufactured using a variety 
of stent and graft materials, usually as a fully 
supported device with a metal frame covered by 
graft material. 

Much of the focus in recent years has been on 
the use of stent grafts to treat abdominal aortic 
aneurysms (AAAs). In these circumstances, 
special modular bifurcated devices are the most 
frequently used to exclude the aneurysmal 
infrarenal aorta and allow flow into both iliac 
arteries. As with any stent graft procedure, it is 
necessary to have a segment of normal-caliber 
vessel above and below the aneurysm to anchor 
the device and produce exclusion of the sac 
from pressure (and preferably flow as well). 
Initially, only about 5% of infrarenal AAAs 
could be treated with this technique, but this 
expanded to 25% to 30% of infrarenal AAAs 
with the advent of bifurcated and aorto-uniiliac 



grafts. At the present time, 50% or more of 
infrarenal AAAs fulfill the necessary criteria to 
allow stent grafting depending on device avail- 
ability and the criteria applied for iliac access 
and sealing. Common reasons for unsuitability 
are an inadequate aortic "neck" below the renal 
arteries; poor access, with small-caliber or tor- 
tuous iliac arteries; and extensive iliac aneurys- 
mal disease. To some extent, problems with iliac 
access can be dealt with by using iliac or 
iliac-femoral conduits, but this diminishes 
some of the advantages with regard to morbid- 
ity and recovery time. Extensive iliac aneurys- 
mal disease can also be handled by coiling or 
covering the internal iliac arteries, but this 
is not without some risk of added morbidity 
compared to standard endovascular repair. 
Although undoubtedly a feasible technique with 
good short-term outcomes, concerns about the 
long-term results, especially the durability of 
the devices, mean that patients require lifelong 
surveillance, and in the United Kingdom the 
technique is offered only as part of an ongoing 
study. Lifelong surveillance and participation in 
organized data collection or a clinical trial are 
also recommended in the U.S., but commercial 
use of stent grafts for endovascular aneurysm 
repair has been accepted in most parts of the 
country following Food and Drug Adminis- 
tration (FDA) approval of the first devices in 
1999. 

Although most commonly applied to 
infrarenal AAAs, stent grafts can be used for 
a number of other applications: to exclude an 
aneurysmal section of a single vessel such as the 
iliac artery or the thoracic aorta; to treat vessel 
rupture, including partial aortic transection fol- 
lowing deceleration injury, or iatrogenic rupture 
from balloon angioplasty; and to treat the acute 
ischemic complications of thoracic aortic dis- 
section. In the case of thoracic aortic dissection, 
this is achieved by stent grafting the true lumen 
covering the entry tear, which usually lies just 
beyond the origin of the left subclavian artery. 
As a result there is a shift of blood flow from the 
false to the true lumen and a resolution of many 
of the ischemic complications that frequently 
occur due to compression of the true lumen by 
a high-pressure false lumen. Although coverage 
of the entry tear achieves the acute objectives, 
stable long-term fixation of the device should 
be in the normal aorta proximal and distal to 
the dissection to prevent erosion of the device 




> 



250 



VASCULAR SURGERY 



through the weakened segment of the aortic 
wall. Treatment of acute dissections requires a 
thorough understanding of the physiology 
related to flow in the false lumen, and may 
require adjunctive techniques such as balloon 
fenestration of the dissection distally or stent- 
ing of branch vessels. 

The endovascular techniques used during 
stent grafting are essentially those of stenting 
generally. Devices are sized in terms of length 
and diameters to ensure adequate anchorage 
and to produce a seal proximally and distally 
and to cover the aneurysmal section of vessel, 
according to manufacturer guidelines. Appro- 
priate selection of patient anatomy, device type, 
device diameter, and length are all crucial to the 
success of the procedure (Broeders et al., 1997; 
Fillinger, 1999; Wyers et al, 2003). Being large, 
most stent graft devices have to be introduced 
through an arterial cutdown at the groin, 
although recently the successful use of arterial 
closure devices for total percutaneous repair 
have been described. Also, as the devices are 
often bulky and fairly rigid, very stiff 
guidewires, such as the Amplatz Superstiff, Lun- 
derquist (Cook Inc., Bloomington, IN) or Meier 
(Boston Scientific, Boston, MA), are used to 
facilitate tracking of the device into position for 
deployment (Fillinger, 1999; Wyers et al, 2003). 
Accurate deployment is vital to ensure that renal 
or internal iliac arteries are not inadvertently 
covered by the graft material, and this usually 
requires adjusting the gantry angle for the 
aortic neck angle. Deployment for most systems 
involves the unsheathing of the device, but 
deployment mechanisms may include self- 
expansion (e.g., Medtronic AneuRx or Talent), a 
release mechanism for a self-expanding device 
(e.g., Gore Excluder), a combination of these 
(e.g., Cook Zenith), or balloon expansion after 
retraction of the sheath (e.g., Edwards Lifepath). 
Most devices require adjunctive or optional 
balloon inflation to seat the device within the 
vessel. In part this is because hooks or some 
other attachment device may be present to 
help anchor the proximal or distal stents. 
For modular devices, the different parts are 
deployed separately, reducing the size of the 
individual delivery systems. 

At completion, angiography is performed to 
ensure that there is no evidence of leak into the 
aneurysm sac. The key for completion angiog- 
raphy is to confirm the absence of type I (attach- 
ment site) or type III (stent-graft junction) 



endoleaks, which are the most dangerous and 
should be treated prior to leaving the endovas- 
cular suite if at all possible. Type IV "graft 
porosity" endoleaks should seal spontaneously 
in 24 hours, and type II [lumbar artery or infe- 
rior mesenteric artery (IMA) branch] endoleaks 
usually seal spontaneously as well, over a period 
of weeks to months. Care should be taken not to 
assume that the endoleak is a more benign 
variety, but to confirm the source prior to 
removing guidewires and catheters. Confir- 
mation may require maneuvers such as retro- 
grade injection near the distal end point, 
injection within the graft at the location of a 
modular junction, or other techniques. 

Following endovascular aneurysm repair 
(EVAR) it is essential that adequate imaging sur- 
veillance be performed, as set out in Chapter 4. 
This should enable late complications to be 
detected with a view to appropriate treatments, 
as necessary, whether by endovascular or con- 
ventional surgical means. 



Embolotherapy 



Embolization techniques involve the deliberate 
blocking of blood vessels. This may be neces- 
sary to stop arterial hemorrhage, to exclude 
an aneurysm or pseudoaneurysm, to occlude 
a branch vessel that may contribute to an 
"endoleak" as part of endovascular aneurysm 
repair, or to treat vascular tumors and arteri- 
ovenous malformations. It is beyond the scope 
of this chapter to describe the use of emboliza- 
tion in detail for all these indications, but 
some general principles and techniques are 
described. 

In all cases of embolization there are four 
main considerations: 

• What is the anatomy? 

• What needs embolizing? 

• What embolic material should be used? 

• Maintain patient safety. 

Usually noninvasive testing or the clinical sce- 
nario will indicate the likely answers to the 
above considerations. However, an understand- 
ing and knowledge of the vascular anatomy is 
vital. This can usually be helped by high-quality 
angiography, which should be performed before 
embolization begins, during embolization as 
flow dynamics may change, and at completion. 



251 



ENDOVASCULAR APPROACHES AND TECHNIQUES 




Once the anatomy becomes clear, then the target 
for embolization should also become clear. It 
may be a whole vascular bed or a single inflow 
vessel. The type of embolic material is dictated 
by the embolization site and whether perma- 
nent or temporary occlusion is required. During 
embolization procedures, a separate trolley 
should be used for the preparation of the 
embolization material. In addition, to avoid 
accidental injection of the material, all contam- 
inated syringes, contrast, and saline should be 
discarded and replaced. 

Before starting, consideration should be 
given to the approach to the embolization site. 
The shortest, straightest route is usually the 
best. Preliminary angiography may reveal the 
need for equipment, such as guiding catheters 
or microcatheters. Single end-hole catheters 
should always be used, and a guide catheter may 
be used to help ensure that a stable catheter 
position is achieved. When embolizing, always 
consider the effects of collaterals, as they may 
carry embolic material away from their target 
and damage normal tissues, in which case the 
catheter position needs to be closer to the target. 
When embolizing a vessel that is not an end 
artery, it is important to "close the back door" to 
prevent potential back-filling once the entry 
vessel is occluded. If this is not done, then a 
lesion will not have been effectively treated, and 
as the "front door" has been closed, further 
access for embolization will be impossible. 

The main embolic agents to choose from are 
the following: 

Particles: for example, Gelfoam, polyvinyl 

alcohol (PVA) microspheres 
Mechanical devices: for example, coils and 

balloons 
Liquids: for example, sclerosants, glues 

When particles are injected they flow into 
distal vessels, occluding the vessels according to 
the age of the particles used. Correct selection 
of the size of the particles is important; small 
particles may pass through a lesion, causing 
undesirable embolization, whereas large parti- 
cles may not successfully occlude the vascular 
bed. Gelfoam is a temporary embolic material, 
most commonly used by cutting up a sheet of 
Gelfoam into pledgets about 1 mm X 1 mm, that 
when mixed with contrast can be injected 
through a conventional catheter. PVA is a per- 
manent embolic agent that comes in a range of 



sizes from 150 to 1000 |J.m in diameter. When 
injecting particles, it is important to ensure that 
full stasis is not obtained before all embolic 
material in the catheter has been injected; oth- 
erwise further injection will produce significant 
reflux. Flush the embolic material mixed with 
contrast from the catheter with saline, and then 
it is safe to perform angiography. 

Coils are permanent embolic agents that act 
as a focus for thrombosis within the vessel. They 
are metallic, but covered by thrombogenic 
threads. They are best used to occlude a single 
vessel or number of discrete feeding vessels to 
isolate a vessel or area of the circulation. Coils 
come in a variety of sizes. The wire diameter 
varies from 0.014 to 0.038 inch and is important 
to consider, as a large gauge will not pass down 
a small-lumen catheter, such as a micro catheter. 
Coil length also varies, with shorter coils being 
most easily managed. The formed coil diameter 
dictates the size the coil will form in the vessel 
and should be appropriate for the location being 
embolized: too small and the coil may migrate; 
too large and it may not form well in the vessel. 
Always check the stability of the catheter posi- 
tion prior to deploying the first coil. This is done 
by advancing the pusher wire to the end of the 
catheter and ensuring the catheter tip does not 
recoil or advance. The appropriate wire is stiff 
enough to push the coil but not so stiff as to 
dislodge the catheter. A Wholey wire has a 
straight flexible tip with a prominent radio- 
paque mark on the tip to distinguish it from the 
coil being pushed. If this wire is too stiff, a 
Bentson or other more flexible wire might 
be preferable. The coil is introduced into the 
catheter by pressing the coil introducer into the 
catheter hub and using the appropriate pusher 
wire to push the coil into the catheter, then out 
the distal end of the catheter, deploying the coil 
in the appropriate place. 

Liquid agents are permanent agents and the 
most potent. They have the potential to rapidly 
produce hemodynamic changes resulting in 
inadvertent embolization and can be difficult to 
control in this respect. As such, their use should 
be limited to those with the most experience. 
Absolute alcohol causes immediate thrombosis 
as it enters a vessel. It is the ultimate permanent 
embolic agent and produces so much pain that 
patients usually require general anesthesia. Its 
principal use is for vascular malformations. 
Glues, such as cyanoacrylate, solidify on contact 
with ionic substances in blood, and are injected 




> 



252 



VASCULAR SURGERY 



with lipiodol and dextrose solutions to ensure 
they do not solidify in the catheter. Once again, 
it is an unforgiving material but can be useful 
and does not produce the severe pain likely with 
other liquid agents. In general, liquid agents are 
best mixed with a contrast agent if they are not 
already radiodense, so the amount of injection 
and extent of delivery can be detected under 
continuous fluoroscopic guidance. 



References 



Broeders IA, Blankensteijn JD, Olree M, Mali W, Eikelboom 

BC. (1997) J Endovasc Surg 4:252-61. 
Fillinger ME (1999) Surg Clin North Am 79:451-75. 
Weaver FA, Comerota AJ, Youngblood M, Froehlich J, 

Hosking JD, Papanicolaou G. (1996) J Vase Surg 24: 

513-21. 
Wyers MC, Fillinger MF, Schermerhorn ML, et al. (2003) J 

Vase Surg 38:730-8. 



Index 



Abdomen, vascular injuries to, 128-130 
Abdominal aortic aneurysms, 12, 13, 193-207 
clinical features of, 194 
complications of, 204 
definition of, 193 
diameter of, 196-197 
endovascular repair of, 202-204, 249-250 

anesthesia for, 67 

long-term costs of, 25 
epidemiology of, 193 
familial, 192 
inflammatory, 206-207 
non-ruptured (elective), management of, 196-204 

anesthesia for, 66 

aneurysmal expansion rate in, 197 

aneurysmal size in, 196-197 

complications of, 200-202 

endovascular repair technique, 202-204 

open repair technique, 199-202 

patient comorbidity in, 197 

preoperative assessment in, 198-199 

proximal extent of aneurysm in, 197 

surgeon's audited results in, 198 
pathogenesis of, 192 
radiological investigations of, 29, 35-36 
risk factors for, 193-194 
ruptured 

gender differences in, 193 

leaking, 205 

as mortality cause, 193,204-206 

operative repair of, 198, 204-206 

in peripheral arterial disease patients, 53 

symptoms of, 205 
screening for, 196 
Accreditation Council for Graduate Medical 
Education (ACGME), 153 



Acrocyanosis, 79 

Activated clotting time (ACT), 41 

Activated partial thromboplastin time (aPTT),41, 

43 
Activated protein C resistance (APCR), 48 
Acute mesenteric ischemia (AMI), 231-234 
Adrenaline, 172 
P-Adrenergic antagonists, 59 
Advanced Trauma Life Support (ATLS), 129 
Afterload, cardiac, aortic cross-clamping-related 

increase in, 68 
Agency for Healthcare Research and Quality 

(AHRQ), 149 
Alternative medical therapy, for lower limb 

ischemia, 95 
Amaurosis fugax, 161, 163 
Ambulatory venous pressure (AVP), 107 
American Board of Surgery, 153 
Amputations 

Buerger's disease-related, 87 

combat injuries-related, 125 

concomitant vascular and nerve injuries-related, 
130 

in diabetic patients, 58 

lower limb ischemia-related, 95, 97-98, 100 

peripheral vascular disease-related, 53 
Analgesia, 241 
"Anatomical snuffbox" 

as arteriovenous fistula access site, 142 

pulse in, 13 
Anesthesia, for vascular surgery, 65-72, 241 

for carotid endarterectomy, 69-70, 172, 176 

for endovascular interventions, 66-67 

general, 65-66, 67-69, 69, 172, 176 

local anesthesia, 240 

in carotid endarterectomy, 172 

for lower extremity vascular surgery, 70-71 



253 




> 



254 



INDEX 



Anesthesia, for vascular surgery {cont.) 

for upper extremity vascular surgery, 71-72 
locoregional, for carotid endarterectomy, 172 
regional, 65-66, 69-70, 71-72 

contraindications to, 71 
risks associated with, 65-66 
tumescent, 67 
Aneurysms, 191-219. See also Pseudoaneurysms 
aortic. See Aortic aneurysms 
axillary arterial, 218 
axillosubclavian arterial, 218-219 
"berry," 88 
carotid arterial, 218-219 

false, 158 

true, 158, 159 
classification of, 191-192 
definition of, 191 
false, 158,202 
femoral arterial, 213-214 
iliac arterial, 214 
mesenteric arterial, 34, 35, 216 
pathogenesis of, 192 
popliteal arterial, 13, 210-213 

duplex imaging evaluation of, 22-23 

as limb ischemia cause, 101, 104 

management of, 211-213 

popliteal pulse in, 93 

ruptured, 211-212 
rupture of, 192,211-212 
subclavian arterial, 185, 186,218 
traumatic, 13 
visceral arterial, 214-217 
Angiitis, cutaneous leukocytoclastic, 85 
Angina, mesenteric, 34-35 
Angiogenic therapy, for limb ischemia, 98 
Angiography 

of abdominal aortic aneurysms, 195-196 
in blunt abdominal trauma patients, 129 
carotid, 156, 159, 165, 178-179, 188 

of the carotid arch, 25 
catheter, 26-28 

of abdominal aortic aneurysms, 35-36 

complications of, 29 

of endoleaks, 36 

of peripheral vascular disease, 31 

pulmonary, 33 
complications related to, 140 
computed tomography 

of peripheral vascular disease, 31 

of the proximal common carotid artery, 187 

pulmonary, 25 
contrast 

of carotid disease, 165 

of lower limb ischemia, 95 
of cystic adventitial disease, 89 
digital subtraction, 26 

of carotid artery disease, 156, 159 

of the proximal common carotid artery, 187 



of the subclavian artery, 186-187 
for endoleak detection, 36, 250 
magnetic resonance, 30 
of carotid disease, 165, 166 
of the mesenteric arteries, 35 
of the proximal common carotid artery, 187 
of pulmonary embolism, 33 
of the renal arteries, 35 
of the subclavian artery, 186-187 
post-angioplasty, 245 

of renal artery stenosis, 35, 223-224 
of subclavian arterial aneurysms, 217 
of thoracoabdominal aortic aneurysms, 209 
Angioplasty 

percutaneous transluminal 

as fibromuscular dysplasia treatment, 88 
mesenteric, 234-235 
as renal artery stenosis treatment, 226 
as supra-aortic trunk disease treatment, 
184-185 
as renal artery stenosis treatment, 225-226 
of the subclavian artery, 187 
techniques in, 244-246 
Angioplasty balloons, 245 
Angiotensin-converting enzyme inhibitors, 88, 

222 
Angiotensin II, captopril blockage of, 225 
Angiotensin II receptor antagonists, 78 
Ankle block, 71 

Ankle-brachial index (ABI), 19, 150-151 
in chronic venous insufficiency, 109 
definition of, 15 
in diabetes mellitus, 19 
effect of exercise testing on, 20 
in lower limb ischemia, 93, 94, 95 
in peripheral arterial disease, 53, 58 
Antibiotics, indications for use of, 241 
Anticoagulation therapy, 66, 99-100, 127 
Antihypertensive therapy, guidelines for, 59 
Antineutrophil cytoplasmic antibodies (ANCAs), 79, 

80,83 
Antioxidant therapy, 8 
Antiphospholipid antibody syndrome, 50 
Antiplatelet therapy, 1, 54-56, 166-167, 172 
Antiplatelet Trialists Collaboration, 56, 166, 167 
Antithrombin III deficiency, 48 
Aorta 

abdominal 

aneurysms of. See Abdominal aortic aneurysms 
diameter of, 193 
palpation of, 14 
thoracic, trauma-related disruption of, 128 
traumatic injuries to, 37, 128 
Aortic aneurysms 

abdominal. See Abdominal aortic aneurysms 
thoracoabdominal, 207-210 
Aortic dissection, 36-37, 100 
Aortic reconstruction, complications of, 137 



255 



INDEX 




Aortic surgery, open 

anesthesia for, 67-69 

pharmacological considerations for, 68-69 
Aortitis, 81,208 

Aortobifemoral bypass, long-term results of, 96 
Aortofemoral bypass, 68, 101-102, 102 
Apolipoproteins, 4-5 

L-Arginine, as lower limb ischemia treatment, 95 
Arterial bypass. See also specific types of arterial 
bypass 

as limb ischemia treatment, 101-102 
Arterial pressure index (API), Doppler, 126 
Arterial reconstruction, complications of, 134-135 
Arteriography. See also Angiography 

of abdominal aortic aneurysms, 195-196 

arch, of innominate artery occlusive disease, 183 

of axillary artery aneurysms, 218 

of popliteal arterial aneurysms, 21 1 
Arteriovenous access grafts 

autogenous. See fistulas, arteriovenous 

nonautogenous, 141, 143 
complications of, 145-146 
Arteriovenous malformations, pulmonary, 33-34 
Arteritis 

giant cell, 80, 160, 163, 181 

radiation, 160, 185 

Takayasu's, 32, 80-81, 157, 159, 163, 181, 185 
Artery of Adamkiewicz, ligation of, as paraplegia 

cause, 202 
Aspirin, as hemorrhage risk factor, 39, 40 
Aspirin therapy, 54-55, 76, 166, 167, 241 
Asymptomatic Carotid Atherosclerosis Study 

(ACAS), 169, 170-171, 173, 179 
Asymptomatic Carotid Surgery Trial (ACST), 169, 

170-171,173 
Atherectomy, 247-248 
Atheroma, 5-6 
Atherosclerosis, 1-8 

of the carotid artery, 156-157 

clinical history of, 10 

epidemiology of, 1-2 

of the innominate artery, 181 

lipid metabolism in, 3-5 

lipid profiles in, 59 

as lower limb ischemia cause, 91, 92, 93 

pathogenesis of, 5-7 

prevention of, 7-8 

radiation-induced, 181 

regression of, 6-7 

risk factors for, 2-5 

of the subclavian artery, 185 

as systemic vascular disorder, 53 

theories of, 5-7 

as thoracoabdominal aortic aneurysm cause, 208 
Atherosclerotic plaques, 5-7 

carotid, 157-158 

fibrous, 5 

fibrous cap of, 5, 157 



rupture of, 7, 157-158 

stable versus unstable, 7 
Atrial fibrillation, 156, 167-168 
Atriofemoral bypass grafting, 68 
Atrophie blanche, 105, 108 
Atropine, 241 
Auscultation, 14 
Axillary artery 

cystic adventitial disease of, 89 

traumatic injuries to, 130 

as vascular access site, 243 
complications of, 140 
Axillary artery/vein dissection, anesthesia for, 71 
Axillobifemoral bypass, in abdominal trauma 

patients, 130 
Axillofemoral bypass, 66, 68, 71, 102 
Axillosubclavian artery aneurysm, 217 
Azathioprine, as giant cell arteritis treatment, 160 

B 

Bandaging, multilayer lymphedema (MLLB), 120, 

121 
Banding, of arteriovenous fistulas, 145 
Benzodiazepines, 241 

Benzpyrenes, as lymphedema treatment, 122 
Beta blockers, use in peripheral arterial disease 

patients, 59 
Bleeding disorders, 39-42. See also Coagulation 

disorders 
Blood flow 

mesenteric, 231 

normal venous, 106-107 
Blue digit syndrome, 31 
Blunt trauma, 125 

abdominal, 128-129 

to the carotid artery, 127 

to the extremities, 130, 131 

to the neck, 127, 160 
Bovine branch anomaly, 182 
Brachial artery 

distal, cystic adventitial disease of, 89 

as vascular access site, 243 
Brachial plexus, traumatic injuries to, 126, 130 

clinical assessment of, 128 
Brachial plexus blocks, 71-72 
Breast cancer, mortality rate in, 54 
Breast cancer treatment, as lymphedema-related 

pain cause, 120 
British Hypertension Society, 59 
Bruits 

carotid, 14, 93, 164-165, 183 

subclavian, 183 

vascular injury-related, 126 
Buerger, Leo, 86 
Buerger's disease, 31, 86-87, 91 
Buerger's test, 14 
Bullets, velocity of, 126 
Bupivacaine, 66, 240 




256 



INDEX 



Calcitonin gene-related peptide, 74, 78-79 
Calcium channel blockers, as Raynaud's 

phenomenon treatment, 76, 77-78 
Calf muscle, venous blood pumping function of, 

106-107 
Capillaroscopy, 76 
Capillary refill test, 13 
CAPRIE (Clopidogrel versus Aspirin in Patients at 

Risk of Ischemic Events) study, 56, 167 
Captopril, 225 

Carbon dioxide, as vascular contrast media, 27-28 
Cardiac failure, high-output, 145 
Cardiovascular disease, as mortality cause, 1 
Carotid arteries 

aneurysms of, 218-219 
false, 158 
true, 158, 159 
dissection of, 127, 129, 158-159, 163 
external, in carotid endarterectomy, 171 
internal 

in carotid endarterectomy, 171, 172-173 
elongation and kinking of, 158 
fibromuscular dysplasia of, 158 
occlusive disease of, vertebral artery occlusive 

disease-associated, 189 
thrombosis of, as stroke cause, 156 
proximal common, occlusive disease of, 187-188 
stenosis of, 22, 182 
tortuous, 33, 158 
traumatic injuries to, 127, 160 
Carotid artery disease, 155-180 
clinical presentation of, 161-163 
epidemiology of, 155 
investigation of, 164-166 
management of, 166-175 
medical, 166-168 
surgical, 169-175 
pathology of, 157-160 
postoperative management of, 175-179 
prognosis of, 164 

radiological investigations of, 32, 33 
Carotid body tumors (CBTs), 32, 160 
Carotid bypass, 175 
Carotid-sub clavian bypass, 187 
Carotid surgery, anesthesia in, 69-70 
Catheterization 

central venous, 141, 142 
epidural, 66 

equipment for, 237-240 
as vascular injury cause, 126 
Catheters, 238-240 

central venous, 141, 142, 143-144, 146-147 

complications related to, 140, 146-147 

for embolotherapy, 251 

flushing of, 101, 147,239,240-241 

placement of, 244 

types of, 239-240 



CAVATAS study, 180 
Cavography, 109 
Celiac artery 

aneurysms of, 216 

duplex ultrasound examination of, 234 
Cerebral perfusion pressure (CPP), 69 
Cervical plexus block, 69, 70 
Cervical rib, 76, 185, 187, 217 
Charles operation, for lymphedema, 122 
Chest, vascular injuries to, 128 
Chewing tobacco, as Buerger's disease cause, 86 
Cholesterol, hepatic production of, 3 
Churg, Jakob, 84 
Churg-Strauss syndrome, 84-85 
Chylomicrons, 3, 4 
Cilostazol (Pletal), 61-62, 95 
Claudication 

as amputation cause, 95 

chronic limb ischemia-related, 10 

cystic adventitial disease-related, 88 

exercise testing evaluation of, 20 

innominate artery occlusive disease-related, 181 

intermittent, 60 

drug therapy for, 61-62 

dyslipidemia-related, 58 

effect of smoking cessation on, 2 

exercise programs for, 60 

lower limb ischemia-related, 53, 91, 92-93, 95, 

98 
medical management of, 54 
natural history of, 53 
risk factors for, 55, 58 
of the jaw, 163 
neurogenic, differentiated from vascular 

claudication, 92-93 
thoracic outlet syndrome-related, 186 
upper-limb, 1 1 
venous, 108 
Clinical status, 150-151 
Clopidogrel, 56, 166, 167,241 
Clopidogrel versus Aspirin in Patients at Risk of 

Ischemic Events (CAPRIE) study, 56 
Closure devices, arterial, 25, 26, 242 
Clotting disorders. See Coagulation disorders 
Coagulation cascade, 42 
Coagulation disorders 
acquired, 46-47 

complications related to, 139-140 
inherited, 42-46 
Cogan's syndrome, 81-82 
Coils, embolic, 251 
Colic artery, aneurysms of, 217 
Colonic ischemia, postoperative, 202, 206 
Colonoscopy, 202 

Colorectal cancer, mortality rate in, 54 
Common femoral artery, orthopedic surgery-related 

injuries to, 126 
Compartment pressures, measurement of, 103 



257 



INDEX 




Compartment syndrome, 102-103, 103, 130 
Compression injuries, abdominal, 128-129 
Compression therapy, 110-111, 121 
Computed tomography (CT), 28-30. See also 
Angiography, computed tomography 

of abdominal aortic aneurysms, 36, 195, 199, 
205 

of arteriovenous fistulas, 123 

of carotid disease, 165-166 

of inflammatory abdominal aortic aneurysms, 
207 

of innominate artery occlusive disease, 183 

of lower limb ischemia, 93, 95 

of lymphedema, 119 

of popliteal arterial aneurysms, 21 1 

of subclavian arterial aneurysms, 217 

of thoracic trauma, 128 

of thoracoabdominal aortic aneurysms, 208 
Congestive heart failure, incidence of, 2 
Connective tissue disorders, Raynaud's 

phenomenon-related, 75, 76 
Continuous quality improvement (CQI), 152-153 
Contrast agents, 26, 27-28 

first use of, 25-26 

nephrotoxicity of, 27, 33, 95, 224 

reactions to, 242 
Cooperative Study of Renovascular Hypertension, 

221-222 
Corona phlebectatica (ankle/malleolar flare), 105, 

108 
Coronary artery disease (CAD) 

carotid disease-related, 65 

cholesterol as risk factor for, 6 

incidence of, 2 

as mortality cause, 1 

peripheral arterial disease-related, 53 

postoperative, 133 

prevention of, with cholesterol-lowering therapy, 
7,8 

supra-aortic trunk disease-associated, 183 
Coronary-subclavian steal syndrome, 186 
Corticosteroids, as giant cell arteritis treatment, 80, 

160 
Cost outcomes, in vascular surgery, 152-153 
Cranial nerve palsy, 159, 218-219 
C-reactive protein, in giant cell arteritis, 80 
Critical closing pressure (CCP), in lower limb 

ischemia, 93 
Cross-clamping, aortic, effect on cardiac afterload, 

68 
Crush injuries, 126, 128 
Cryoglobulinemia, essential mixed, 85 
Cryoglobulins, 85-86 
Cyanoacrylate, 252 

Cyanosis, Raynaud's phenomenon-related, 75 
Cyclophosphamide, 84, 160 
Cystic adventitial disease, 88-89 
Cytokines, 6, 157 



Debridement, 22, 130 
Deceleration injuries, 126, 128-129 
Decongestive lymphedema therapy (DLT), 120 
Deep venous reconstruction, as varicose veins 

treatment, 112-113 
Deep venous thrombosis 

abdominal aortic aneurysm repair-related, 202 

chronic venous insufficiency-related, 107 

hypercoagulability-related, 61 

as lymphedema cause, 118 

medical management of, 54 
Dense granule deficiency, 46 
Dermatitis, chronic venous insufficiency-related, 

105,108,110 
Diabetes mellitus 

ankle-brachial index (ABI) in, 19 

as atherosclerosis risk factor, 1,2 

hypertension management in, 59, 167 

lower limb ischemia associated with, 91, 97 

negative correlation with abdominal aortic 
aneurysms, 193-194 

peripheral arterial disease associated with, 55, 58, 
59 
Dialysis Outcomes and Practice Patterns Study 

(DOPPS), 142 
Dialysis patients, vascular access in, 141-148 
Diazepam, 66-67 

Digital pressures, measurement of, 20 
Diphenhydramine, 66-67 
Dipyridamole, 56, 166, 241 
Distal revascularization-interval ligation (DRIL), 

145 
Dorsalis pedis artery, absence of, 19 
Dressings, for chronic venous insufficiency 

management, 109-110 
Drug injection, intraarterial, adverse effects of, 

16-17,131 
Drugs. See also names of specific drugs 

as bleeding risk factor, 39, 40 
Dysarthria, 163 
Dysfibrinogenemias, 49 
Dyshypoplasminogenemias, 49 
Dyslipidemia, management of, 58-59 
Dysphagia, 163 
Dysplasia, fibromuscular, 87-88 

of the internal carotid artery, 158 

radiological investigations of, 35 

as renal artery stenosis cause, 222, 225 

as stroke cause, 157 

subtypes of, 158 



Eczema, 105, 108, 120 
Edema 

chronic venous insufficiency-related, 108-109 

pulmonary, "flash," 224 
Ehlers-Danos syndrome, 158-159 




» 



258 



INDEX 



Elastases, 7 

Elastic bandages, 110-111, 121 
Elderly patients, brachial emboli in, 12 
Electrocardiography, preoperative, 198 
Elephantiasis, as lymphedema cause, 118 
Embolectomy. See also Embolotherapy 

as lower limb ischemia treatment, 100-101, 103 
Embolic agents, 251-252 
Embolism 

cardiac, as ischemic stroke cause, 156 
cholesterol, 161, 162 
common sources of, 11 
distal, of the supra-aortic trunk, 189 
endovascular abdominal aortic aneurysm repair- 
related, 204 
as lower limb ischemia cause, 98-99, 100-101, 

103,104 
mesenteric, 232, 233 
pulmonary, 32-33, 202 
as upper limb ischemia cause, 16, 17 
as upper limb vascular disease cause, 1 1 
Embolotherapy, 250-252 
Endarterectomy 
carotid, 169-179 

anesthesia for, 66, 69-70 
atherocsclerotic plaque excision in, 157 
bifurcation, of the proximal common carotid 

artery, 188 
cerebral protection in, 69 
comparison with carotid angioplasty, 179-180 
effect on stroke risk, 169-171 
monitoring during, 66 
neurological deficits associated with, 

176-178 
operative principles of, 171-175 
postoperative management of, 175-179 
preoperative hypertension control for, 167 
traditional versus eversion, 174 
in vertebral artery stenosis/occlusion, 189 
transaortic renal, 226 
transthoracic innominate, 183, 184 
Endoleaks, 36, 204, 250 

Endothelin-1, in Raynaud's phenomenon, 74-75 
Endovascular approaches and techniques, 237-252. 
See also Embolotherapy; Stenting; Stent 
grafting 
for abdominal aortic aneurysms, 67, 202-204 

conversion to open repair, 204 
access techniques, 242-244 
anesthesia in, 66-67 
for aneurysmal disease, 249-250 
for arterial occlusive disease, 244-249 
for arterial trauma, 131 
for carotid aneurysms, 218, 219 
equipment for, 237-240 
medications used in, 240-242 
radiological investigations with, 25-26 
for renal artery stenosis, 226-227 



for supra-aortic trunk disease, 184-185 

for Takayasu's arteritis, 81 

for thoracoabdominal aortic aneurysms, 210 

vascular intervention devices for, 242 
Epidural anesthesia, 66, 71 
Errors, medical, 149 

Erythrocytes, in Raynaud's phenomenon, 75 
Erythrocyte sedimentation rate, 80, 83, 198 
Eryfhromelalgia, 79 
Estrogen, 39, 75 

European Carotid Surgery Trial (ECST), 169-170 
EuroQol, 152 
Exercise 

health benefits of, 60 

lymphedema-exacerbating effects of, 122 
Exercise testing, 20 

in lower limb ischemia patients, 94-95 
Exercise therapy 

for lower limb ischemia patients, 95 

for peripheral arterial disease patients, 60 



Factor deficiencies, 42-45 
Factor inhibitors, 46-47 
Fasciotomy, 102-103, 130 
Fatty streaks, 5 
Femoral arteries 

aneurysms of, 13,213-214 

superficial 

aneurysms of, 13 

distal, occlusive disease of, 96 

as hemodialysis access conduit, 147 

traumatic injuries to, 130-131 

as vascular access site, 243 
Femoral artery punctures, complications of, 131, 140 
Femoral-femoral bypass, 96, 101-102 
Femoral-popliteal bypass, 96 
Femoral shaft fractures, 126 
Femoral-tibial bypass grafting, 97 
Fentanyl, 67 
fibrinogen, 41 

fibrinogen abnormalities, 45 
filiariasis, as lymphedema cause, 118 
fistulas 

aortocaval, 194 

aortoduodenal, 194 

arteriovenous, 123, 124, 128, 131, 141, 142-143 
complications of, 144-145 

carotid-cavernous sinus, 128 
flavonoids, as lymphedema treatment, 122 
fluid management, perioperative, in patients with 

ruptured aortic aneurysms, 201 
Framingham Study, 6 
Functional status, 151-152 



Gadolinium chelate, 28 

Gangrene, 17, 91, 92, 93, 94, 96-97, 131 



259 



INDEX 




Gastroduodenal artery, aneurysms of, 216-217 

Gingko, 40, 95 

Glomerulonephritis, Wegener's granulomatosis- 
related, 84 

Glucocorticoids, as Takayasu's arteritis treatment, 
81 

Glucose tolerance test, 58 

Glues, embolic, 252 

Glyceryl trinitrate, 241 

Glycopyrronium bromide, 241 

Grafts, peripheral vascular. See also Stent grafting 
complications related to, 134-139 
infrainguinal, duplex imaging of, 22 
lower limb, occlusion of, 100, 136-137 
as lower limb ischemia risk factor, 91 
polytetrafluoroethylene, 96, 97, 102, 135, 

136 
prosthetic, complications of, 137-139 
in supra-aortic trunk disease, 183-184 

Granulomatosis, Wegener's, 83-84 

Gray platelet syndrome, 46 

Greater saphenous vein, radiofrequency or laser 
ablation of, 67 

Greater saphenous vein grafts, 135 

Growth factor therapy, for limb ischemia, 98 

Guidewires, 237-238, 244-245 

introduction through needles, 244 

Gunshot wounds, 126, 129 

H 

Hand surgery, anesthesia for, 72 

Health Plan Employer Data and Information Set 

(HEDIS), 151 
Heart Protection Study, 58, 59 
Hemangiomas, 123 
Hematomas 

of neck incisions, 176 

retroperitoneal, 129 

vascular injury-related, 126, 127, 129 
Hemianopia, homonymous, 162, 163 
Hemophilia, 42-43 
Hemorrhage 

abdominal, trauma-related, 129 

chronic venous insufficiency- related, 109 

femoral artery puncture-related, 131 

intracranial, carotid endarterectomy-related, 
177-178 

perioperative, in abdominal aortic aneurysm 
patients, 201 

thrombolytic therapy- related, 248 

vascular injury-related, 126, 127 

venous, trauma-related, 130 
Henoch, Edouard, 85 
Heparin 

in catheter saline flushes, 101, 147,239,240-241 

as contraindication to epidural catheter use, 
66 

as hypercoagulability treatment, 47, 48 



intraoperative administration of, 173,240-241 

as thrombocytopenia cause, 50-51 
Hepatic artery, aneurysms of, 215-216 
Hepatitis C virus, 85, 86 
Herbal remedies, as hemorrhage risk factor, 39,40, 

41 
Hexopal (inositol nicotinate), 61-62 
High-density lipoprotein, 2, 4, 168 
Hollenhorst plaques, 161 
Homan operation, for lymphedema, 122 
Homocysteine, 3, 48-59 
Homocystinemia, 61 
Horner syndrome, 163 

partial, 159 
Human immunodeficiency virus (HIV) infection, 

118 
Hunter, William, 125 
Hydro thorax, 37 

3-Hydroxy-3-mefhylglutaryl coenzyme (HMG CoA) 
reductase, inhibition of, 3, 5. See also Statin 
therapy 
5-Hydroxytryptamine. See Serotonin 
Hypercholesterolemia, 1,6, 193 
Hypercoagulable disorders 

acquired, 47, 50-51, 60-61 

inherited, 47, 48-50, 60, 61 

as ischemic stroke cause, 157 

peripheral arterial disease-associated, 60-61 

supra-aortic trunk disease-associated, 183-184 

as vascular thrombosis cause, 139-140 
Hyperhomocystinemia, 47, 48-49 
Hyperhomocystinuria, 48-49 
Hyperlipidemia, 2, 168 

Hyperperfusion syndrome, as stroke cause, 178 
Hyperplasia, myointimal, 135 
Hypertension 

as abdominal aortic aneurysm risk factor, 193 

as atherosclerosis risk factor, 1, 3 

diabetes mellitus-related, 2 
management of, 59, 167 

drug therapy guidelines for, 59 

fibromuscular dysplasia-related, 88 

incidence of, 2 

as peripheral arterial disease risk factor, 59-60 

postoperative, endarterectomy-related, 176 

renovascular, 22 1 -229 

diagnostic evaluation of, 223-224 
pathophysiology of, 222-223 
renal artery stenosis-related, 225-226 
treatment of, 225-228 

as stroke risk factor, 3, 167 
Hypotension 

endarterectomy-related, 176 

vascular injury-related, 126, 128 

I 

Ileal artery, aneurysms of, 217 
Ileus, laparotomy-related, 201-202 




> 



260 



INDEX 



Iliac arteries 

aneurysms of, 214 

cystic adventitial disease of, 89 

external, orthopedic surgery-related injuries to, 
126 
Iloprost, 76, 78 

Imaging. See Radiological investigations 
Impotence, abdominal aortic aneurysm repair- 
related, 202 
Infections 

arteriovenous access graft- related, 146 

arteriovenous fistula-related, 145 

central venous catheter-related, 144, 146-147 

prosthetic graft-related, 137-139 
Inferior mesenteric artery, in abdominal aortic 

aneurysm repair, 199-200,202 
Inferior vena cava 

as hemodialysis access conduit, 147 

traumatic injuries to, 130 
Inflammation 

as aneurysmal disease cause, 192, 206 

as atherosclerosis cause, 6 
Infrainguinal bypass, as acute ischemia treatment, 

102 
Innominate artery 

occlusive disease of, 181-185 

traumatic injuries to, 128 
Inositol nicotinate (Hexopal), 61-62 
Institute of Medicine, "To Err is Human" report, 149 
Intercostal arteries, in thoracoabdominal aortic 

aneurysm repair, 209 
Intermediate-density lipoprotein, 3, 4-5 
Internal jugular veins, traumatic injury to, 127 
Internal mammary artery revascularization, as 

coronary-subclavian steal syndrome cause, 
186 
International normalized ratio (INR), 41 
International Society for Cardiovascular Surgery, 

151 
Interscalene technique, of deep cervical plexus 

block, 70 
Intravenous immunoglobulin, as Kawasaki's disease 

treatment, 82 
Ischemia 

colonic, postoperative, 202, 206 

limb. See Limb ischemia 

mesenteric, 16, 231-234 

popliteal arterial aneurysm-related, 211-212 

Raynaud's phenomenon-related, 76 

visceral. See Visceral ischemic syndromes 
Itching, chronic venous insufficiency- related, 108 



Jejunal artery, aneurysms of, 217 



Kaposi's sarcoma, 118 
Kawasaki, Tomisahu, 82 



Kawasaki's disease, 82 

Ketanserin, 78 

Klippel-Trenaunay- Weber syndrome, 117 

Knee injuries, 126, 131 

Kocher maneuver, 130 

Korean War, vascular surgery during, 125 



Lacunar infarcts, 156, 164 

Language disorders, carotid disease-related, 163 

Laparotomy, as ileus cause, 201-202 

Laser ablation, of the greater saphenous vein, 67 

Leap Frog Group, 149-150 

Lecithin- cholesterol acetyltransferase, 4 

Letessier-Meige syndrome, 117 

Lidocaine, 66, 67, 240 

Lifestyle risk factors, clinical evaluation of, 

10 
Limb ischemia 

lower. See Lower limb ischemia 
upper limb 

clinical examination of, 16-17 
radiological investigations of, 32 
vascular injury-related, 126 
Limb pressures, segmental, 19-20, 21 
Limb reduction procedures, as lymphedema 

treatment, 122 
Limbs, vascular injuries to, 130-131 
Limb salvage, in acute lower limb ischemia, 

103 
Lipedema, 119 

Lipid-lowering therapy, 7, 8, 58-59 
Lipid metabolism, 3-5 
Lipid profile, "diabetic," 58 
Lipodermatosclerosis, 105, 108 
Lipoprotein (a), 2, 4 
Lipoprotein lipase, 4 
Liver, cholesterol production in, 3 
Local anesthesia, 240 

in carotid endarterectomy, 172 

in lower extremity vascular surgery, 

70-71 
in upper extremity vascular surgery, 
71-72 
Long saphenous vein, 106 
Long saphenous vein surgery, for varicose veins, 

111-112 
Lorazepam, 67 
Low-density lipoprotein, 1,2,3, 4-5, 168 

small-density, 2, 4 
Lower limb(s), pallor of, 14 
Lower limb ischemia, 91-104 
acute, 98-104 

clinical examination of, 16 
diagnostic studies of, 99 
five P's of, 99 
outcomes of, 103-104 
patient history of, 98-99 



261 



INDEX 




physical examination of, 99 
treatment of, 99-103 
chronic, 10, 92-98 

abdominal aortic aneurysm-related, 194 
clinical examination of, 16 
diagnostic studies of, 94-95 
outcomes of, 98 
patient history of, 92-93 
physical examination of, 93-94 
treatment of, 95-98 
critical, 97 
acute, 10, 54 
amputations in, 97-98 
hypertension treatment in, 59, 60 
in peripheral vascular disease patients, 53 
thrombolytic therapy for, 248 
etiology and presentation of, 91-92 
postoperative, in abdominal aortic aneurysm 

patients, 201 
subclavian artery aneurysm-related, 217 
treatment of 

amputation, 97-98 
medical, 95 

revascularization, 95-97 
Lung cancer, mortality rate in, 54 
Lupus anticoagulant, 50 
Lymph, formation of, 121-122 
Lymphadenitis, 118 
Lymphangiography, 119 
Lymphangioma circumscriptum, 116 
Lymphangioma diffusum, 116 
Lymphangiomas, 116 
Lymphangiosarcoma, 116, 119 
Lymphangitis, 118 
Lymphatic drainage, manual lymphatic (MLD), 120, 

121 
Lymphatic system, anatomy and functions of, 

113-114 
Lymphedema, 113-122 

acute inflammatory episodes (AIEs) associated 

with, 118, 121 
clinical assessment of, 115-116 
definition of, 114 
differential diagnosis of, 115 
epidemiology of, 1 1 5 
etiological classification of, 109 
factitious, 119 
familial, 117 

lymphatic system in, 114 
management of, 120-122 
pathophysiology of, 114 
primary, 114, 116-118 
relationship with chronic venous insufficiency, 

118-119 
secondary, 114, 118-119 
Lymphedema congenita, 117 
Lymphedema praecox, 117 
Lymphorrhea, 120-121 



M 

Macrophages, 6, 157, 192 
Magnetic resonance imaging (MRI). See also 
Angiography, magnetic resonance 

of abdominal aortic aneurysms, 195 

of carotid disease, 165-166 

of popliteal arterial aneurysms, 211 

of thoracoabdominal aortic aneurysms, 208-209 
Mannitol, 69 

Marfan syndrome, 46, 158-159 
Matrix metalloproteineases, 157, 158, 192 
Mean arterial pressure (MAP), during carotid 

endarterectomy, 69 
Medical Outcomes Short Form 36 (SF-36), 151-152 
Mesenteric arteries 

aneurysms of, 34, 35, 216 

radiological investigations of, 34-35 

superior 

aneurysms of, 216 

duplex ultrasound examination of, 234 
embolization of, as ischemia cause, 231 
thrombosis of, as ischemia cause, 231, 233 
traumatic injuries to, 129-130 

thrombosis of, 231, 232, 233-234 
Mesenteric ischemia, acute, 231-234 
Mesenteric venous thrombosis, 235-236 
Midazolam, 241 
Middle cerebral artery, 156 
Milroy's disease, 117 
Morphine, 241 

Motor deficits, carotid disease-related, 162-163 
Multiple Risk Factor Intervention Trial (MRfiT), 2 
Mupirocin, 146 

Muscle death, acute limb ischemia-related, 99, 100 
Myocardial infarction, 7, 53, 201 
Myxedema, pretibial, 119 

N 

Naftidrofuryl (Praxilene), 61-62 

National Committee for Quality assurance (NCQA), 

151 
National Veterans Administration Surgical Quality 

Improvement Program (NSQIP), 149 
Neck 

anatomic zones of, 127 
innervation of, 69-70 
traumatic injuries to, 126, 127-128, 160 
Neck incisions, hematoma of, 176 
Nephropathy 

contrast agent-induced, 27, 33, 95, 224 
diabetic, lower limb ischemia associated with, 91 
ischemic, 221, 223, 228 
Nerve injuries, vascular injuries-associated, 126, 

130 
Neurological deficits, post-carotid endarterectomy, 

176-178 
Neurological examination, of vascular injury 
patients, 126 




> 



262 



INDEX 



Neuropathy 
diabetic, 71 
ischemic, 104 
Neutrophils, in Raynaud's phenomenon 

pathophysiology, 75 
Nicotine replacement therapy (NRT), 57 
Nifedipine, 76,241 
Nitroglycerin, 68-69, 78, 241 
Nitroprusside, 68, 69 
Nonsteroidal anti-inflammatory drugs, as 

hemorrhage risk factor, 39, 40 
Noone-Milroy syndrome, 117 
North American Symptomatic Carotid 

Endarterectomy Trial (NASCET), 166, 

169-170,174 



Obesity 

adverse health effects of, 57-58 

management of, 57 

in peripheral arterial disease patients, 58 
Ocular disorders, carotid disease-related, 161-162 
Ocular-ischemia syndrome, 161, 162 
Opiates, 241 

Optic nerve, ischemia of, 161, 162 
Orthopedic injuries, treatment for, 130 
Orthopedic procedures, as vascular injury cause, 126 
Osier- Weber-Rendu disease, 46 
Ostial lesions, stenting of, 246 
Outcome measures, in vascular surgery, 149-154 
Ovarian vein reflux, imaging of, 109 
Oxfordshire Community Stroke Project, 164 
Oxypentifylline (Trental), 61-62 



Padma Basic, 95 

Pallor, 14, 16, 75,99 

Palpation, 13-14, 194, 207, 211 

Pancreatic artery, aneurysms of, 216-217 

Pancreaticoduodenal artery, aneurysms of, 216-217 

Papaverine, 241 

Paralysis, acute limb ischemia-related, 99 

Paraplegia, 128, 202 

Paresthesia, acute limb ischemia-related, 99 

Patching, in carotid endarterectomy, 174-175 

Patient satisfaction, 151 

Penetrating trauma, 125 

abdominal, 128 

to the extremities, 130 

to the neck, 127 

thoracic, 128 

weapon-related, 125 
Pentoxifylline, 95 

Perforator ligation, as venous disease treatment, 112 
Peripheral vascular disease 

asymptomatic, in diabetic patients, 58 

atherosclerosis risk factor treatment in, 1 

cholesterol-lowering therapy for, 7 



as coronary artery disease risk factor, 65 

diabetes mellitus associated with, 58 

evaluation instruments for, 152 

as lower limb ischemia cause, 91 

medical management of, 53-63 

vascular risk factor modification, 53-61 

mortality rate in, 53, 54 

radiological investigations of, 31 
Pethidine, 241 
Phleboliths, 123 
Physicians' Health Study, 56 
Platelet-derived growth factor, 54, 135 
Platelet disorders, 45-46 
Platelet factor, 4, 54 
Platelets, in Raynaud's phenomenon 

pathophysiology, 75 
Pletal (cilostazol), 61-62 
Plethysmography, 21, 109 
Pneumothorax, 37 

tension, 128 
Poiseuille's law, 95 
Polyangiitis, microscopic, 85 
Polyarteritis nodosa, 82-83, 85 
Polytetrafluoroethylene grafts, 96, 97, 102, 135, 136 
POPADAD (Prevention of Progression of Arterial 

Disease in Diabetes), 55, 58 
Popliteal artery 

aneurysms of, 13,210-213 

duplex imaging evaluation of, 22-23 
as limb ischemia cause, 101, 103 
management of, 211-213 
popliteal pulse in, 93 
ruptured, 211-212 

cystic adventitial disease of, 89 

occlusive disease of, surgical treatment of, 96 

orthopedic surgery-related injuries to, 126 

traumatic injuries to, 131 

as vascular access site, 243 
Popliteal pulse, in popliteal aneurysm, 93 
Postimplant syndrome, 204 
Postthrombotic syndrome (PTS), 107 
Praxilene (naftidrofuryl), 61-62 
Prazosin, 74 
Prednisolone, as Wegener's granulomatosis 

treatment, 84 
Pregnancy, "white leg" of, 107 
Prevention of Progression of Arterial Disease in 

Diabetes (POPADAD), 55, 58 
Prilocaine hydrochloride, 240 
Primary valvular incompetence (PVI), 107 
Pringle's maneuver, 129 
Prostate cancer, mortality rate in, 54 
Proteases, 7 

Protein C deficiency, 49 
Protein S deficiency, 49 
Prothrombin time (PT),41 
Pseudoaneurysms 

of arterial puncture sites, 131, 140 



263 



INDEX 




arteriovenous access graft-related, 146 

carotid, 127-128 
Pulmonary arteries, imaging of, 32-34 
Pulmonary disease 

implication for anesthesia, 71 

postoperative, 133-134 
Pulseless disease. See Arteritis, Takayasu's 
Pulselessness, acute limb ischemia-related, 99 
Pulselessness, vascular injury-related, 126 
Pulses 

abdominal, 14 

carotid, 13-14 

lower limb, 13 

subclavian, 13 

ulnar, 13 

upper limb, 13 
Pulse volume recordings (PVRs), 20-21 
Purpura, Henoch-Schbnlein, 85 



Quality-adjusted life years (QALYs), 153 

Quality analysis (QA), 153 

Quality Interagency Coordination Task Force, 149 



Radial arteries 

distal, cystic adventitial disease of, 89 
traumatic injuries to, 130 
as vascular access site, 143, 243 
Radiation, as arteritis cause, 160, 185 
Radiofrequency ablation, of the greater saphenous 

vein, 67 
Radiological investigations, 25-38. See also specific 
radiological imaging modalities in 
complications of, 140 
controversies in, 25 
radiation safety during, 30-31 
of trauma, 37 

of vascular diseases, 31-37 
Radionuclide investigations, mesenteric, 34 
Raynaud, Maurice, 73 
Raynaud's phenomenon, 73-79 
primary, 73, 75 

differentiated from secondary, 75-76 
secondary, 73, 74, 75-76 
thoracic outlet syndrome-related, 186 
treatment for, 76-79 
Recombinant tissue-type plasminogen activator, 248 
Renal arteries 

aneurysms of, 216 
radiological investigations of, 35 
resistive index (RI) in, 223 
stenosis of, 221-229 
bruits associated with 
diagnostic evaluation of, 223-224 
etiology of, 222 

experimental models of, 222-223 
medical management of, 225-226 



natural history of, 225 
pathophysiology of, 222-223 
renal-to-aortic ratio (RAR) for, 223 
surgical management, 223, 224, 225-228, 246 
traumatic injuries to, 130 
Renal function, postoperative, in abdominal aortic 

aneurysm patients, 201, 206 
Renal insufficiency. See also Nephropathy, ischemic 
contrast agent-related exacerbation of, 95, 224 
in lower limb ischemia patients, 97 
renal artery stenosis-related, 224 
Renal-to-aortic ratio (RAR), 223 
Renal vein, left, operative exposure of, 227 
Renal vein renin assays, 224-225 
Renography, isotope, 225 
Reperfusion syndrome, as stroke cause, 178 
Residency training programs, outcomes measures 

competency requirement in, 153 
Resistive index (RI),in the renal artery, 223 
Rest pain, 10, 11,92,93,98 
Resuscitation, in abdominal vascular injury 

patients, 129 
Retinal artery, occlusion of, 161, 162 
Rheumatoid arthritis, as lymphedema cause, 

119 
Rib fractures, 128 
Rubor, 14, 15,75 



Saline, heparinized, 101, 147,239,240-241 
Saphenofemoral junction, incompetence of, 14-16, 

16 
Saphenopopliteal junction, ligation of, 112 
Saphenous vein, as carotid endarterectomy patch, 

174-175 
SAPPHIRE study, 180 
Schonlein, Johann, 85 
Sclerotherapy, for chronic venous insufficiency, 

111 
Scottish Intercollegiate Guidelines Network (SIGN), 

59 
Sedation, 241 
Seldinger technique, 26 
Selective serotonin reuptake inhibitors, as 

Raynaud's phenomenon treatment, 78 
Sensory deficits, carotid disease-related, 162-163 
Serotonin, 54, 75 
Serotonin antagonists, as Raynaud's phenomenon 

treatment, 78 
Short saphenous vein, 106 
Short saphenous vein surgery, for varicose veins, 

112 
Shunting, in carotid surgery, 69, 173-174 
Simvastatin, 8, 59 

Sinus nerve, in carotid endarterectomy, 173 
Sistrunk operation, for lymphedema, 122 
Skin perfusion pressure (SPP), 21-22 
"Slipped heart syndrome," 12 




> 



264 



INDEX 



Smoking 

as abdominal aortic aneurysm cause, 193 

as atherosclerosis cause, 1,2 

as Buerger's disease cause, 86, 87 

by occlusive vascular disease cause patients, 12 

as peripheral arterial disease cause, 1, 57 

as stroke cause, 167 

by supra-aortic trunk disease patients, 183 
Smoking cessation 

cardiovascular health benefits of, 2 

effect on stroke risk, 167 

in lower limb ischemia patients, 95 

methods, 57 

in peripheral arterial disease patients, 1, 57 

as weight gain cause, 58 
Society for Vascular Surgery, 151 
Speech disorders, carotid disease-related, 163 
Splenic artery, aneurysms of, 215 
Stab wounds, 126 
Starling's forces, 114 
Statin therapy, 7-8, 59 
Steal, vascular, 145, 146 
Stemmer's sign, 115-116 
Stent grafting, 248 

of abdominal aortic aneurysms, 202-204, 249-250 
migration of stent in, 204 

of carotid artery aneurysms, 219 

devices for, 249, 250 

postimplantation imaging of, 36 

types of stents, 246-247 
Stenting 

intravascular, 246-247 

of renal artery stenosis, 226-227 

of supra-aortic trunk disease, 184-185 

techniques in, 246 

of thoracoabdominal aortic aneurysms, 210 
Sticky platelet syndrome, 49 
Strauss, Lotte, 84 
Streptokinase, 101,248 
"String of sausages" sign, 233 
Stroke 

atrial fibrillation-related, 167-168 

in carotid endarterectomy patients, 176-178 

in carotid territory. See Carotid artery disease 

completed, 161 

definition of, 155 

in evolution, 161 

hemorrhagic, 156 

incidence of, 2 

investigation of, 164-166 

ischemic, 156-157, 164 

as mortality cause, 164 

perioperative, in supra-aortic trunk disease 
patients, 183-184 

peripheral arterial disease-related, 53 

prevention of 

with antiplatelet therapy, 166-167 
with carotid endarterectomy, 169-179 



with risk factor modification, 167-168 
vertebrobasilar symptoms of, 161, 163, 188-189 
Subclavian arteries 

aneurysms of, 185, 186,217 

as arteriovenous fistula access site, 142 

occlusive disease of, 185-187 

cerebrovascular symptoms of, 1 1 
revascularization of, 187 
right, stenosis of, 185 
traumatic injuries to, 128, 130 
Subclavian steal, 186 
Subclavian vein, compression of, 186 
Subfascial endoscopic perforator surgery (SEPS), 

duplex ultrasound-guided, 112 
Suction drains, 175 
"Sunset foot," 14 

Sympathectomy, as Raynaud's phenomenon 
treatment, 74, 76-79 



Takayasu, Mikito, 80 

Thiazides, 59 

Thompson operation, for lymphedema, 122 

Thoracic outlet syndrome, 185, 186,217 

Thoracic spine, traumatic injuries to, 128 

Thoracotomy 

emergent, in thoracic vascular injury patients, 
128 

in thoracoabdominal aortic aneurysm repair, 209, 
210 
Thrombectomy, 249 
Thrombin inhibitors, 47 

Thromboangiitis obliterans. See Buerger's disease 
Thromboasthenia, Glanzmann's, 45, 75 
Thrombocytopenia, heparin-induced, 50-51 
Thromboembolism. See also Deep venous 
thrombosis 

carotid artery aneurysm-related, 218 

as ischemic carotid territory stroke cause, 156 

subclavian artery fhromboembolism-related, 185 

venous, hypercoagulability-related, 48 
Thromboendarterectomy, 227, 235 
p-Thromboglobulin, 54 
Thrombolytic therapy, 101, 248-249 
Thrombophilia 

acquired, 60-61 

inherited, 60, 61, 105-106 

mesenteric venous thrombosis-related, 235 

peripheral arterial disease-related, 60-61 
Thrombophilia screen, 41 
Thrombophlebitis, superficial venous, 118 
Thrombosis. See also Deep venous thrombosis 

as acute lower limb ischemia cause, 98-99, 104 

arteriovenous access graft- related, 146 

arteriovenous fistula-related, 145 

central venous catheter-related, 147 

of chronic arterial stenosis, 10 

of collateralized chronic stenosis, 16 



265 



INDEX 




hypercoagulability- related, 139-140 

post-endarterectomy, 176-177 
Thromboxane A 2 , 54, 75 
Tibial nerve, traumatic injuries to, 131 
Ticlopidine, 166-167 
Tissue factor, 42 
Tobacco use. See also Smoking 

as Buerger's disease cause, 86, 87 
Toe-brachial index (TBI), in lower limb ischemia, 94 
Topical agents 

for chronic venous insufficiency-related 
dermatitis, 110 

for lymphedema-related eczema, 120 
Trancutaneous oxygen testing (TcP0 2 ), 97-98 
Transient ischemic attacks (TIAs), 161 

definition of, 155 

epidemiolgy of, 155 

innominate artery occlusive disease-related, 182 

types of, 161 
Trauma 

as acute upper limb vascular disease cause, 11-12 

as axillary artery aneurysm cause, 218 

as carotid artery aneurysm cause, 218 

as innominate artery occlusive disease cause, 181 

as mortality cause, 125 

vascular. See Vascular injuries 
Trendelenburg test, 14-15 
Trental (oxypentifylline), 61-62 
Triglycerides, 4, 58 

U 

Ulceration 

chronic venous, 109 

epidemiology of, 105-106 

management of, 113 

physical therapy for, 110 
foot/lower limb 

differential diagnosis of, 14 

ischemia-related, 91, 92, 93-94, 96-97 

skin perfusion pressure evaluation of, 21-22 
lymphedema-related, 116 
Ulnar arteries 

distal, cystic adventitial disease of, 89 
traumatic injuries to, 130 
Ultrasonography 

of abdominal aortic aneurysms, 194-195, 198-199 
Doppler 

of arterial disease, 15-16, 19-20,21 

of arteriovenous fistulas, 123 

fixed waved, 15-16 

waveform analysis in, 21 
duplex, 22-23, 150-151 

of arteriovenous fistulas, 123 

of axillary artery aneurysms, 218 

carotid, 32, 164-165 

of chronic venous insufficiency, 109 

of cystic adventitial disease, 89 

of popliteal arterial aneurysms, 211 



post-carotid endarterectomy, 178 

of the renal artery, 223 

of subclavian arterial aneurysms, 217 

of the subclavian artery, 186 

of upper limb vascular disease, 32 
of inflammatory abdominal aortic aneurysm, 207 
United Kingdom Prospective Diabetes Study group, 

167 
University of Iowa, 67 
Ureterolysis, 207 
Urokinase, 101,248 



Varices, pelvic, imaging of, 109 
Varicose veins, 12 

arteriovenous fistula-associated, 123 

clinical evaluation of, 14-15 

epidemiology of, 105-106 

physical examination of, 108 

radiofrequency or laser ablation treatment of, 67 

recurrent, surgical treatment of, 112 

secondary, 107 

surgical treatment of, 111-113 
Vascular access, 141-148 

complications of, 144-147 

desperate, 147-148 

equipment for, 237-240 

principles of, 141 

standards for, 141-142 

techniques in, 242-244 

for thrombolytic therapy, 248 

types of, 142-144 
Vascular disease. See also Peripheral vascular 
disease 

clinical evaluation of, 9-18 
clinical examination in, 12-18 
clinical history in, 9-12 
patient's characteristics, 9 
radiological studies in, 18 

diabetes mellitus-related, 2 

lipoproptein (a) as risk factor for, 4 

nonatherosclerotic, 73-89 

miscellaneous disorders, 86-89 
vasculitis, 79-86 
vasospastic disorders, 73-79 

noninvasive examination of, 19-23 

upper limb occlusive, 11-12 
Vascular examination, noninvasive, 19-23 
Vascular injuries, 125-132 

abdominal, 128-130 

combat-related, 125 

diagnosis of, 126 

of the extremities, 126, 130-131 

iatrogenic, 126, 131 

imaging of, 37 

mechanism of injury in, 126 

in the neck, 126, 127-128 

thoracic, 128 




> 



266 



INDEX 



Vascular surgery. See also specific surgical 
procedures 
complications of, 133-140 
Vasculitis, 79-86 

cryoglobulinemic, 85-86 
large-vessel, 80-82 
medium-vessel, 82-83 
small-vessel, 83-85 
Vasoconstriction, Raynaud's phenomenon-related, 

73 
Vasodilation, erythromelalgia-related, 79 
Vasodilators, 241 

Vasospasm, Raynaud's phenomenon-related, 73 
Vasospastic disorders, 73-79 
Vein grafts, historical background of, 125 
Velocity analyses, 22 
Venoarteriolar reflex, lymphedema-related loss of, 

116 
Venous disease 

chronic, clinical evaluation of, 17 
deep, symptoms of, 12 
superficial, symptoms of, 12 
symptoms of, 12 
Venous function, normal, 106-107 
Venous insufficiency 
chronic, 17, 105-113 

classification of, 105, 106 
clinical assessment of, 107-109 
epidemiology of, 105-106 
nonsurgical management of, 109-111 
pathophysiology of, 107 
relationship with lymphedema, 118-119 
surgical management of, 111-113 
symptoms of, 108 
Venous systems, deep and superficial, 106-107 
Vertebral arteries 

effect of subclavian stenosis on, 11 
occlusive disease of, 188-190 



traumatic injuries to, 128 
Vertebrobasilar symptoms, of stroke, 161, 163, 

188-189 
Very-low density lipoprotein (VLDL), 2, 3, 4 
Veterans Administration, Surgical Quality 

Improvement Program (NSQIP), 149 
Vietnam Vascular Registry, 125 
Vietnam War, vascular surgery during, 125 
Visceral arteries, aneurysms of, 214-217 
Visceral ischemic syndromes, 231-236 

acute mesenteric ischemia, 231-234 

chronic visceral ischemia, 234-235 

mesenteric venous thrombosis, 235-236 
Visuospatial neglect, 163 
von Willebrand disease, 43-44 
von Willebrand factor, 41 

W 

Walking Impairment Questionnaire (WIQ), 152 

WALLSTENT study, 180 

Wegener, Friedrich, 83 

Wegener's granulomatosis, 83-84 

Weight gain, smoking cessation-related, 58 

Weight-loss programs, 57, 58 

World Wars I and II, vascular surgery development 

during, 125 
Wound healing, evaluation of, 21-22 



Xanthelasma, 12 

X-ray exposure, safety precautions for, 30-31 

X-rays 

abdominal, of abdominal aortic aneurysms, 
194 

chest, of traumatic injuries, 37, 128 



Yellow-nail syndrome, 117