Skip to main content

Full text of "Surgery-Book_1aim.net"

See other formats


Current Clinical Strategies 



Surgery 



Sixth Edition 

The University of California, Irvine, Manual of 
Surgery 



Samuel Eric Wilson, MD 

Professor and Chairman 
Department of Surgery 
College of Medicine 
University of California, Irvine 

Bruce M, Achauer, MD 
John A. Butler, MD 
David A. Chamberlin, MD 
Paul D. Chan, MD 
Marianne Cinat, MD 
Roger Crumley, MD 
Alex Di Stante, MD 
C. Garo Gholdoian, MD 
Ian L. Gordon, MD, PhD 
Joshua Helman, MD 
James G. Jakowatz, MD 
Fernando Katie, MD 
Michael E. Lekawa, MD 
I. James Sarfeh, MD 
Michelle Schultz, MD 
Harry Skinner, MD 
Charles Theuer, MD 
Russell A. Williams, MD 



Current Clinical Strategies Publishing 

www. ccspublishing. com/ccs 



Digital Book and Updates 

Purchasers of this book may download the digital book 
and updates for Palm, Pocket PC, Windows and 
Macintosh. The digital books can be downloaded at the 
Current Clinical Strategies Publishing Internet site: 

www.ccspublishing.com/ccs/surg.htm 



Copyright © 2006 Current Clinical Strategies Publishing . 
All rights reserved. This book, or any parts thereof, may 
not be reproduced, photocopied, or stored in an 
information retrieval network without the permission of 
the publisher. The reader is advised to consult the drug 
package insert and other references before using any 
therapeutic agent. The publisher disclaims any liability, 
loss, injury, or damage incurred as a consequence, 
directly or indirectly, of the use and application of any of 
the contents of this text. 

Current Clinical Strategies Publishing 

27071 Cabot Road 

Laguna Hills, California 92653 

Phone: 800-331-8227 or 949-348-8404 

Fax: 800-965-9420 or 949-348-8405 

Internet: www.ccspublishing.com/ccs 

E-mail: info@ccspublishing.com 



Printed in USA ISBN 1929622-57-0 



Surgical Documentation 

S. E. Wilson, MD 

Surgical History and Physical 
Examination 

Identifying Data: Patient's name, age, race, sex; 
referring physician. 

Chief Compliant: Reason given by patient for seeking 
surgical care and the duration of the symptom. 
History of Present Illness (HPI): Describe the course of 
the patient's illness, including when it began, character of 
the symptoms; pain onset (gradual or rapid), precise 
character of pain (constant, intermittent, cramping, 
stabbing, radiating); other factors associated with pain 
(defecation, urination, eating, strenuous activities); loca- 
tion where the symptoms began; aggravating or relieving 
factors. Vomiting (color, character, blood, coffee-ground 
emesis, frequency, associated pain). Change in bowel 
habits; rectal bleeding, character of blood (clots, bright or 
dark red), trauma; recent weight loss or anorexia; other 
related diseases; past diagnostic testing. 
Past Medical History (PMH): Previous operations and 
indications; dates and types of procedures; serious 
injuries, hospitalizations; diabetes, hypertension, peptic 
ulcer disease, asthma, heart disease; hernia, gallstones. 
Medications: Aspirin, anticoagulants, hypertensive and 
cardiac medications, diuretics. 
Allergies: Penicillin, codeine, iodine. 
Family History: Medical problems in relatives. Family 
history of colon cancer, cardiovascular disease. 
Social History: Alcohol, smoking, drug usage, 
occupation, daily activity. 
Review of Systems (ROS): 

General: Weight gain or loss; loss of appetite, fever, 
fatigue, night sweats. Activity level. 
HEENT: Headaches, seizures, sore throat, masses, 
dentures. 

Respiratory: Cough, sputum, hemoptysis, dyspnea 
on exertion, ability to walk up flight of stairs. 
Cardiovascular: Chest pain, orthopnea, claudication, 
extremity edema. 

Gastrointestinal: Dysphagia, vomiting, abdominal 
pain, hematemesis, melena (black tarry stools), 
hematochezia (bright red blood per rectum), constipa- 
tion, change in bowel habits; hernia, hemorrhoids, 
gallstones. 

Genitourinary: Dysuria, hesitancy, hematuria, 
discharge; impotence, prostate problems, urinary 
frequency. 

Gynecological: Last menstrual period, gravida, para, 
abortions, length of regular cycle and periods, birth 
control. 

Skin: Easy bruising, bleeding tendencies. 
Neurological: Stroke, transient ischemic attacks, 
weakness. 

Surgical Physical Examination 

General appearance: Note whether the patient looks 

"ill," well, or malnourished. 

Vital Signs: Temperature, respirations, heart rate, blood 

pressure, weight. 

Eyes: Pupils equally round and react to light (PERRL); 

extraocular movements intact (EOMI). 
Neck: Jugular venous distention (JVD), thyromegaly, 

masses, bruits; lymphadenopathy; trachea midline. 
Chest: Equal expansion, dullness to percussion; rales, 

rhonchi, breath sounds. 
Heart: Regular rate and rhythm (RRR), first and second 

heart sounds; murmurs (grade 1-6), pulses (graded 0- 

2+). 
Breast: Skin retractions, erythema, tenderness, masses 

(mobile, fixed), nipple discharge, axillary or 

supraclavicular node enlargement. 
Abdomen: Contour (flat, scaphoid, obese, distended), 

scars, bowel sounds, bruits, tenderness, masses, liver 

span; splenomegaly, guarding, rebound, percussion 

note (dull, tympanic), pulsatile masses, costovertebral 

angle tenderness (CVAT), abdominal hernias. 
Genitourinary: Inguinal hernias, testicles, varicoceles; 

urethral discharge, varicocele. 
Extremities: Skin condition, edema (grade 1-4+); 

cyanosis, clubbing, pulses (radial, ulnar, femoral, 

popliteal, posteriortibial, dorsalis pedis; simultaneous 

palpation of radial and femoral pulses). Grading of 

pulses: = absent; 1+ weak; 2+ normal; 3+ very 

strong (arterial dilation). 
Rectal Exam: Masses, tenderness, hemorrhoids, 

prostate masses; bimanual palpation, guaiac test for 

occult blood. 
Neurological: Mental status, cranial nerves, gait, 



strength (graded 0-5); tendon reflexes, sensory 

testing. 
Laboratory Evaluation: Electrolytes (sodium, 
potassium, bicarbonate, chloride, BUN, creatinine), 
glucose, liver function tests, INR/PTT, CBC with 
differential; X-rays, ECG (if older than 35 yrs or 
cardiovascular disease), urine analysis. 
Assessment (Impression): Assign a number to each 
problem and discuss each problem. Begin with most 
important problem and rank in order. 
Plan: Discuss surgical plansfor each numbered problem, 
including preoperative testing, laboratory studies, 
medications, antibiotics, endoscopy. 



Preoperative Preparation of the 
Surgical Patient 

1 . Reviewthe patient's history and physical examination, 
and write a preoperative note assessing the patient's 
overall condition and operative risk. 

2. Preoperative laboratory evaluation: Electrolytes, 
BUN, creatinine, INR/PTT, CBC, platelet count, UA, 
ABG, pulmonary function test. Chest x-ray (>35 yrs 
old), EKG (if older then 35 yrs old or if cardiovascular 
disease). Type and cross for an appropriate number of 
units of blood. No screening laboratory tests are 
required in the healthy patient. 

3. Skin preparation: Patient to shower and scrub the 
operative site with germicidal soap (Hibiclens) on the 
night before surgery. On the day of surgery, hair 
should be removed with an electric clipper or shaved 
just prior to operation. 

4. Prophylactic antibiotics or endocarditis 
prophylaxis if indicated. 

5. Preoperative incentive spirometry on the evening 
prior to surgery may be indicated for patients with 
pulmonary disease. 

6. Thromboembolic prophylaxis should be provided for 
selected, high-risk patients. 

7. Diet: NPO after midnight. 

8. IV and monitoring lines: At least one 1 8-gauge IV for 
initiation of anesthesia. Arterial catheter and 
pulmonary artery catheters (Swan-Ganz) if indicated. 
Patient to void on call to operating room. 

9. Medications. Preoperative sedation as ordered by 
anesthesiologist. Maintenance medicationsto be given 
the morning of surgery with a sip of water. Diabetics 
should receive one half of their usual AM insulin dose, 
and an insulin drip should be initiated with hourly 
glucose monitoring. 

10. Bowel preparation 

Bowel preparation is required for upper or lower Gl tract 

procedures. 

Antibiotic Preparation for Colonic Surgery 

Mechanical Prep: Day 1 : Clear liquid diet, laxative (milk 
of magnesia 30 cc or magnesium citrate 250 cc), tap 
water or Fleet enemas until clear. Day 2: Clear liquid 
diet, NPO, laxative. Day 3: Operation. 

Whole Gut Lavage: Polyethylene glycol electrolyte 

solution (GoLytely). 

Day 1 : 2 liters PO or per nasogastric tube over 5 
hours. Clear liquid diet. Day 2: Operation. 

Oral Antibiotic Prep: One day prior to surgery, after 
mechanical or whole gut lavage, give neomycin 1 
gm and erythromycin 250 mg at 1 p.m., 2 p.m., 11 
p.m. 

11. Preoperative IV antibiotics: Initiate preoperatively 
and give one dose during operation and one dose of 
antibiotic postoperatively. Cefotetan (Cefotan), 1 gm 
IV q12h, for bowel flora, or cefazolin (Ancef), 1 gm 
IVPB q8h x 3 doses, for clean procedures. 

12. Anticoagulants: Discontinue Coumadin 5 days 
preop and check PT; stop IV heparin 6 hours prior to 
surgery. 



Admitting and Preoperative Orders 

Admit to: Ward, ICU, or preoperative room. 

Diagnosis: Intended operation and indication. 

Condition: Stable 

Vital Signs: Frequency of vital signs; input and output 
recording; neurological or vascular checks. Notify 
physician if blood pressure <90/60, >160/1 10; pulse 
>1 10; pulse <60; temperature >101 .5; urine output 
<35 cc/h for >2 hours; respiratory rate >30. 

Activity: Bed rest or ambulation; bathroom privileges. 

Allergies: No known allergies 

Diet: NPO 

IV Orders: D5 1/2 NS at 100 cc/hour. 

Oxygen: 6 L/min by nasal canula. 

Drains: Foley catheter to closed drainage. Nasogastric 
tube at low intermittent suction. Other drains, tubes, 



dressing changes. Orders for irrigation of tubes. 

Medications: Antibiotics to be initiated immediately 
preoperatively; additional dose during operation and 
1 dose of antibiotic postoperatively. Cefotetan 
(Cefotan), 1 gm IV q12h, for bowel flora, or cefazolin 
(Ancef) 1 gm IVPB q8h x 3 doses;) for clean 
procedures. 

Labs and Special X-Rays: Electrolytes, BUN, creatinine, 
INR/PTT, CBC, platelet count, UA, ABG, pulmonary 
function tests. Chest x-ray (if >35 yrs old), EKG (if 
older then 35 yrs old or if cardiovascular disease). 
Type and cross for an appropriate number of units of 
biood. 



Preoperative Note 

Preoperative Diagnosis: 

Procedure Planned: 

Type of Anesthesia Planned: 

Laboratory Data: Electrolytes, BUN, creatinine, CBC, 

INR/PTT, UA, EKG, chest x-ray; type and screen for 

blood or cross match if indicated; liver function tests, 

ABG. 

Risk Factors: Cardiovascular, pulmonary, hepatic, renal, 

coagulopathic, nutritional risk factors. 

American Surgical Association (ASA) grading of 

surgical risk: 1= normal; 2= mild systemic disease; 3= 

severe systemic disease; 4= disease with major threat to 

life; 5= not expected to survive. 

Consent: Document explanation to patient of risks and 

benefits of the procedure and alternative treatments. 

Document patient's or guardian's informed consent and 

understanding of the procedure. Obtain signed consent 

form. 

Allergies: 

Major Medical Problems: 

Medications: 

Special Requirements: Signed blood transfusion 

consent form; documentation that breast procedure 

patients have been given an information brochure. 



Brief Operative Note 

This note should be written in chart immediately after the 

surgical procedure. 

Date of the Procedure: 

Preoperative Diagnosis: 

Postoperative Diagnosis: 

Procedure: 

Operative Findings: 

Names of Surgeon and Assistants: 

Anesthesia: General endotracheal, spinal, epidural, 

regional or local. 

Estimated Blood Loss (EBL): 

Fluids and Blood Products Administered During 

Procedure: 

Urine output: 

Specimens: Pathology specimens, cultures, blood 

samples. 

Intraoperative X-rays: 

Drains: 

Condition of Patient: Stable 



Operative Report 

This full report should be dictated at the conclusion of the 

surgical procedure. 

Identifying Data: Name of patient, medical record 

number; name of dictating physician, date of dictation. 

Attending Surgeon and Service: 

Date of Procedure: 

Preoperative Diagnosis: 

Postoperative Diagnosis: 

Procedure Performed: 

Names of Surgeon and Assistants: 

Type of Anesthesia Used: 

Estimated Blood Loss (EBL): 

Fluid and Blood Products Administered During 

Operation: 

Specimens: Pathology, cultures, blood samples. 

Drains and Tubes Placed: 

Complications: 

Consultations Intraoperatively: 

Indications for Surgery: Brief history of patient and 

indications for surgery. 

Findings: Describe gross findings and frozen section 

results relayed to operating room. 

Description of Operation: Position of patient; skin prep 

and draping; location and types of incisions; details of 

procedure from beginning to end, including description of 

surgical findings, both normal and abnormal. 



Intraoperative studies or x-rays; hemostatic and closure 
techniques; dressings applied. Needle and sponge 
counts as reported by operative nurse. Patient's condition 
and disposition. Send copies of report to surgeons and 
referring physicians. 



Postoperative Check 

A postoperative check should be completed on the 
evening after surgery. This check is similar to a daily 
progress note. 



Example Postoperative Check 



Date/time: 

Postoperative Check 

Subjective: Note any patient complaints, and note 

the adequacy of pain relief. 

Objective: 

General appearance: 

Vitals: Maximum temperature in the last 24 

hours (T max ), current temperature, pulse, 

respiratory rate, blood pressure. 

Urine Output: If urine output is less than 30 cc 

per hour, more fluids should be infused if the 

patient is hypovolemic. 

Physical Exam: 

Chest and lungs: 

Abdomen: 

Wound Examination: The wound should be 

examined for excessive drainage or bleeding, 

skin necrosis, condition of drains. 

Drainage Volume: Note the volume and 

characteristics of drainage from Jackson-Pratt 

drain or other drains. 

Labs: Post-operative hematocrit value and 
other labs. 

Assessment and Plan: Assess the patient's 
overall condition and status of wound. Comment on 
abnormal labs, and discuss treatment and 
discharge plans. 



Postoperative Orders 

1 . Transfer: From recovery room to surgical ward when 
stable. 

2. Vital Signs: q4h, l&O q4h x 24h. 

3. Activity: Bed rest; ambulate in 6-8 hours if 
appropriate. Incentive spirometer q1h while awake. 

4. Diet: NPO x 8h, then sips of water. Advance from 
clear liquids to regular diet as tolerated. 

5. IV Fluids: IV D5 LR or D5 1/2 NS at 125 cc/h (KCL, 
20 mEq/L if indicated), Foley to gravity. 

6. Medications: 

Cefazolin (Ancef) 1 gm IVPB q8h x 3 doses; if 
indicated for prophylaxis in clean cases OR 

Cefotetan (Cefotan) 1 gm IVq12h x2 doses for clean 
contaminated cases. 

Meperidine (Demerol) 50 mg IV/IM q3-4h prn pain 

Hydroxyzine (Vistaril) 25-50 mg IV/IM q3-4h prn 

nausea OR 

Prochlorperazine (Compazine) 10 mg IV/IM q4-6h prn 
nausea or suppository q 4h prn. 

7. Laboratory Evaluation: CBC, SMA7, chest x-ray in 
AM if indicated. 



Postoperative Surgical Management 

I. Postoperative day number 1 

A. Assess the patient's level of pain, lungs, cardiac 
status, flatulence, and bowel movement. Examine 
for distension, tenderness, bowel sounds; wound 
drainage, bleeding from incision. 

B. Discontinue IV infusion when taking adequate PO 
fluids. Discontinue Foley catheter, and use in-and- 
out catheterization for urinary retention. 

C. Ambulate as tolerated; incentive spirometer, 
hematocrit and hemoglobin. 

D. Acetaminophen/codeine (Tylenol #3) 1-2 POq4-6h 
prn pain. 

E. Colace 100 mg PO bid. 

F. Consider prophylaxis for deep vein thrombosis. 

II. Postoperative day number 2 

A. If passing gas or if bowel movement, advance to 
regular diet unless bowel resection. 

B. Laxatives: Dulcolax suppository prn or Fleet enema 
prn or milk of magnesia, 30 cc PO prn constipation. 

III. Postoperative day number 3-7 

A. Check pathology report. 

B. Remove staples and place steri-strips. 



C. Consider discharge home on appropriate medica- 
tions; follow up in 1 -2 weeks for removal of sutures. 

D. Write discharge orders (including prescriptions) in 

AM; arrange 
for home health care if indicated. Dictate discharge 
summary and send copy to surgeon and referring 
physician. 



Surgical Progress Note 

Surgical progress notes are written in "SOAP" format. 



Surgical Progress Note 



Date/Time: 

Post-operative Day Number: 

Problem List: Antibiotic day number and 

hyperalimentation day number if applicable. List 

each surgical problem separately (eg, status-post 

appendectomy, hypokalemia). 

Subjective: Describe how the patient feels in the 

patient's own words, and give observations about 

the patient. Indicate any new patient complaints, 

note the adequacy of pain relief, and passing of 

flatus or bowel movements. Type of food the 

patient is tolerating (eg, nothing, clear liquids, 

regular diet). 

Objective: 

Vital Signs: Maximum temperature (T max ) over 

the past 24 hours. Current temperature, vital 

signs. 

Intake and Output: Volume of oral and 

intravenous fluids, volume of urine, stools, 

drains, and nasogastric output. 

Physical Exam: 

General appearance: Alert, ambulating. 

Heart: Regular rate and rhythm, no murmurs. 

Chest: Clear to auscultation. 

Abdomen: Bowel sounds present, soft, 

nontender. 

Wound Condition: Comment on the wound 

condition (eg, clean and dry, good 

granulation, serosanguinous drainage). 

Condition of dressings, purulent drainage, 

granulation tissue, erythema; condition of 

sutures, dehiscence. Amount and color of 

drainage 

Lab results: White count, hematocrit, and 

electrolytes, chest x-ray 
Assessment and Plan: Evaluate each numbered 
problem separately. Note the patient's general 
condition (eg, improving), pertinent developments, 
and plans (eg, advance diet to regular, chest x- 
ray). For each numbered problem, discuss any ad- 
ditional orders and plans for discharge or transfer. 



Procedure Note 

A procedure note should be written in the chart when a 
procedure is performed. Procedure notes are brief 
operative notes. 



Procedure Note 



Date and time: 

Procedure: 

Indications: 

Patient Consent: Document that the indications, 

risks and alternatives to the procedure were 

explained to the patient. Note that the patient was 

given the opportunity to ask questions and that the 

patient consented to the procedure in writing. 

Lab tests: Electrolytes, INR, CBC 

Anesthesia: Local with 2% lidocaine 

Description of Procedure: Briefly describe the 

procedure, including sterile prep, anesthesia 

method, patient position, devices used, anatomic 

location of procedure, and outcome. 

Complications and Estimated Blood Loss 

(EBL): 

Disposition: Describe how the patient tolerated 

the procedure. 
Specimens: Describe any specimens obtained 
and laboratory tests which were ordered. 



Discharge Note 



The discharge note should be written in the patient's 
chart prior to discharge. 



Discharge Note 



Date/time: 

Diagnoses: 

Treatment: Briefly describe treatment provided 

during hospitalization, including surgical 

procedures and antibiotic therapy. 

Studies Performed: Electrocardiograms, CT 

scans. 

Discharge Medications: 

Follow-up Arrangements: 



Discharge Summary 



Patient's Name: 
Chart Number: 
Date of Admission: 
Date of Discharge: 
Admitting Diagnosis: 
Discharge Diagnosis: 
Name of Attending or Ward Service: 
Surgical Procedures, Diagnostic Tests, Invasive 
Procedures: 

Brief History and Pertinent Physical Examination and 
Laboratory Data: Describe the course of the patient's 
disease up to the time the patient came to the hospital, 
and describe the physical exam and pertinent laboratory 
data on admission. 

Hospital Course: Briefly describe the course of the 
patient's illness while in the hospital, including evaluation, 
operation, outcome of the operation, and medications 
given while in the hospital. 

Discharged Condition: Describe improvement or 
deterioration of the patient's condition. 
Disposition: Describe the situation to which the patient 
will be discharged (home, nursing home) and the person 
who will provide care. 

Discharged Medications: List medications and 
instructions and write prescriptions. 
Discharged Instructions and Follow-up Care: Date of 
return for follow-up care at clinic; diet, exercise 
instructions. 

Problem List: List all active and past problems. 
Copies: Send copies to attending physician, clinic, con- 
sultants and referring physician. 



Prescription Writing 



Patient's name: 

Date: 

Drug name, dosage form, dose, route, frequency 

(include concentration for oral liquids or mg strength for 

oral solids): Amoxicillin 125mg/5mL 5 mL PO tid 

Quantity to dispense: mL for oral liquids, # of oral solids 

Refills: If appropriate 

Signature 



Clinical Care of the 
Surgical Patient 



James G. Jakowatz, MD 
Marianne Cinat, MD 



Radiographic Evaluation of Common 
Interventions 

I. Central intravenous lines: 

A.Central venous catheters should be located well 
above the right atrium, and not in a neck vein. 
Pneumothorax should be excluded by checking that 
the lung markings extend completely to the rib cages 
on both sides. An upright, expiratory x-ray may be 
helpful. Hemothorax will appear as opacification or 
a fluid meniscus on chest x-ray. Mediastinal 
widening may indicate great vessel injury. 

B. Pulmonary artery catheters should be located 
centrally and posteriorly and not more than 3-5 cm 
from midline within the mediastinum. 

II. Pulmonary tubes 

A. Endotracheal tubes: Verify that the tube is located 
3 cm below the vocal cords and 2-4 cm above the 
carina. The tip of tube should be at the level of aortic 
arch. 

B. Tracheostomy: Verify by chest x-ray that the tube 
is located half way between the stoma and the 
carina; the tube should be parallel to the long axis of 
the trachea. The tube should be approximately 2/3 
of width of the trachea, and the cuff should not cause 
bulging of the trachea walls. Check for 
subcutaneous air in the neck tissue and for 
mediastinal widening secondary to air leakage. 

C. Chest tubes: A chest tube for pneumothorax 
drainage should be located anteriorly at the mid- 
clavicular line at the level of the third intercostal 
space or in the anterior axillary line directed toward 
the apex at the 4-5th intercostal space. Pleural 
effusions should be drained by locating the tube 
inferior-posteriorly at or about the level of the eighth 
intercostal space and directed posteriorly. 

D. Mechanical ventilation: A chest x-ray should be 
obtained to rule out pneumothorax, subcutaneous 
emphysema, pneumomediastinum or subpleural air 
cysts. Lung infiltrates may diminish or disappear 
because of increased aeration of the affected lung 
lobe. 

III. Gastrointestinal tubes 

A. Nasogastric tubes: Verify that the tube is in the 
stomach and not coiled in the esophagus or trachea. 
The tip of the tube should not be near 
gastroesophageal junction. The standard size 
nasogastric tube is 1 4-1 6 French. Nasogastric tubes 
are used to decompress the stomach. 

B. Feeding tubes are smaller in size (8-12 Fr) than 
nasogastric tubes. They are flexible and are 
frequently used for enteral nutrition . They are passed 
nasally through the stomach and into the duodenum 
or jejunum. The tip is radiopaque, and it should be 
located in the distal stomach. The tube may extend 
through the pylorus into the duodenum. 



Blood Component Therapy 

I. Crystalloids solutions: Sodium is the principle 
component of crystalloid solutions, which is the most 
abundant solute in the extracellular fluid. 

A. Hypotonic solutions include 0.45% normal saline 
and 0.25% normal saline. Hypotonic solutions are 
used as maintenance fluids in adults (0.45% NS) 
and infants (0.25% NS). 

B. Isotonic solutions include normal saline (0.9% 
NaCI; 154 mEq Na and 154 mEq CI) and lactated 
Ringers (1 30meq Na, 1 09 mEq CI, 4 mEq K, 3 mEq 
Ca, lactate as a buffer). Isotonic solutions are used 
for acute resuscitation and volume replacement. 
Approximately 3 cc of crystalloid should be given to 
replace each 1 cc of blood loss. 

C. Hypertonic saline (7.5% NaCI; 1283 mEq Na, 
1283 mEq CI) is used to treat symptomatic 
hyponatremia. Replacement must be done slowly 
to prevent central pontine myelinolysis. 

II. Colloid solution therapy is indicated for volume 
expansion. 

A. Albumin (5% or 25%) is useful for hypovolemia or 
to induce diuresis with furosemide in hypervolemic, 
hypoproteinemic patients. Salt poor albumin is 
used in cirrhosis. 

B. Purified protein fraction (Plasmanate) consists of 
83% albumin and 17% globulin. It is indicated for 
volume expansion as an alternative to albumin. 

C. Hetastarch (Hespan) consists of synthetic colloid 
(6% hetastarch in saline). Hespan is useful for 
volume expansion and raising osmotic pressure. 
Maximum dose is 1500 cc per 24 hours. 
Hetastarch may prolong the partial thromboplastin 
time. 

III. Management of acute blood loss - red blood cell 
transfusions 

A. Hemorrhage should be controlled, and crystalloids 
should be infused until packed red blood cells are 
available to replace losses. In trauma, bleeding 
may require surgical control. If crystalloids fail to 
produce hemodynamic stability after more than 2 
liters have been administered, packed red blood 
cells should be given. 

B. If volume replacement and hemostasis stabilize the 
patient's hemodynamic status, formal type and 
cross match of blood should be completed. In 
exigent bleeding, O negative, lowtiter blood or type 
specific (ABO matched) Rh compatible blood 
should be administered. 

IV. Guidelines for blood transfusion in anemia. 
Consider blood transfusion when hemoglobin is less 
than 8.0 gm/dL and hematocrit is less than 24%. If the 
patient has symptoms of anemia, such as chest pain, 
dyspnea, mental status changes, transfusion should 
be provided. 

V. Blood component products 

A. Packed red blood cells (PRBCs). Each unit 
provides 400 cc of volume, and each unit should 
raise hemoglobin by 1 gm/dL and hematocrit by 
3%. 

B. Platelets are indicated for bleeding due to 
thrombocytopenia or thrombopathy. Each unit 
should raise the platelet count by 5,000-10,000 
cells/mul. Platelets are usually transfused 8-10 
units at a time. Dilutional thrombocytopenia occurs 
after massive blood transfusions. Therefore, 
platelet transfusion should be considered after 8-10 
units of blood replacement. ABO typing is not 
necessary before platelets are given. 

C. Fresh frozen plasma (FFP) is used for bleeding 
secondary to liver disease, dilutional coagulopathy 
(from multiple blood transfusions), or coagulation 
factor deficiencies. ABO typing is required before 
administration of FFP, but cross matching is not 
required. Improvement of INR/PTT usually requires 
2-3 units. One unit of FFP should be administered 
for every 4-6 units of PRBCs. FFP contains all 
clotting factors except factors V and VII. 

D. Cryoprecipitate contains factor VIII, and 
fibrinogen. It is given 8-10 units at a time. 
Cryoprecipitate may be necessary for massive 
transfusions. 

E. Autologous blood. The patient donates blood 
within 35 days of surgery; frozen blood can be 
stored for up to 2 years. Autologous blood is useful 
in elective orthopedic, cardiac, and peripheral 
vascular procedures. 



Fluids and Electrolytes 

I. Maintenance fluid guidelines 

A. Maintenance fluid requirements consist of 4 cc/kg 



for the first 10 kg of body weight, 2 cc/kg for the 
second 10 kg, and 1 cc/kg for each additional kg. 
B. A 70 mg patient has a maintenance fluid 
requirement of approximately 125 mL/hr. 
Maintenance fluids used are D5 1/2 NS with 20 
mEq KCL/liter and D5 1/4 NS with 20 mEq KCI/liter 
in children. 

II. Pediatric patients 

A. Use D5 1/4 NS with 20 mEq KCL/liter. 

B. 24 hour water requirement, kilogram method: 
For the first 10 kg body weight: 100 mL/kg/day 
PLUS for the second 10 kg body weight: 50 
mL/kg/day PLUS for weight above 20 kg: 20 
mL/kg/day. Divide by 24 hours to determine hourly 
rate. 

III. Specific replacement fluids of specific losses 

A. Gastric fluid (nasogastric tube, emesis). D5 1/2 
NS with 20 mEq/liter KCL; replace equal volume of 
lost fluid q6h. 

B. Diarrhea. D5LR with 15 mEq/liter KCL. Provide 1 
liter replacement for each 1 kg or 2.2 lb of lost body 
weight; bicarbonate 45 mEq (1/2 amp) per liter may 
be added. 

C. Bile. D5LR with 25 mEq/liter (1/2 amp) of 
bicarbonate. 

D. Pancreatic. D5LR with 50 mEq/liter (1 amp) 
bicarbonate. 



Evaluation of Postoperative Fever 

I. Clinical evaluation 

A. History. Fever >1 00.4-1 01 F. Determine the 
number of days since operation. 

B. Differential diagnosis. Pneumonia, urinary tract 
infection, thrombophlebitis, wound infection, drug 
reaction. Atelectasis is the most common cause of 
fever less than 48 hrs after operation. 

C. Dysuria, abdominal pain, cough, sputum, 
headache, stiff neck, joint or back pain may be 
present. 

D. IV catheter infection (central or peripheral) is an 
important source of postoperative sepsis. 

E. Fever pattern. Check previous day for fever 
patterns; spiking fevers indicate abscesses. 
Continuous fevers or high fevers indicate vascular 
involvement, such as infected prosthetic grafts or 
septic phlebitis from central IV lines. 

F. Chills or rigors indicate bacteremia. These 
symptoms are usually not associated with 
atelectasis or drug fevers. 

G. Fevers prior to the operation, alcohol use, allergies, 
and recent WBC count and differential counts 
should be assessed. 

II. Physical Exam 

A. General. Temperature, fever curve, tachycardia, 
hypotension. Examine all vascular access sites 
carefully. 

B. HEENT. Pharyngeal erythema, neck rigidity. 

C. Chest. Rhonchi, crackles, dullness to percussion 
(pneumonia), murmurs (endocarditis). 

D. Abdomen. Masses, liver tenderness, Murphy's 
sign (right upper quadrant tenderness with 
inspiration, cholecystitis); ascites. Costovertebral 
angle or suprapubic tenderness. Examine wound 
for purulence, induration, or tenderness. 

E. Extremities. Infected decubitus ulcers or wounds; 
IV catheter tenderness (phlebitis); calf tenderness, 
joint tenderness (septic arthritis). Cellulitis, 
abscesses, perirectal abscess. 

F. Genitourinary. Prostate tenderness; rectal 
fluctuance. Cervical discharge, cervical motion 
tenderness; adnexal tenderness. 

III. Laboratory evaluation. CBC, blood C&S X 2, UA, 
urine C&S; blood, urine, sputum, wound cultures, 
chest x-ray. 

IV. Differential diagnosis 

A. Wound infection, abscesses, intra-abdominal 
abscess, atelectasis, drug fever, pulmonary emboli, 
pancreatitis, alcohol withdrawal, deep vein 
thrombosis, tuberculosis, cystitis, pyelonephritis, 
osteomyelitis; IV catheter phlebitis, sinusitis, otitis 
media, upper respiratory infection, pelvic infection, 
cellulitis; hepatitis, infected decubitus ulcer, 
peritonitis, endocarditis, diverticulitis, cholangitis, 
carcinomas. 

B. Medications associated with fever: H2 blockers, 
penicillins, phenytoin, sulfonamides. 

V. Antibiotics should be initiated if there is any 
possibility of infection. 



Sepsis 



About 400,000 cases of sepsis, 200,000 cases of septic 
shock, and 100,000 deaths from both occur each year. 

I. Pathophysiology 

A. Sepsis is defined as the systemic response to 
infection. In the absence of infection, it is called 
systemic inflammatory response syndrome and is 
characterized by at least two of the following: 
temperature greater than 38"C or less than 36"C; 
heart rate greater than 90 beats per minute; 
respiratory rate more than 20/minute or PaC0 2 less 
than 32 mm Hg; and an alteration in white blood 
cell count (>12,000/mm 3 or <4,000/mm 3 ). 

B. Septic shock is defined as sepsis-induced 
hypotension that persists despite fluid resuscitation 
and is associated with tissue hypoperfusion. 

C. The initial cardiovascular response to sepsis 
includes decreased systemic vascular resistance 
and depressed ventricular function. Low systemic 
vascular resistance occurs. If this initial 
cardiovascular response is uncompensated, 
generalized tissue hypoperfusion results. 
Aggressive fluid resuscitation may improve cardiac 
output and systemic blood pressure, resulting in the 
typical hemodynamic pattern of septic shock (ie, 
high cardiac index and low systemic vascular 
resistance). 

D. Although gram-negative bacteremia is commonly 
found in patients with sepsis, gram-positive 
infection may affect 30-40% of patients. Fungal, 
viral and parasitic infections are usually 
encountered in immunocompromised patients. 



Defining sepsis and related disorders 


Term 


Definition 


Systemic 

inflammatory 
response 
syndrome 
(SIRS) 


The systemic inflammatory response to 
a severe clinical insult manifested by >2 
of the following conditions: Temperature 
>38°C or <36°C, heart rate >90 
beats/min, respiratory rate >20 
breaths/min or PaC0 2 <32 mm Hg, 
white blood ceil count >12,000 
cells/mm 3 , <4000 cells/mm 3 , or >10% 
band cells 


Sepsis 


The presence of SIRS caused by an 
infectious process; sepsis is considered 
severe if hypotension or systemic 
manifestations of hypoperfusion (lactic 
acidosis, oliguria, change in mental 
status) is present. 


Septic shock 


Sepsis-induced hypotension despite 
adequate fluid resuscitation, along with 
the presence of perfusion abnormalities 
that may induce lactic acidosis, oliguria, 
or an alteration in mental status. 


Multiple organ 
dysfunction 
syndrome 
(MODS) 


The presence of altered organ function 
in an acutely ill patient such that 
homeostasis cannot be maintained 
without intervention 



E. Sources of bacteremia leading to sepsis include 
the urinary, respiratory and Gl tracts, and skin and 
soft tissues (including catheter sites). The source 
of bacteremia is unknown in 30% of patients. 

F. Escherichia coli is the most frequently 
encountered gram-negative organism, followed by 
Klebsiella, Enterobacter, Serratia, Pseudomonas, 
Proteus, Providencia, and Bacteroides species. 
Up to 16% of sepsis cases are polymicrobic. 

G. Gram-positive organisms, including 
Staphylococcus aureus and Staphylococcus 
epidermidis, are associated with catheter or line- 
related infections. 

Diagnosis 

A. A patient who is hypotensive and in shock should 
be evaluated to identify the site of infection, and 
monitorfor end-organ dysfunction. History should 
be obtained and a physical examination 
performed. 

B. The early phases of septic shock may produce 
evidence of volume depletion, such as dry 
mucous membranes, and cool, clammy skin. After 
resuscitation with fluids, however, the clinical 
picture resembles hyperdynamic shock, including 
tachycardia, bounding pulses with a widened 
pulse pressure, a hyperdynamic precordium on 
palpation, and warm extremities. 

C. Signs of infection include fever, localized 
erythema or tenderness, consolidation on chest 
examination, abdominal tenderness, and 
meningismus. Signs of end-organ hypoperfusion 
include tachypnea, oliguria, cyanosis, mottling of 



the skin, digital ischemia, abdominal tenderness, 
and altered mental status. 
, Laboratory studies should include of arterial 
blood gases, lactic acid level, electrolytes, renal 
function, liver enzyme levels, and chest 
radiograph. Cultures of blood, urine, and sputum 
should be obtained before antibiotics are 
administered. Cultures of pleural, peritoneal, and 
cerebrospinal fluid may be appropriate. If 
thrombocytopenia or bleeding is present, tests for 
disseminated intravascular coagulation should 
include fibrinogen, d-dimer assay, platelet count, 
peripheral smear for schistocytes, prothrombin 
time, and partial thromboplastin time. 



Manifestations of Sepsis 


Clinical features 


Laboratory findings 


Temperature instability 


Respiratory alkaloses 


Tachypnea 


Hypoxemia 


Hyperventilation 


Increased serum lactate 


Altered mental status 


levels 


Oliguria 


Leukocytosis and 


Tachycardia 


increased neutrophil 


Peripheral vasodilation 


concentration 




Eosinopenia 




Thrombocytopenia 




Anemia 




Proteinuria 




Mildly elevated serum 




bilirubin levels 



III. Treatment of septic shock 

A. Early management of septic shock is aimed at 
restoring mean arterial pressure to 65 to 75 mm 
Hg to improve organ perfusion. Clinical clues to 
adequate tissue perfusion include skin 
temperature, mental status, and urine output. 
Urine output should be maintained at >20 to 30 
mL/hr. Lactic acid levels should decrease within 
24 hours if therapy is effective. 

B. Intravenous access and monitoring 

1. Intravenous access is most rapidly obtained 
through peripheral sites with two 16- to 
1 8-gauge catheters. More stable access can be 
achieved later with central intravenous access. 
Placement of a large-bore introducer catheter in 
the right internal jugular or left subclavian vein 
allows the most rapid rate of infusion. 

2. Arterial lines should be placed to allow for more 
reliable monitoring of blood pressure. 
Pulmonary artery catheters measure cardiac 
output, systemic vascular resistance, pulmonary 
artery wedge pressure, and mixed venous 
oxygen saturation. These data are useful in 
providing rapid assessment of response to 
various therapies. 

C. Fluids 

1. Aggressive volume resuscitation is essential in 
treatment of septic shock. Most patients require 
4 to 8 L of crystalloid. Fluid should be 
administered as a bolus. The mean arterial 
pressure should be increased to 65 to 75 mm 
Hg and organ perfusion should be improved 
within 1 hour of the onset of hypotension. 

2. Repeated boluses of crystalloid (isotonic 
sodium chloride solution or lactated Ringer's 
injection), 500 to 1,000 mL, should be given 
intravenously over 5 to 10 minutes, until mean 
arterial pressure and tissue perfusion are 
adequate (about 4 to 8 L total over 24 hours for 
the typical patient). Boluses of 250 mL are 
appropriate for patients who are elderly or who 
have heart disease or suspected pulmonary 
edema. Red blood cells should be reserved for 
patients with a hemoglobin value of less than 1 
g/dL and either evidence of decreased oxygen 
delivery or significant risk from anemia (eg, 
coronary artery disease). 

D.Vasoactive agents 

1. Patients who do not respond to fluid therapy 
should receive vasoactive agents. The primary 
goal is to increase mean arterial pressure to 65 
to 75 mm Hg. 

2. Dopamine (Intropin) traditionally has been 
used as the initial therapy in hypotension, 
primarily because it is thought to increase 
systemic blood pressure. However, dopamine is 
a relatively weak vasoconstrictor in septic 
shock. 

3. Norepinephrine (Levophed) is superior to 
dopamine in the treatment of hypotension 
associated with septic shock. Norepinephrine is 
the agent of choice for treatment of hypotension 
related to septic shock. Dobutamine (Dobutrex) 
should be reserved for patients with a 



persistently low cardiac index or underlying left 
ventricular dysfunction. 



Hemodynamic effects of vasoactive agents 


Agent 


Dose 


Effect 






CO 


MA 
P 


SVR 


Dopamin 

e 

(Inotropin 

) 


5-20 

mcg/kg/mi 

n 


2+ 


1 + 


1 + 


Norepin- 
ephrine 
(Levophe 

d) 


0.05-5 

mcg/kg/mi 

n 


-101+ 


2+ 


2+ 


Dobutam 
ine 

(Dobutre 
x) 


5-20 

mcg/kg/mi 

n 


2+ 


-101+ 




Epinephri 
ne 


0.05-2 

mcg/kg/mi 

n 


2+ 


2+ 


2+ 


Phenylep 
hrine 
(Neo-Syn 
ephrine) 


2-10 

mcg/kg/mi 

n 


-10 


2+ 


2+ 



E. Antibiotics should be administered within 2 hours 
of the recognition of sepsis. Use of vancomycin 
should be restricted to settings in which the 
causative agent is most likely resistant 
Enterococcus, methicillin-resistant Staphylococcus 
aureus, or high-level penicillin-resistant 
Streptococcus pneumoniae. 



Recommended Antibiotics in Septic Shock 


Suspected 
source 


Recommended antibiotics 


Pneumonia 


Second- or third-generation 
cephalosporin (cefuroxime, ceftazidime, 
cefotaxime, ceftizoxime) plus macrolide 
(antipseudomonal beta lactam plus 
aminoglycoside if hospital-acquired) 


Urinary tract 


Ampicillin plus gentamicin (Garamycin) 
or third-generation cephalosporin 
(ceftazidime, cefotaxime, ceftizoxime) 


Skin or soft 
tissue 


Nafcillin (add metronidazole [Flagyl, 
Metro IV, Protostat] or clindamycin if 
anaerobic infection suspected) 


Meningitis 


Third-generation cephalosporin 
(ceftazidime, cefotaxime, ceftizoxime) 


Intra-abdominal 


Third-generation cephalosporin 
(ceftazidime, cefotaxime, ceftizoxime) 
plus metronidazole or clindamycin 


Primary 
bacteremia 


Ticarcillin/clavulanate (Timentin) or 
piperacillin/tazobactam(Zosyn) 



Dosages of Antibiotics Used in Sepsis 


Agent 


Dosage 


Ceftizoxime (Cefizox) 


2gmlVq8h 


Ceftazidime (Fortaz) 


2glVq8h 


Cefotaxime (Claforan) 


2 gm q4-6h 


Cefuroxime (Kefurox, 
Zinacef) 


1.5glVq8h 


Cefoxitin (Mefoxin) 


2 gm q6h 


Cefotetan (Cefotan) 


2gmlVq12h 


Piperacillin/tazobactam 
(Zosyn) 


3.375-4.5 gm IV q6h 


Ticarcillin/clavulanate (Tim- 
entin) 


3.1 gmlVq4-6h (200-300 
mg/kg/d) 


Ampicillin 


1-3.0 gm IV q6h 



Agent 


Dosage 


Ampicillin/sulbactam 
(Unasyn) 


3.0 gm IVq6h 


Nafcillin (Nafcil) 


2 gm IV q4-6h 


Piperacillin, ticarcillin, 
mezlocillin 


3gmlVq4-6h 


Meropenem (Merrem) 


1 gm IVq8h 


Imipenem/cilastatin 
(Primaxin) 


1.0gmlVq6h 


Gentamicin or tobramycin 


2 mg/kg IV loading dose, 
then 1.7 mg/kg IVq8h 


Amikacin (Amikin) 


7.5 mg/kg IV loading dose, 
then 5 mg/kg IVq8h 


Vancomycin 


1 gmlVq12h 


Metronidazole (Flagyl) 


500 mg IV q6-8h 


Clindamycin (Cleocin) 


600-900 mg IV q8h 


Linezolid (Zyvox) 


600mglV/POq12h 


Quinupristin/dalfopristin 
(Synercid) 


7.5 mg/kg IVq8h 



1. Initial treatment of life-threatening sepsis 
usually consists of a third-generation 
cephalosporin (ceftazidime, cefotaxime, 
ceftizoxime)orpiperacillin/tazobactam(Zosyn). 
An aminoglycoside (gentamicin, tobramycin, or 
amikacin) should also be included. 
Antipseudomonal coverage is important for 
hospital- or institutional-acquired infections. 
Appropriate choices include an 
antipseudomonal penicillin, cephalosporin, or 
an aminoglycoside. 

2. Methicillin-resistant staphylococci. If line 
sepsis or an infected implanted device is a 
possibility, vancomycin should be added to the 
regimen to cover for methicillin-resistant Staph 
aureus and methicillin-resistant Staph 
epidermidis. 

3. Vancomycin-resistant enterococcus (VRE): 
An increasing number of enterococcal strains 
are resistant to ampicillin and gentamicin. The 
incidence of vancomycin-resistant 
enterococcus (VRE) is rapidly increasing. 

a. Linezolid (Zyvox) is an oral or parenteral 
agent active against vancomycin-resistant 
enterococci, including E. faecium and E. 
faecalis. Linezolid is also active against 
methicillin-resistant staphylococcus aureus. 

b. Quinupristin/dalfopristin (Synercid) is a 
parenteral agent active against strains of 
vancomycin-resistant enterococcusfaecium, 
but not enterococcus faecalis. Most strains 
of VRE are enterococcusfaecium. 

F. Other therapies 

1. Hydrocortisone (100 mg every 8 hours) in 
patients with refractory shock significantly 
improves hemodynamics and survival rates. 
Corticosteroids may be beneficial in patients 
with refractory shock. 

2. Activated protein C (drotrecogin alfa 
[Xigris]) has antithrombotic, profibrinolytic, and 
anti-inflammatory properties. Activated protein 
C reduces the risk of death by 20%. Activated 
protein C is approved for treatment of patients 
with severe sepsis who are at high risk of 
death. Drotrecogin alfa is administered as 24 
mcg/kg/hr for 96 hours. There is a small risk of 
bleeding. Contraindications are 
thrombocytopenia, coagulopathy, recent 
surgery or recent hemorrhage. 

References: See page 112. 



Nutrition in the Surgical Patient 



Nutritional requirements are based on the patient's 
nutritional needs, stress and severity of illness. 
Requirements are divided into non-protein calories per 
kilogram (npc/kg) and grams of protein per kilogram 
(gm protein/kg) per 24-hour period. 



Nutritional Requirements 


Patient 


Non-protein 
calories/kg 


Protein 


Well- 
nourished, 
unstressed 


20-25 
npc/kg/day 


1 gm/kg 


Minimal stress 
(post-op) 


25-30 
npc/kg/day 


1-1.2 gm/kg 


Moderate 

stress 

(multiple 

trauma, 

infection) 


30-35 
npc/kg/day 


1.2-1.5 gm/kg 


Severe stress 
(severe 
sepsis, critical 
illness) 


35-40 
npc/kg/day 


1.5-2.0 gm/kg 


Extreme 
stress (burns> 
40% body 
surface area) 


40-45 
npc/kg/day 


2.0-2.5 gm/kg 



A. Sources of non-protein calories 

1 . Carbohydrate solutions contain dextrose, 
which contains 3.4 kcal/gm 

2. Lipid solutions contain 9.1 kcal/gm 

B. Protein calories. Amino acid solutions contain 
protein in a concentration of 4 kcal/gm 

. Enteral nutrition 

A. Enteral nutrition is more physiologic and technically 
easier to administer than parenteral nutrition. 
Enteral nutrition can be administered via 
nasogastric, nasoduodenal or nasojejunal tubes, or 
gastrostomy or jejunostomy tubes. 

B. Continuous enteral infusion 

1. Initial enteral solution infusion starts at 30 m/hr. 
increase rate by 30 mL at 4-hour intervals as 
tolerated until the final rate is achieved. 
Residual volume should be measured every 4 
hours; hold feedings for 1 hour if the residual is 
greater than 2 times the infusion rate. 

2. Gastric/duodenal feedings: Start with full 
strength formula and increase the rate until the 
goal is achieved. 

3. Jejunal feedings: Start with 1/4 strength 
formula. Increase the rate until the goal is 
achieved. Once at goal rate, change to 1/2 
strength formula for 4-8 hours, then % strength 
formula for 4-8 hours, then full strength formula 
for 4-8 hours. This method allows the mucosa of 
the distal small bowel to adjust to the increased 
osmolarity of enteral formulas. 

C. Bolus feedings: Give 50-100 cc enteral nutrition 
every 3 hours initially. Increase by 50 cc each 
feeding until the goal of 250-300 cc q 3-4 hours is 
achieved. Flush tube with 100 cc of water after 
each bolus. 

D. Promotility agents are given to improve gastric 
emptying 

1. Metoclopramide (Reglan) 5-10 mg PO/IV q6h 
OR 

2. Erythromycin 1 25 mg IV or via nasogastric tube 
q8h. 

E. Antidiarrheal Agents 

1. Loperamide (Imodium) 2-4 mg q6h. 

2. Diphenoxylate/atropine (Lomotil) 2.5-5.0 mg q4- 
6h. 



Total Parental Nutrition 



Indications for total parenteral nutritions: 

Prolonged post-operative ileus, inability to take oral 
feedings for more than 5 days, severe malnutrition, 
intestinal fistula, pancreatitis. Total parenteral nutrition 
should be given via a central catheter because of high 
osmolality. 

Standard solutions and components 
A. Dextrose solutions. Various concentrations are 
available. One gram of dextrose yields 3.4 kcal. 



Dextrose Solutions 


Solution 


Concentration 


Calories 


10% 


100 gm/l iter 


340 kcal/liter 


20% 


200 gm/liter 


680 kcal/liter 


50% 


500 gm/liter 


1700 kcal/liter 


70% 


700 gm/liter 


2380 kcal/liter 



B. Lipid solutions consist of lipid emulsions of long- 
chain triglycerides. These are usually given in 500 
cc volumes at 32 cc/hour for 16 hours. 



Lipid Solutions 


Solution 


Concentration 


Calories 


10% 


50 gm/500 cc 


500 kcal 


20% 


100gm-500cc 


1000 kcal 



C.Amino acid solutions supply protein. Various 
types of solutions at various concentrations are 
available. 



Amino Acid Solutions 


Solution 


Concentration 


Indications 


Ami nosy n 7% 


70 gm/liter 


Standard 


Aminosyn-HBC* 
7% 


70 gm/liter 


Hypercatabolism 


Aminosyn-RF 
5.2% 


52 gm/liter 


Renal Failure 


HepatAmine 8% 


80 gm/liter 


Liver Failure 


FreAmine 10% 


100 gm/liter 


Fluid Overload 

(highly 

concentrated) 


*High-branched chain aminoacid formulas may prevent 
muscle breakdown and may prevent hepatic 
encephalopathy. 



D. Electrolyte requirements should be adjusted daily 
based on patient labs. 



Electrolyte Requirements 


Usual concentration 


Range 


Sodium 60 (meg/L) 


0-150 meq/L 


Potassium 20 (meg/L) 


0-80 meq/L 


Acetate* 50 (meg/L) 


50-150 meq/L 


Chloride 50 (meg/L) 


0-1 50 meq/L 


Phosphate 15 (meg/L) 


0-30 meq/L 


Calcium** 4.5 (meg/L) 


0-20 meq/L 


Magnesium 5.0 (meg/L) 


5-1 5 meq/L 


* Acetate is used in addition to chloride to help prevent 
hyperchloremic acidosis. One-third to one half of 
sodium and potassium should be supplied in the form of 
acetate rather than chloride. 

** Calcium should be given as calcium gluconate or 

calcium chloride. One gram of calcium supplies 4.5 meq 
of calcium. 



1 . Other additives 

a. Multivitamins 1 amp daily 

b. Vitamin K 10 mg each week 

c. Trace elements chromium, copper, 
manganese, zinc, selenium 

E. Ordering Total Parenteral Nutrition 

1. Step One. Determine the daily non-protein 
calories (dextrose and lipid) and grams or protein 
(amino acid) that the patient needs. 

Non-protein calory requirement/kg/day = wt in kg 
x npc requirement/kg/d 

Protein requirement = wt in kg x protein 



requirement/kg/d 



Step Two. Non-protein calories consist of lipids 
and carbohydrate (dextrose) solutions. The 
amount of each component should be determined . 
500cc of 10% Intralipid solution will supply 
approximately 500 npc kcal. The patient will 
require the remaining non-protein calories from 
the dextrose solution. If using D50, the volume of 
D50 = npc x 1000 cc/1700 kcal. 
Step Three. Protein calories are supplied by 
amino acid solutions. 



Vol of 7% Aminosyn = gm protein required/dx 100 
cc/7 gm 

4. Step Four. Combine the above volumes to 
determine total volume and rate. The dextrose 
and amino acid solutions are mixed together and 
given over 24 hours. The lipid solution is infused 
separately over 24 hours. 

F. Methods of delivery 

1 . Continuous infusion of the solutions over 24 
hours is the most common method of 
administration. The TPN solution should be 
initiated slowly at 40 cc/hr for the first 24 hours. 
The rate can then be gradually increased by 30 
cc/hr every four hours until the goal rate is 
reached. 

2. Cyclic total parenteral nutrition 12-hour night 
schedule. Taper continuous infusion in the 
morning by reducing the rate to half of the 
original rate for one hour. Further reduce the 
rate by half for an additional hour, then 
discontinue. Restart TPN in the afternoon. 
Taper at the beginning and end of cycle. 

G. Laboratory examinations 

1. Baseline Labs: CBC, electrolytes, liverfunction 
tests, prealbumin, transferrin, triglyceride level, 
chest x-ray for line placement 

2. DailyLabs: Electrolytes, calcium, phosphorous 
until stable; glucometer checks with insulin 
sliding scale every 4-6 hours 

3. Weekly Labs: CBC, electrolytes, calcium, 
phosphorous, liver function tests, triglyceride 
level (4-6 hours after completion of lipid infusion; 
should be maintained <200 mg/dl) 

4. Nutritional Assessment to determine 
adequacy of nutritional supplementation: 

a. Prealbumin or transferrin weekly 

b. 24-hour urine for urine urea nitrogen (to 
calculate nitrogen balance 

III. Peripheral parenteral nutrition (PPN) can be 
delivered via peripheral veins. 

A. The goal of PPN is to provide enough non-protein 
calories to prevent catabolism and the breakdown 
of visceral proteins. Peripheral parenteral nutrition 
is not meant to create a positive nitrogen balance 
or anabolic state, and it should be used for short- 
term support only. 

B. PPN usually consists of a 3% amino acid solution 
mixed with dextrose 20% or glycerol. Intralipids 
(10% or 20%) can also be given peripherally to 
supply extra calories. 



Central Venous Catheterization 

I. Indications for central venous catheter can- 
nulation: Monitoring of central venous pressures in 
shock or heart failure; management of fluid status; 
administration of total parenteral nutrition; prolonged 
antimicrobial therapy or chemotherapy. 

II. Location of catheterization site 

A. The internal jugular approach should not be used 
in patients with a carotid bruit, carotid stenosis, or 
an aneurysm. 

B. The subclavian approach should be avoided in 
patients with emphysema or bullae. 

C. The external jugular or internal jugular approach by 
direct cut-down may be preferable in patients with 
coagulopathy or thrombocytopenia. 

D. If a chest tube already in place, the catheter should 
be placed on the same side as the chest tube. 

III. Technique of insertion into the external jugular 

vein 

A. The external jugular vein courses from the angle of 
the mandible to behind the middle of clavicle, 
where it joins with the subclavian vein. Place 
patient in Trendelenburg's position, and apply 
digital pressure to the external jugular vein above 
clavicle to distend the vein. Cleanse the skin with 
Betadine iodine solution using sterile technique. 



Inject 1% lidocaine to produce a skin weal. 

B. With an 18-gauge, thin-wall needle, advance the 
needle into the vein. Then pass a J-guidewire 
through the needle; the wire should advance with- 
out resistance. Remove the needle, maintaining 
control over the guidewire at all times. Nick the skin 
with a No. 1 1 scalpel blade. 

C. With guidewire in place, pass the central catheter 
over the wire, and remove the guidewire after the 
catheter is in place. Cover the catheter hub with a 
finger to prevent air embolization. 

D. Attach a syringe to the catheter hub, and ensure 
that there is free back-flow of dark venous blood. 
Attach the catheter to an intravenous infusion at a 
keep open rate. Secure the catheter in place with 
2-0 silk suture and tape. 

E. Obtain a chest x-ray to confirm position and rule 
out pneumothorax. The catheter should be remo- 
ved and changed within 3-4 days. 

IV. Internal jugular vein cannulation. The internal 
jugularvein is positioned behind the sternocleidomas- 
toid muscle, lateral to the carotid artery. The catheter 
should be placed at a location at the upper confluence 
of the two bellies of sternocleidomastoid at the level of 
cricoid cartilage. 

A. Place the patient in Trendelenburg's position, and 
turn the patient's head to the contralateral side. 
Choose a location on the right or left. If lung 
function is symmetrical and no chest tubes are in 
place, the right side is preferred because of the 
direct path to the superior vena cava. Prepare the 
skin with Betadine solution using sterile technique 
and drape the area. Infiltrate the skin and deeper 
tissues with 1% lidocaine. 

B. Palpate the carotid artery. Using a 22-gauge scout 
needle and syringe, direct the needle toward the 
ipsilateral nipple at a 30 degree angle to the neck. 
While aspirating, advance the needle until the vein 
is located and blood back flows into the syringe. 

C. Remove the scout needle and advance an 18- 
gauge, thin wall, catheter-over-needle (with an at- 
tached syringe) along the same path as the scout 
needle. When back flow of blood is noted into the 
syringe, advance the catheter into the vein. 
Remove the needle and confirm back flow of blood 
through the catheter and into the syringe. Remove 
the syringe and cover the catheter hub with a finger 
to prevent air embolization. 

D. With the catheter in position, advance a guidewire 
through the catheter. The guidewire should 
advance easily without resistance. 

E. With the guidewire in position, remove the catheter 
and use a No. 11 scalpel blade to nick the skin. 
Place the central vein catheter over the wire, hol- 
ding the wire secure at all times. Pass the catheter 
into the vein, and suture the catheter to the skin 
with O silk suture. Tape the catheter in place, and 
connect it to an IV infusion at a keep open rate. 

F. Obtain a chest x-ray to rule out pneumothorax and 
confirm position. 

V. Subclavian vein cannulation 

A. The subclavian vein is located in the angle formed 
by the medial 1/3 of clavicle and the first rib. 

B. Position the patient supine with a rolled towel 
located longitudinally between the patient's scap- 
ulae, and turn the patient's head towards the con- 
tralateral side. Prepare the area with Betadine 
iodine solution, and, using sterile technique, drape 
the area and infiltrate 1% lidocaine into the skin 
and tissues. 

C. Use a 16-gauge needle, with syringe attached, to 
puncture the mid-point of the clavicle, advancing 
until the clavicle bone and needle come in contact. 

D. Then slowly probe down until the needle slips 
under the clavicle. Advance the needle slowly 
towards the vein until the needle enters the vein, 
and a back flow of venous blood enters the 
syringe. Remove the syringe, and cover the 
catheter hub with a finger to prevent air 
embolization. 

E. With the 16-gauge catheter in position, advance a 
0.89 mm x 45 cm guidewire through the catheter. 
The guidewire should advance easily without re- 
sistance. With the guidewire in position, remove 
the catheter, and use a No. 1 1 scalpel blade to nick 
the skin. Pass the dilator over the wire. 

F. Place the central line catheter over the wire, hol- 
ding the wire secure at all times. Pass the catheter 
into the vein, and suture the catheter to the skin 
with 2-0 silk suture, tape the catheter in place and 
connect to IV infusion. Obtain a chest x-ray to 
confirm the position of the catheter tip and rule out 
pneumothorax. 



Pulmonary Artery Catheterization 

1 . Cannulate a vein using the technique above, such as 
the subclavian vein or internal jugular. Advance a 
guidewire through the cannula, and remove the 
cannula. Nick the skin with a number 1 1 scalpel blade 
adjacent to the guidewire, and pass a number 8 
French introducer over the wire and into the vein. 
Connect the introducer to an IV fluid infusion at a 
keep open rate, and suture introducer to the skin with 
2-0 silk. 

3. Pass the proximal end of the pulmonary artery 
catheter (Swan Ganz) to an assistant for connection 
to a continuous flush transducer system. 

4. Flush the distal and proximal ports with heparin 
solution, removing all bubbles, and check balloon in- 
tegrity by inflating 2 cc of air. Check pressure transd- 
ucer response by moving the distal tip quickly. 

5. Pass the catheter through the introducer into the vein 
10-20 cm, then inflate the balloon, and advance the 
catheter until the balloon is in or near the right atrium. 

6. The correct distance to the entrance of the right atrium 
is determined from the site of insertion: 

Right antecubital fossa: 35-40 cm. 
Left antecubital fossa: 45-50 cm. 
Right internal jugular vein: 10-15 cm. 
Subclavian vein: 10 cm. 
Femoral vein: 35-45 cm. 

7. Run a continuous monitoring strip to record pressures 
as the PA catheter is advanced. Advance the balloon , 
inflated with 0.8-1.0 cc of air, while monitoring 
pressures and wave forms. Advance the catheterthro- 
ugh the right ventricle into the main pulmonary artery 
until the catheter enters a distal branch of the pul- 
monary artery and is stopped by impaction (as 
evidenced by a pulmonary wedge pressure wave 
form). 

8. Do not advance catheterwith balloon deflated, and do 
not withdraw the catheter with the balloon inflated. 
After placement, obtain a chest x-ray to verify that the 
tip of catheter is no farther than 3-5 cm from the 
midline, and no pneumothorax is present. 



Normal Pulmonary Artery Catheter 
Values 

Right atrial pressure 1-7mmHg 

RVP Systolic 15-25 mmHg 

RVP Diastolic 8-15 mmHg 

Pulmonary artery pressure 

PAP Systolic 15-25 mmHg 

PAP Diastolic 8-15 mmHg 

PAP Mean 10-20 mmHg 

PCWP 6-12 mmHg 

Cardiac Output 3.5-5.5 L/min 

Cardiac Index 2.0-3.2 L/min/nr 

Systemic Vascular Resistance 800-1 200 dyne/- 
sec/cm 2 



Venous Cutdown 

Procedures 

1 . Obtain a venous cutdown tray, or a minor procedure 
tray and instrument tray with a silk suture (3-0, 4-0) 
and a catheter. This procedure will require sterile 
gloves, sterile towels/drapes, 4x4 gauze sponges, 
povidone-iodine solution, 5 cc syringe, 25 gauge 
needle, 1 % lidocaine with epinephrine, adhesive tape, 
scissors, needle holder, hemostat, scalpel and blade, 
3-0 or 4-0 silk suture. 

2. Apply a tourniquet proximal to the site, and identify the 
vein. Remove the tourniquet and prep the skin with 
povidone-iodine solution and drape the area. Infiltrate 
the skin with 1% lidocaine, then incise the skin 
transversely. 

3. Spread the incision long-wise in the direction of the 
vein with a hemostat and dissect adherent tissue from 
the vein. Lift the vein and pass two chromic or silk ties 
(3-0 or 4-0) behind the vein. 

4. Tie off the distal suture, using the upper tie for traction. 
Make a transverse nick in the vein. If necessary, use a 
catheter introducer to hold the lumen of the vein open. 

5. Make a small stab incision in the skin distal to the main 
skin incision, and insert a plastic catheter or IV cannula 
through the incision, then insert it into vein. Tie the 
proximal suture, and attach an infusion of IV fluid. 
Release the tourniquet. Suture the skin with silk or 
nylon, and apply a dressing. 



Arterial Line Placement 

Procedure 

1. Obtain a 20 gauge, 1 1/2-2 inch, catheter-over-needle 
assembly (Angiocath), arterial line setup (transducer, 
tubing, pressure bag containing heparinized saline), 
armboard, sterile dressing, 1 % lidocaine, 3 cc syringe, 
25 gauge needle, and 3-0 silk. 

2. The radial artery should be used. Use the Allen test to 
verify patency of the radial artery and adequacy of 
ulnar artery collaterals. Place the extremity on an 
armboard with a gauze roll behind the wrist to 
maintain hyperextension. 

3. Prep with povidone-iodine and drape the wrist area. 
Infiltrate 1% lidocaine using a 25 gauge needle. 
Choose a site where the artery is most superficial and 
as distal as possible on the wrist. 

4. Palpate the artery with the left hand, and use other 
hand to advance a 20 gauge catheter-over-needle into 
the artery at a 30 degree angle to the skin. When a 
flash of blood is seen, hold the needle in place and 
advance the catheter into the artery; occlude the 
artery with manual pressure while pressure tubing is 
connected. 

5. If a needle and guide-wire kit is used, advance the 
guidewire into the artery, and pass the catheter over 
the guide-wire. 

6. Suture the catheter in place with 3-0 silk, and apply a 
dressing. 



Cricothyrotomy 



I. Needle cricothyrotomy 

A. Obtain a 12-14 gauge, catheter-over-needle 
(Angiocath or Jelco), 6-12 mL syringe, 3 mm 
pediatric endotracheal tube adapter, oxygen tubing, 
and a high flow oxygen source. 

B. Locate the cricothyroid membrane (the notch 
between the thyroid cartilage and cricoid cartilage). 
Cleanse the neck area with povidone-iodine 
solution, and inject 2% lidocaine with epinephrine 
if the patient is conscious. 

C. With a 12 or 14 gauge, catheter-over-needie 
assembly on the syringe, advance needle through 
the cricothyroid membrane at a 45 degree angle 
directed inferiorly. Apply back pressure on the 
syringe until air is aspirated. 

D. Advance the catheter and remove the needle, then 
attach the hub to a 3 mm endotracheal tube 
adapter connected to oxygen tubing. 

E. Administer oxygen at 15 liters per minute for 1-2 
seconds on, then 4 seconds off. Air flow is 
controlled with a Y-connector or a hole in the side 
of the tubing. 

F. The needle cricothyrotomy should be replaced with 
oral endotracheal intubation as soon as possible. A 
needle cricothyrotomy should not be used for more 
than 45 minutes, since exhalation of C0 2 is 
inadequate. 

II. Surgical cricothyrotomy 

A. Obtain a #5-#7 tracheostomy tube; tracheostomy 
tube adapter to connect to bag-mask ventilator; 
povidone-iodine solution, sterile gauze pads, 
scalpel handle, and hemostat. 

B. Clean the neck area with povidone-iodine. Locate 
the thyroid and cricoid cartilages; the cricothyroid 
membrane extends between these two cartilages. 

C. Infiltrate the overlying skin with 2% lidocaine with 
epinephrine if the patient is conscious. Stabilize the 
thyroid cartilage with the left hand, and make a 
vertical incision through the skin and subcutaneous 
tissues overlying the cricothyroid membrane, 
avoiding the large vessels that are located laterally. 

D. Make a stab incision inferiorly in the cricothyroid 
membrane with the point of the blade. Insert the 
knife handle, and rotate the handle 90 degrees to 
open the incision; or use a hemostat to dilate the 
opening. Gently insert the endotracheal tube and 
secure with tape. A tracheostomy tube or an 
endotracheal tube may be used. 

E. The surgical cricothyrotomy should be replaced 
with a formal tracheostomy within 24 hours. 

References: See page 112. 



Trauma 

Michael E. Lekawa, MD 



Management of the Trauma Patient 

I. Primary Survey of the Trauma Patient: The primary 
survey should identify immediate life threatening 
injuries. 

A = Assess airway maintenance with cervical 

spine protection. 
B = Assess breathing and administer assisted 

ventilation if required; rule out tension 

pneumothorax. 
C = Assess circulation and control hemorrhage. 
D = Assess disability and neurologic status 

(determine the level of consciousness with 

Glasgow Coma Scale). 
E = Exposure: Completely undress the patient 

and prevent hypothermia. 

II. Resuscitation phase: The primary survey and 
resuscitation of the patient should be done 
simultaneously. 

A. Assess airway and alleviate obstruction. Establish 
a definitive airway for patients with a GCS of less 
than 8 or hemodynamic instability. Protect the 
cervical spine until fractures have been excluded. 

B. Give oxygen and manage tension pneumothorax 
with needle or tube thoracostomy. 

C. Control hemorrhage by direct pressure or by 
surgical ligation. At least 2 large bore IVs should be 
places, and infuse 2-3 liters of warm Ringer's 
lactate solution (LR) as needed. Administer type 
specific or O-negative blood if the response to LR 
is inadequate. Send blood for type and cross and 
hemoglobin. 

D. If the patient has a decreased level of 
consciousness, treat hypoxemia and shock, and 
evaluate for intracranial space-occupying lesion. 

E. Give warm fluids, keep the room warm, and cover 
the patient with warm blankets. Small doses of 
short acting narcotics (Fentanyl) or 
benzodiazepines may be given as needed. 

III. Ongoing assessment and treatment 

A. Change to cross matched blood when available. 

B. Monitor for coagulopathy. The PT/PTT and 
fibrinogen level should be monitored, and fresh 
frozen plasma, cryoprecipitate or platelets should 
be administered as indicated. 

C. A nasogastric tube should be placed for 
decompression of the stomach (caution if facial 
fracture or unstable cervical spine). 

D. Shock 

1. A Foley catheter should be placed to evaluate 
urine output. Adequate resuscitation is 
suggested by improvement in physiologic 
parameters such as heart rate, systolic 
pressure, ventilatory rate, distal perfusion and 
capillary refill, pulse oximetry, arterial blood gas, 
and urine output. 

2. Reassess ABCs prior to beginning secondary 
survey. 

IV. Secondary survey 

A. Obtain an abbreviated history, including allergies, 
medications, past illness, last meal, 
event/mechanism (AMPLE history). 

B. Evaluate the completely undressed patient, front 
and back, and from head to toe. Evaluate each 
system (head and neck, chest, abdomen, 
perineum, musculoskeletal, vascular and 
neurologic). 

C. Obtain x-rays of the chest, cervical spine, and 
pelvis. Perform peritoneal lavage, and/or CT-scan 
as needed. Unstable patients should not be sent to 
the radiology department. 

D. Laboratory studies: Send type and cross for six 
units or more of packed red blood cells; complete 
blood count, platelet count, creatinine, glucose, 
ethanol level, pregnancy test, arterial blood 
gasses, UA, and urine toxicology screens. 

V. Treatment of shock 

A. Maintain airway, breathing, and circulation 
(ABCs). Rapid exsanguinating injuries take 
precedence over other injuries, including head 
injuries. 

B. Initial stabilization: Control external bleeding with 
direct external pressure. Place two 14 or 16 gauge 
intravenous lines and type and cross for packed 
red blood cells. If there is insufficient time to cross 
match, give type O-negative blood. Type specific 
blood should be given if time permits. 

C. For hypotensive patients, give an initial fluid 
challenge of 2 liters of LR over 5-10 min or 20 



ml/kg in children over 5-10 min. Assess response 

to initial fluid challenge by checking blood pressure 

and heart rate. Patients who respond with only a 

transient increase in blood pressure should be 

rechallenged with LR or blood transfusion. Blood 

loss may be continuing in these patients. 

D. Patients who do not respond to initial fluid 

challenge may have had either extensive blood 

loss or continuing bleeding, which must be 

identified (chest, abdomen, extremities, pelvis). 

Surgical intervention should be initiated. Other 

causes of hypotension include tension 

pneumothorax and cardiac tamponade. 

VI. Empiric management of coagulopathy. Consider 

empiric administration of 1 unit FFP for every 4 units 

of packed red blood cells, and consider 10 units 

platelets (or 1 unit of single donor platelets) per 6 units 

PRBC. 



Penetrating Abdominal Trauma 

I. Gun shot wounds 

A. All abdominal gun shot wounds require exploratory 
laparotomy. Tangential wounds that do not 
penetrate the peritoneal cavity may be assessed by 
peritoneal lavage or laparoscopy if the wound is 
located on the anterior abdominal wall. 

II. Stab wounds and other penetrating abdominal 
trauma 

A. Exploratory laparotomy is required if an acute 
abdomen is present or if signs of visceral injury, 
shock, hypertension, upper or lower Gl bleeding, 
evisceration or pneumoperitoneum is present. 

B. If the patient is stable and the abdominal fascia has 
been penetrated or if disruption cannot be ruled out 
by local exploration, diagnostic peritoneal lavage 
(DPL) or 24 hours of serial exams should be 
completed. 

C. Consider tetanus prophylaxis as indicated. 



Blunt Abdominal Trauma 

I. Physical findings of peritonitis or pneumoperitoneum 
on x-ray require exploratory laparotomy. 

II. If the patient has a non-acute abdomen 

A. If the patient is stable with a clinically evaluable 
abdomen who does not undergo exploratory 
laparotomy, serial abdominal exams should be 
performed. If significant tenderness or peritoneal 
signs are noted, the patient should be evaluated by 
diagnostic peritoneal lavage, CT, or laparotomy. 

B. If the clinical evaluation is inadequate, perform 
diagnostic peritoneal lavage or CT-scan to rule out 
intra-abdominal injury. 

C. If the patient is not stable (systolic blood pressure 
<100 mmHg, HR >100, decreasing hemoglobin) 
and abdominal injury is possible, diagnostic 
peritoneal lavage should be done rather than CT- 
scan. If lavage is positive, laparotomy is required. 

D. If a CT-scan shows isolated splenic or liver injury, 
and the patient remains stable, the patient may be 
observed in the ICU. Other injuries should be 
assessed with laparotomy. CT-scan is less 
sensitive for intestinal or diaphragmatic injury. 

E. If there is a significant head injury, intoxication, or 
distracting injury (eg, multiple rib fractures, pelvic 
fracture, extremity fracture), the abdominal exam is 
unreliable. These patients must be evaluated by 
diagnostic peritoneal lavage or CT-scan. 

F. If the patient is to undergo a prolonged orthopedic 
or neurosurgical procedure, the abdomen should 
be evaluated with diagnostic peritoneal lavage or 
CT-scan before the procedure. A diagnostic 
peritoneal lavage can be done in the operating 
room. 

III. Diagnostic peritoneal lavage 

A. Insert a nasogastric tube and Foley catheter to 
decompress the stomach and the bladder. Restrain 
or sedate the patient if necessary. Prep and drape 
the periumbilical region with Betadine solution and 
sterile towels. A site should be selected above or 
below umbilicus. If the patient has a pelvic fracture 
or if pregnant, the site should be located above the 
umbilicus. 

B. Infiltrate the skin and subcutaneous tissue with 1% 
lidocaine with epinephrine. Incise the skin with a 
1 .5 cm vertical incision through the subcutaneous 
tissue down to fascia. Use a No. 1 1 scalpel blade 
to make a 2-3 mm stab incision into the fascia. 
Apply traction to both sides of fascial incision with 
towel clips. An assistant should apply strong 
upward traction on clips. Dissect bluntly with a 



small hemostat to the peritoneum, then grasp and 
incise the peritoneum, and introduce a lavage 
catheter into the pelvis. 

C. Aspirate with a 12 cc syringe. If 10 cc of blood is 
returned, the lavage should be considered "grossly 
positive" which mandates an immediate 
laparotomy. If the aspirate is negative, instill 1 liter 
of LR or saline from a pressure bag. Periodically 
agitate the abdomen. When only a small amount of 
fluid remains in the bag, drop bag to the floor, and 
drain the fluid by siphon action. 

D. During the procedure, keep a sponge packed in the 
wound and hold the catheter in place. After at least 
400 cc of fluid have been removed, clamp the 
tubing and withdrawthe catheter. Close the fascial 
defect with heavy absorbable suture, and staple 
the skin. 

E. Previous abdominal surgery, morbid obesity and 
advanced cirrhosis are relative contraindication to 
diagnostic peritoneal lavage. If diagnostic 
peritoneal lavage is indicated, it should be done by 
open, rather than the closed, Seldinger technique. 

IV. Criteria for a positive peritoneal lavage 

A. Gross blood; red blood cell count <100,000 
cells/mm 3 (or 5-10,000 cells/mm 3 ) white blood cell 
count >500 cells/mm 3 . Presence of food particles, 
bile, feces, or bacteria on Gram stain. Exit of 
lavage fluid via a chest tube or bladder catheter. 

B. Amylase >20 IU/L; alkaline phosphates >3 IU. 



Head Trauma 

I. Initial management of head trauma 

A. Support airway, breathing, and circulation (ABCs). 
A cervical spine injury should be considered to be 
present in any patient with multisystem trauma. 

B. Intravenous resuscitation solutions should consist 
of isotonic Ringer's lactate (LR) or normal saline 
(NS). Fluids should be infused until the patient is 
euvolemic. 

C. Make an initial assessment of the patient during the 
primary survey (alert, voice, pain, unresponsive). 

D. Perform a mini-neurologic examination and repeat 
frequently (GCS, motor/lateralizing signs). 

E. A history, including the mechanism of injury, past 
medical history, drug intake, should be completed. 



Glasgow Coma Scale Assessment of Level of 
Consciousness 


Eye Opening 


Points 


Spontaneous 


4 


To speech 


3 


To pain 


2 


None 


1 


Verbal Response 


Oriented 


5 


Confused 


4 


Inappropriate words 


3 


Incomprehensible sounds 


2 


None 


1 


Best Motor Response 


Obeys 


6 


Localities 


5 


Withdraws 


4 


Flexion 


3 


Extension 


2 


None 


1 



Examine the skull depressed skull fractures, 
Battle's sign (blood in the ear canal or ecchymosis 
over mastoid process), Raccoon's eyes (periorbital 
ecchymosis), or rhinorrhea. If any of these signs 
are present, the patient requires admission and a 
neurosurgical consult. Nasogastric and 
nasotracheal intubation are contraindicated in 



patients with significant facial trauma because a 
cribriform plate fracture may be present. 

II. Secondary management of head trauma 

A. All patients with significant head trauma should be 
admitted for at least 24 hours of serial neurological 
exams unless Glasgow coma scale is 1 5 and there 
is only brief amnesia of events, without loss of 
consciousness. Such patients may be discharged 
with instructions if reliable observation is ensured. 

B. If the Glasgow coma scale is 14 or less, or if loss of 
consciousness was for more than a few seconds, 
a head CT-scan should be obtained. 

C. If the mechanism of injury was significantly violent 
(rollover of vehicle) or if massive upper torso 
trauma, or if any lateralizing neurologic deficits, a 
head CT-scan should be obtained. 

D. If the Glasgow coma scale is less than 8 or if 
unequal pupils, lateralizing deficits, or open head 
injury, there is a high probability of a subdural, 
epidural, or intracerebral bleed or diffuse axonal 
injury. This patient requires ICU admission after 
obtaining a CT-scan of the head and a 
neurosurgical consultation. 

III. Ongoing management of head trauma 

A. Continually reassess ABCs, ECG, systolic blood 
pressure, heart rate, and pulse oximeter. Serial 
hemoglobin or hematocrit should be obtained. 

B. Isolated head injuries rarely cause hypotension. If 
hypotension is present, the cause should be 
vigorously sought. Secondary causes of brain 
injury, such as hypoxia and hypotension, should be 
managed immediately. 

C. Stress ulcer prophylaxis with H 2 -blockers 
(ranitidine, cimetidine) or sucralfate should be 
administered. 

D. Sequential compression stockings should be 
applied if the Glasgow coma scale is less than 13, 
or if spinal cord injury or pharmacologic paralysis is 
present. 

E. Mannitol 1 gm/kg is used to treat elevated 
intracranial pressure, especially in normotensive 
patients with pupillary abnormalities, or lateralizing 
signs. Steroids are not indicated in acute head 
injuries. Hyperventilation may be used for short 
periods in select patients. 

F. Open head wounds should be cleaned and 
repaired. 

G. Tetanus prophylaxis should be given with 0.5 cc 
tetanus toxoid IM, with or without tetanus Ig 250 IU, 
IM, as indicated. 

H. Patients with an abnormal head CT-scan, 
neurologic deficit or a sustained Glasgow coma 
scale less than 14 require early neurosurgery 
consultation. 



Thoracic Trauma 

I. Management of traumatic pneumothorax 

A. Give high-flow oxygen and immediately insert a 
chest tube. Aggressive hemodynamic and 
respiratory resuscitation should be initiated. 

B. Tension pneumothorax should be treated 
immediately with a needle thoracostomy, followed 
by insertion of a chest tube. 

II. Technique of chest tube insertion 

A. The patient should be placed in the supine 
position, with involved side elevated 10-20 
degrees. The arm should be abducted at 90 
degrees. The usual site of insertion is the anterior 
axillary line at the level of the fourth intercostal 
space (nipple line). Cleanse the skin with Betadine 
iodine solution and drape the field. The 
intrathoracic tube distance can be estimated by the 
distance between the lateral chest wall and the 
apex of the lung. The intrathoracic tube distance 
should be marked with a clamp. 

B. Infiltrate 1% lidocaine into the skin, subcutaneous 
tissues, intercostal muscles, periosteum, and 
pleura using a 25-gauge needle. Use a scalpel to 
make a transverse skin incision, 2 centimeters 
wide, located over the rib just inferior to the 
interspace where the tube will penetrate the chest 
wall. 

C. A Kelly clamp should be used to bluntly dissect a 
subcutaneous tunnel from the skin incision, 
extending just over the superior margin of the rib. 
The nerve, artery, and vein located just below each 
rib should be avoided. 

D. Bluntly dissect over the rib and penetrate the 
pleura with the clamp, and open the pleura 1 
centimeter. 

E. With a gloved finger, explore the subcutaneous 
tunnel, and palpitate the lung medially. Exclude 
possible abdominal penetration, and verify correct 



location within the pleural space, use a finger to 
remove any local pleural adhesions. 

F. Use the Kelly clamp to grasp the tip of the 
thoracostomy tube (36 F, Argyle, internal diameter 
12 mm), and direct it into the pleural space in a 
posterior, superior direction for pneumothorax 
evacuation. Guide the tube into the pleural space 
until the last hole is inside the pleural space. 

G. Attach the tube to 20 cm H 2 of suction. Suture the 
tube to the skin of the chest wall using O silk. An 
untied, vertical, mattress suture may be placed to 
close the skin when the chest tube is removed in a 
few days. 

H. Apply Vaseline gauze, 4x4 gauze sponges, and 
elastic tape. Obtain a chest x-ray to verify correct 
tube placement and to evaluate re-expansion of 
the lung. 
III. Indications for thoracotomy after trauma 

A. >1500 mL blood from chest tube on insertion. 

B. >200mL blood/hour from chest tube thereafter (for 
2-4 hours). 

C. Massive air leak such that lung will not re-expand 
after a properly placed and functioning chest tube 
has been inserted. 



Tension Pneumothorax 

I. Clinical signs 

A. Tension pneumothorax will manifest as severe 
hemodynamic and respiratory compromise, a 
contralateral^ deviated trachea, and decreased or 
absent breath sounds on the involved side. 

B. Signs of tension pneumothorax may include 
hyperresonance to percussion on the affected side; 
jugular venous distention, and asymmetrical chest 
wall motion with respiration. 

II. Radiographic findings. Loss of lung markings of 
ipsilateral side is common ly seen. Flattening or 
inversion of the ipsilateral hemidiaphragm and 
contralateral shifting of the mediastinum are usually 
present. Flattening of the cardiomediastinal contour, 
and spreading of the ribs on the ipsilateral side may 
be apparent. 

III. Acute management of tension pneumothorax. 
Place a chest tube as described above. A temporary 
large-bore IV catheter may be inserted at the level of 
the second intercostal space at the mid-clavicular line 
into the pleural space, until the chest tube is placed. 



Flail Chest 

I. Clinical evaluation 

A. Flail chest occurs after two or more adjacent ribs 
become fractured in two locations. Flail chest 
usually occurs secondary to severe, blunt chest 
injury. The fractured ribs allow a rib segment, 
without bony continuity with the chest wall, to move 
freely during breathing. Hypoxia may result from 
underlying pulmonary contusion. 

B. Arterial blood gases should be measured if 
respiratory compromise is significant. If the fracture 
is in the left lower rib cage, splenic injury should be 
excluded with CT-scan. 

II. Management of flail chest 

A. Aggressive pulmonary suctioning and close 
observation for any signs of respiratory 
insufficiency or hypoxemia are recommended. 
Endotracheal intubation and positive-pressure 
ventilation is indicated for significant cases of flail 
chest if oxygenation is inadequate. Associated 
injuries, such as pneumothorax and hemothorax, 
should be treated with tube thoracostomy. 

B. Pain control with epidural or intercostal blockade 
may eliminate the need for intubation. Intubation is 
required if there are significant injuries with 
massive pulmonary contusion or poor pulmonary 
reserve. 

C. If mechanical ventilation is required, the ventilator 
should be set to assist control mode to put the flail 
segment at rest for several days. Thereafter, a trial 
of low rate, intermittent, mandatory ventilation may 
be attempted to check for return of flail, prior to 
attempting extubation. 



Massive Hemothorax 

I. Clinical evaluation 

A. Massive hemothorax is defined as greater than 
1500 mL of blood lost into the thoracic cavity. It 
most commonly occurs secondary to penetrating 
injuries. 



B. Signs of massive hemothorax include absence of 
breath sounds and dullness to percussion on the 
ipsilateral side and signs of hypovolemic shock. 

Management 

A. Volume deficit should be replaced. The chest cavity 
should be decompressed with a chest tube. Two 
large-bore intravenous lines or a central venous 
line should be inserted. A cardiothoracic surgeon 
should be consulted as soon as possible. 

B. A chest tube should be inserted. The site of 
insertion should be at the level of the fifth or sixth 
intercostal space, along the midaxillary line, 
ipsilateral to the hemothorax. The chest tube 
should be inserted in a location away from the 
injury. 

C. Penetrating wounds should be cleaned and closed. 
The wound should be covered with Vaseline 
impregnated gauze to decrease the likelihood of 
tension pneumothorax. 

D. A thoracotomy is indicated if blood loss continues 
through the chest tube at a rate greater than 200 
mL/hr for 2-3 consecutive hours. If the site of 
wound penetration is medial to the nipple anteriorly 
or medial to the scapula posteriorly, it has a higher 
probability of being associated with injury to the 
myocardium and the great vessels. If the wound is 
belowthe fourth intercostal space, abdominal injury 
should be excluded. 

E. Consider tetanus prophylaxis and empirical 
antibiotic coverage. 



Cardiac Tamponade 



Clinical evaluation 

A. Cardiac tamponade most commonly occurs 
secondary to penetrating injuries. Cardiac 
tamponade can also occur when a central line 
penetrates the wall of the right atrium. 

B. Cardiac tamponade is often manifested by Beck's 
triad of venous pressure elevation, drop in the 
arterial pressure, and muffled heart sounds. 

C. Other signs include hypovolemic shock, pulseless 
electrical activity (electromechanical dissociation), 
low voltage ECG, and enlarged cardiac silhouette 
on chest x-ray. 

D. Kussmaul's sign, rise in venous pressure with 
inspiration, may be present. Pulsus paradoxus or 
elevated venous pressure may be present. 

Management 

A. Pericardiocentesis or placement of a pericardial 
window is indicated if the patient is unresponsive to 
fluid resuscitation measures for hypovolemic 
shock. 

B. All patients who have a positive pericardiocentesis 
(recovery of non-clotting blood) due to trauma 
require open thoracotomy with inspection of the 
myocardium and the great vessels. Cardiothoracic 
surgery should be consulted. 

C. Intravenous fluids or blood should be given as 
temporizing measures on the way to the operating 
room. 

D. Other causes of hemodynamic instability or 
electromechanical dissociation that may mimic 
cardiac tamponade include tension pneumothorax, 
massive pulmonary embolism, or hypovolemic 
shock. 

E. Subxiphoid pericardial window is an equally rapid 
and safer procedure than pericardiocentesis if 
equipment and experienced surgical personnel are 
available. 



Other Life-Threatening Trauma 
Emergencies 

I. Cardiac contusions 

A. Arrhythmias are the most common consequence of 
cardiac contusions. Pump failure can also occur. 

B. Treatment. The patient should receive cardiac 
monitoring for 24 hours or longer if arrhythmias are 
present. If pump failure is suspected, cardiac 
function should be assessed with an 
echocardiogram or Swan Ganz catheter. Inotropic 
support should be provided. 

II. Pulmonary contusions 

A. Pulmonary contusions are the most common 
potentially fatal chest injuries. Respiratory failure 
and hypoxemia may develop gradually over 
several hours. The clinical severity of hypoxia does 
not correlate well with chest x-ray, however, a 
contusion visible the initial CHEST X-RAY predicts 
a need for mechanical ventilation. 

B. If pulmonary compromise is mild and there is no 



other injury, patients can be managed without 
intubation. 
C. Treatment of severe contusions, especially with 
multiple injuries consists of intubation, positive 
pressure ventilation, and PEEP. 

III. Traumatic aortic transection 

A. Diagnosis of traumatic aortic transection requires 
a high index of suspicion after severe chest 
trauma. 

B. The chest x-ray may showa widened mediastinum, 
obscured aortic knob, and a left pleural cap. The 
diagnostic standard remains aortogram, although 
transesophageal echocardiogram and spiral CT- 
scan are also useful. Management consists of 
immediate surgical repair. 

IV. Pelvic fracture 

A. Fracture of the pelvis can produce exsanguinating 
hemorrhage. Diagnosis is by physical examination, 
plain x-ray films, and CT-scan. 

B. Hemorrhage is often difficult or impossible to 
control at laparotomy. Most bleeding is venous, 
and may be decreased by external fixation of the 
pelvis. Arterial bleeding sometimes occurs, and 
requires angiographic embolization. 

C. Pelvic fractures are often associated with 
abdominal injury. Diagnostic peritoneal lavage can 
be utilized to establish the presence of internal 
hemorrhage, although CT-scan is preferred. 
Associated bladder or urethral injuries are also 
common. 

V. Traumatic esophageal injuries 

A. Clinical evaluation 

1. Esophageal injuries are usually caused by 
penetrating chest injuries, severe blunt trauma 
to the abdomen, nasogastric tube placement, 
endoscopy, or by repeated vomiting 
(Boerhaave's syndrome). 

2. After rupture, esophageal contents leak into the 
mediastinum, followed by immediate or delayed 
rupture into the pleural space (usually on left), 
with resulting empyema. 

3. A high index of suspicion is required in 
transthoracic penetrating injuries. 
Transmediastinal penetrating injuries mandate 
a search for great vessel, tracheobronchial, and 
esophageal injuries. 

B. Treatment of esophageal injuries. Surgical 
therapy consists of primary surgical repair of the 
esophagus, with drainage, or esophageal diversion 
in the neck and a gastrostomy. Perforated tumors 
should be resected. Empiric broad spectrum 
antibiotic therapy should be initiated. 

References: See page 112. 



Burns 

Bruce M. Achauer, MD 

Over 50,000 people are hospitalized every year for burn 
injuries, and more than one million people are burned 
each year in the US. Burn injuries cause over 5000 

deaths each year in the US. 

I. Initial assessment 

A. An evaluation of the Airway, Breathing, and 
Circulation (the ABCs) should receive first priority. 
The history should include the time, location and 
circumstances of the injury, where the patient was 
found, and their condition. Past medical and social 
history, current medication usage, drug allergies, 
and tetanus status should be rapidly determined. 

B. Smoke inhalation causes more than 50% of fire- 
related deaths. Patients sustaining an inhalation 
injury may require aggressive airway intervention. 
Most injuries result from the inhalation of toxic 
smoke; however, super-heated air may rarely 
cause direct thermal injury to the upper respiratory 
tract. 

C. Patients who are breathing spontaneously and at 
risk for inhalation injury should be placed on high- 
flow humidified oxygen. Patients trapped in 
buildings or those caught in an explosion are at 
higher risk for inhalation injury. These patients may 
have facial burns, singeing of the eyebrows and 
nasal hair, pharyngeal bums, carbonaceous 
sputum, or impaired mentation. A change in voice 
quality, striderous respirations, orwheezing may be 
noted. The upper airway may be visualized by 
laryngoscopy, and the tracheobronchial tree should 
be evaluated by bronchoscopy. Chest radiography 
is not sensitive for detecting inhalation injury. 

D. Patients who have suffered an inhalation injury are 
also at risk for carbon monoxide (CO) poisoning. 
The pulse oximeter is not accurate in patients with 



CO poisoning because only oxyhemoglobin and 
deoxyhemoglobin are detected. CO-oximetry 
measurements are necessary to confirm the 
diagnosis of CO poisoning. Patients exposed to CO 
should receive 100% oxygen using a 
nonrebreather face mask. Hyperbaric oxygen 
(HBO) therapy reduces the half-life of CO to 23 
minutes. 
II. Burn assessment 

A. After completion of the primary survey, a 
secondary survey should assess the depth and 
total body surface area (TBSA) burned. 

B. First-degree burns involve the epidermis layer of 
the skin, but not the dermal layer. These injuries 
are characterized by pain, erythema, and lack of 
blisters. First-degree burns are not considered in 
calculation of the TBSA burned. 

C. Second-degree burns are subdivided into 
superficial and deep partial-thickness burns. 

1. Superficial partial-thickness burn injury 
involves the papillary dermis, containing pain- 
sensitive nerve endings. Blisters or bullae may 
be present, and the burns usually appear pink 
and moist. 

2. Deep partial-thickness burn injury damages 
both the papillary and reticular dermis. These 
injuries may not be painful and often appear 
white or mottled pink. 

D. Full-thickness or third-degree burns involve all 
layers of the epidermis and dermis. They appear 
white or charred. These burns usually are painless 
because of destruction of nerve endings, but the 
surrounding areas are extremely painful. Third- 
degree burns are treated with skin grafting to limit 
scarring. 

E. Fourth-degree burns involve structures beneath 
the subcutaneous fat, including muscle and bone. 

F. Estimation of total body surface area burn is based 
upon the "rule of nines." 



Assessment of Percentage of Burn Area 


Head 


9% 


Anterior Torso 


18% 


Posterior Torso 


18% 


Each Leg 


18% 


Each Arm 


9% 


Genitalia/perineum 


1% 



Classification of Burns 



Less than 15% TBSA burns in adults or less than 10% 
TBSA burns in children or the elderly with less than 2% 
full-thickness injury 



Partial- and full-thickness burns of 15-25% TBSA in 
young adults, 10-20% in children younger than 10 and 
adults older than 40 

Full-thickness burns less than 10% TBSA, not involving 
special care area. 



Major Burns 



Greater than 25% TBSA burns in young adults or 

greater than 20% TBSA in children younger than 10 and 

adults older than 40. 

Full-thickness burns of 10% or greater. All burns of 

special care areas that are likely to result in either 

functional or cosmetic impairment (ie, face, hands, ears, 

or perineum). 

All burns complicated by inhalation injury, high-voltage 

electrical injury, or associated major trauma. High-risk 

patients include infants, the elderly, and patients with 

complicated medical problems. 



III. Management of moderate to severe burns 

A. Initial fluid resuscitation-The Parkland Formula 

1. Initiation of fluid resuscitation should precede 
initial wound care. In adults, IV fluid 
resuscitation is usually necessary in second- or 
third-degree burns involving greater than 20% 
TBSA. In pediatric patients, fluid resuscitation 
should be initiated in all infants with burns of 
10% or greater TBSA and in older children with 
burns greater than 15% or greater TBSA. 

2. Two large-bore IV lines should be placed. 



Lactated Ringer's solution is the most commonly 
used fluid for burn resuscitation. 

3. The Parkland formula is used to guide initial 
fluid resuscitation during the first 24 hours. The 
formula calls for 4 cc/kg/TBSA burn (second and 
third degree) of lactated Ringer's solution over 
the fast 24 hours. Half of the fluid should be 
administered over the first eight hours post 
burn, and the remaining half should be 
administered over the next 16 hours. The 
volume of fluid given is based on the time 
elapsed since the burn. 

4. A urine output of 0.5-1.0 mL/kg/h should be 
maintained. Patients with significant burns 
should have a Foley catheter inserted in order 
to monitor urine output. 

B. A nasogastric (NG) tube should be placed in 
patients with burns involving 20% or more TBSA in 
order to prevent gastric distention and emesis 
associated with a paralytic ileus. 

C. Antibiotics is not generally recommended for 
initial management of burns but may be considered 
in the young child with suspected streptococcal 
infection. 

D. Laboratory studies should include a complete 
blood count, electrolytes, serum glucose, BUN, and 
creatinine. Additional laboratory studies may 
include an ABG, CO HB level, urinalysis, urine 
myoglobin, coagulation studies, blood type and 
screen, toxicology screen, serum ethanol level, and 
serum protein. Patients with known heart disease 
or patients at risk for cardiac complications require 
a baseline ECG. A chest x-ray and other 
radiographs should be obtained as indicated. 

E. Pain control. After completion of the primary and 
secondary survey, priority should be given to pain 
management. Morphine 0.1 mg/kg IV or 
meperidine (Demerol) 1 -2 mg/kg IV should be used 
on an hourly basis. 

F. Complications associated with eschar formation 
may occur in circumferential, deep partial- or full- 
thickness burns. Emergent escharotomy can 
relieve life- or limb-threatening constrictions 
caused by circumferential burns. 

G. Tetanus toxoid (0.5 cc) should be administered as 
indicated. If the wound is >50% of the body surface 
area, 250 units of tetanus immune globulin should 
also be administered. 

H. Burn center transfer. Most burn patients require 
only outpatient care. Most moderate burns and all 
major burns require hospitalization. Burns involving 
more than 15% of the total body surface area in 
adults, or more than 10% in children, should be 
transferred to a burn center. Patients with other 
severe injuries, extremes of age or inhalation injury 
should also be admitted to a burn center. 
IV. Management of minor burns 

A. Home wound care, pain medication, and active 
range-of-motion exercises should be prescribed. 
Some patients require physical therapy. 

B. Tetanus status should be assessed. The burn 
wound should be cleansed with a mild soap and 
water or saline. Devitalized tissue or ruptured 
blisters should be debrided using aseptic tech- 
nique. Unruptured blisters should be left intact. 

C. Most outpatient burns are managed with closed 
dressings applied with topical antibiotics. A thin 
layer of sulfadiazine 1 % (Silvadene) cream can be 
applied to a sterile gauze using a tongue blade, 
and then the gauze should be applied to the burn. 
Dressings are changed daily or twice daily and can 
be removed under running water. 

D. Silver sulfadiazine should be avoided in patients 
with allergies to sulfonamides, pregnant women 
approaching or at term, or in newborn infants 
during the first two months of life. Silvadene 
therapy may rarely cause a transient leukopenia 
and staining of sun-exposed skin. 

References: See page 112. 



Pulmonary Disorders 

Charles F. Chandler, MD 

Airway Management and Intubation 

Orotracheal Intubation: 

Endotracheal tube size (Interior diameter): 
Women 7.0-9.0 mm 
Men 8.0 -10.0 mm. 

1. Prepare functioning suction apparatus. Have bag 
and mask apparatus 

setup with 100% oxygen; and ensure that the patient 
can be adequately bag ventilated and that suction 
apparatus is available. 

2. If sedation and/or paralysis is required, consider 

rapid sequence induction as follows: 

-Fentanyl (Sublimaze) 50 meg increments IV (1 

meg/kg) with: 
-Midazolam hydrochloride (Versed) 1 mg IV q2-3 
min, max 0.1-0.15 mg/kg; 2-5 mg IV doses q1- 
4h prn followed by: 
-Succinylcholine (Anectine) 0.6-1.0 mg/kg, at 
appropriate intervals. 
Note: These drugs may cause vomiting; therefore, 
cricoid cartilage pressure should be applied during 
intubation (Sellick maneuver). 

3. Position the patient's head in "sniffing" position 
with head flexed at neck and extended. If necessary 
elevate the head with a small pillow. 

4. Ventilate the patient with a bag mask apparatus and 
hyperoxygenate with 100% oxygen. 

5. Hold endoscope handle with left hand, and use 
right hand to open the patient's mouth. Insert blade 
along the right side of mouth to the base of tongue, 
and push the tongue to the left. If using curved blade, 
advance to the vallecula (superior to epiglottis), and 
lift anteriorly, being careful not to exert pressure on 
the teeth. If using a straight blade, place it beneath the 
epiglottis and lift anteriorly. 

6. Place endotracheal tube (ETT) into right corner of 
mouth and pass it through the vocal cords; stop just 
after the cuff disappears behind the vocal cords. If 
unsuccessful after 30 seconds, stop and resume bag 
and mask ventilation before reattempting. If 
necessary, use stilette to maintain the shape of the 
ETT; remove stilette after intubation. Application of 
lubricant jelly at the balloon facilitates passage 
through the vocal cords. 

7. Inflate cuff with syringe, keeping cuff pressure <20 
cm H 2 and attach the tube to an Ambu bag or 
ventilator. Confirm bilateral, equal expansion of the 
chest and equal bilateral breath sounds. Auscultate 
the abdomen to confirm that the ETT is not in the 
esophagus. If there is any question about proper ETT 
location, repeat laryngoscopy with tube in place to be 
sure it is located endotracheal; remove tube 
immediately if there is any doubt about proper 
location. Secure the tube with tape and note 
centimeter mark at the mouth. Suction the oropharynx 
and trachea. 

8. Confirm proper tube placement with a chest X-ray. 
The tip of the ETT should be between the carina and 
thoracic inlet, or level with the top of the aortic notch. 



Ventilator Management 

I. Indications for Ventilatory Support: Respirations 
>35, vital capacity <15 mL/kg, negative inspiratory 
force <-25, p02 <60 on 50% O,, pH <7.2, pCO, >55, 
severe, hypercapnia, hypoxia, severe metabolic 
acidosis. 

II. Initiation of ventilatory support 

A. Intubation 

1. Prepare suction apparatus, laryngoscope, 
endotracheal tube. Clear airway and place oral 
airway, then hyperventilate with bag and mask 
attached to high flow oxygen. 

2. Midazolam (Versed) 1-2 mg IV boluses until 
sedated. 2-5 mg IV doses q1-4h prn. 

3. Intubate, inflate cuff, ventilate with bag, 
auscultate chest, and suction trachea. 

B. Initial orders. FiO, = 100%, PEEP = 3-5 cm H 2 0, 
assist control 8-14 breaths/min, tidal volume = 800 
mL (10-15 mL/kg ideal body weight), set rate so 
that minute ventilation (VE) is approximately 10 
L/min. Alternatively, use intermittent mandatory 
ventilation mode with tidal volume and rate to 
achieve near-total ventilatory support. Consider 
pressure support in addition to IMV at 5-15 cm 
H,C\ 



. ABG should be obtained in 30 min, CHEST X-RAY 
for tube placement, measure cuff pressure q8h 
(maintain <20 mmHg), pulse oximeter, arterial line, 
or monitor end tidal CO,. Maintain oxygen 
saturation >90-95%. 

. Ventilator management 

1. Decreased minute ventilation. Evaluate 
patient and rule out complications (endotracheal 
tube malposition, cuff leak, excessive 
secretions, bronchospasms, pneumothorax, 
worsening pulmonary disease, sedative drugs, 
pulmonary infection). Readjust ventilator rate to 
maintain mechanically assisted minute 
ventilation of 10 L/min. If peak airway pressure 
(AWP) is >45 cm H 2 0, decrease tidal volume to 
7-8 mL/kg (with increase in rate if necessary) or 
decrease ventilator flow rate. 

2. Arterial saturation >94% and pO z >100, 
reduce FI0 2 (each 1 % decrease in FIO, reduces 
pO, by 7 mmHg); once FIO, is <60%, PEEP 
may be reduced by increments of 2 cm H 2 until 
PEEP is 3-5 cm H 2 0. Maintain O, saturation of 
>90% (pO, >60). 

3. Arterial saturation <90% and p02 <60, 
increase FI0 2 up to 60-100%, then consider 
increasing PEEP by increments of 3-5 cm H 2 
(PEEP >10 requires a PA catheter). Add 
additional PEEP until oxygenation is adequate 
with an FI0 2 of <60%. 

4. Excessively low pH, (pH <7.33): Increase rate 
and/or tidal volume. Keep peak airway pressure 
<40-50 cm H 2 if possible. 

5. Excessively high pH (>7.48): Reduce rate 
and/or tidal volume to lessen hyperventilation. If 
patient is breathing rapidly above ventilator rate, 
sedate patient. 

6. Patient "fighting" ventilator: Consider 
intermittent mandatory ventilation or 
spontaneous intermittent mandatory ventilation 
mode, or add sedation with or without paralysis 
(exclude complications or other causes of 
agitation). Paralytic agents should not be used 
without concurrent amnesia and/or sedation. 

7. Sedation 

a. Midazolam (Versed) 0.05 mg/kg IVP x1 , then 
0.02-0.1 mg/kg/hr IV infusion. Titrate in 
increments of 25-50%. 

b. Lorazepam (Ativan) 1-2 mg IV ql-2h pm 
sedation or 0.005 mg/kg IVP x1 , then 0.025- 
0.2 mg/kg/hr IV infusion. Titrate in increments 
of 25-50%. 

c. Morphine sulfate 2-5 mg IV q5min, max dose 
20-30 mg OR 0.03-0.05 mg/kg/h IV infusion 
(50-100 mg in 500 mL D5W) titrated OR 

d. Propofol (Diprivan): 50 mcg/kg bolus over 5 
min. then 5-50 mcg/kg/min. Titrate in 
increments of 5 mcg/kg/min. 

8. Paralysis 

a. Pancuronium (Pavulon) is the agent of 
choice in most settings. This agent should be 
avoided in patients with a history of asthma 
(causes mast cell degranulation) and heart 
disease (vagolytic). Load with 0.08 mg/kg, 
then start a continuous infusion of 0.02-0.03 
mg/kg/hr. 

b. Vecuronium (Norcuron) is an analog of 
pancuronium that does not cause 
hypertension or mast cell degranulation. It is 
the agent of choice with cardiac disease or 
hemodynamic instability. However, it has the 
highest incidence of post-neuromuscular 
blockade paralysis. Load with 0.1 mg/kg, 
then start a continuous infusion of 0.06 
mg/kg/hr. 

c. Cisatracurium (Nimbex) is an isomer of 
atracurium with less histamine release and 
hypotension. It has no hepatic or renal 
excretion. The dosage is 0.15 mg/kg IV, then 
0.3 mcg/kg/min IV infusion; titrate between 
0.5-1.0 mcg/kg/min. 

d. All patients on neuromuscular blocking 
agents should be continuously monitored for 
degree of blockade with a peripheral nerve 
stimulator to a train of four of 90-95%. 

9. Loss of tidal volume: If a difference between 
the tidal volume setting and the delivered 
volume occurs, check for a leak in the ventilator 
or inspiratory line. Check for a poor seal 
between the endotracheal tube cuff or 
malposition of the cuff in the subglottic area. If a 
chest tube is present, check for air leak. 

10. Weaning from mechanical ventilation 

a. Consider extubation in patients with 
cardiovascular stability, a high Pa0 2 /Fi0 2 
ratios (>200) on low PEEP, and those able to 



protect their airway. 

The most common weaning method is the 
daily trial of spontaneous breathing. While at 
the bedside, the patient is placed on CPAP 
and allowed to breath on his own. If the 
respiratory rate/tidal volume ratio is <100, 
patients are rested on the previous settings 
until the next day. 

If they fail the T-tube trial, or their respiratory 
rate to tidal volume ratio is >100, they are 
rested on the previous settings until the next 
day. 



Epistaxis 

Roger Crumley, MD 

Almost all persons have experienced a nosebleed at 
some time, and most nosebleeds resolve without 
requiring medical attention. Prolonged epistaxis, 
however, can be life-threatening, especially in the elderly 
or debilitated. 

I. Pathophysiology 

A. Anterior epistaxis, in the anterior two thirds of the 
nose, is usually visible on the septum and is the 
most common type of epistaxis. The anterior 
portion of the septum has a rich vascular supply 
known as Kiesselbach's plexus or Little's area, and 
most epistaxis originates in this region. Anterior 
bleeding can often be stopped by pinching the 
cartilaginous part of the nose. 

B. Posterior epistaxis from the posterior third of the 
nose accounts for 10% of nosebleeds. Bleeding is 
profuse because of the larger vessels in that 
location. It usually occurs in older patients, who 
have fragile vessels because of hypertension, 
atherosclerosis, coagulopathies, or weakened tis- 
sue. Posterior bleeds require aggressive treatment 
and hospitalization. 

II. Causes of epistaxis 

A. Trauma. Nose picking, nose blowing, or sneezing 
can tear or abrade the mucosa and cause bleed- 
ing. Other forms of trauma include nasal fracture 
and nasogastric and nasotracheal intubation. 

B. Desiccation. Cold, dry air and dry heat contribute 
to an increased incidence of epistaxis during the 
winter. 

C. Irritation. Upper respiratory infections, sinusitis, 
allergies, topical decongestants, and cocaine 
sniffing may cause bleeding. 

D. Less common causes of anterior epistaxis include 
Wegener's granulomatosis, mid-line destructive 
disease, tuberculosis, syphilis, and tumors. 
Epistaxis is exacerbated by coagulopathy, blood 
dyscrasia, thrombocytopenia, or anticoagulant 
medication (NSAIDs, warfarin), hepatic cirrhosis, 
and renal failure. 

E. Hypertension complicates active bleeding by 
promoting rigid arteries, and arteriosclerosis 
weakens vessels. 

III. Clinical evaluation of epistaxis 

A. The airway, breathing and circulation should be 
maintained. Hemodynamic evaluation for 
tachycardia, hypotension, or light-headedness 
should be completed immediately. Hypovolemic 
patients should be resuscitated with fluids and 
packed red blood cells. Oxygen should be 
administered and intravenous access established. 
When inserting the intravenous line, it is convenient 
to obtain blood for complete blood count and, if 
clinically indicated, type and screen, coagulation 
profile, and electrolytes. 

B. After stabilization, the site, cause, and amount of 
bleeding should be determined. Most patients do 
not require resuscitation. Posterior epistaxis in an 
elderly and debilitated patient can be life- 
threatening. 

C. Determine the side of bleeding. Unilateral nose 
bleeding suggests anterior epistaxis in 
Kiesselbach's plexus. Bilateral bleeding suggests 
posterior epistaxis caused by overflow around the 
posterior septum. 

D. Determine whether epistaxis is anterior or pos- 
terior: When the patient is upright, blood drains 
primarily from the anterior part of the nose in 
anterior bleeding, or it drains from the nasopharynx 
in posterior bleeding. 

E. Assess the duration of the nosebleed and any 
inciting incident (eg, trauma). Swallowed blood 
from epistaxis may cause melena. Hypertension, 
bleeding disorders, diabetes, alcoholism, liver 
disease, pulmonary disease, cardiac disease and 
arteriosclerosis should be assessed. 



F. Medications including aspirin, NSAIDs, warfarin, 
nasal sprays, and oxygen via nasal cannula should 
be sought. 

G. Blood tests. Hematologic tests include CBC, 
platelet count, INR, partial thromboplastin time, and 
blood for type and cross. The hematocrit does not 
immediately drop in acute hemorrhage. 

IV. Localization of the site of bleeding 

A. Sedation. When sedation is required, midazolam 
(Versed), 1-2 mg IV in adults and 0.035-0.2 mg/kg 
IV in children is recommended; overmedication 
may threaten the cough reflex which protects the 
airway. 

B. Drape the patient, and furnish an emesis basin. 
Keep the patient sitting upright or leaning forward. 
A gown, gloves, mask, and protective eyewear 
should be worn because patients may inadvertently 
cough blood. 

C. A nasal speculum and a suction apparatus with a 
#10 Frazier tip are used to aspirate blood from the 
nose and oropharynx. A bright headlight should be 
used. 

D. Anesthesia and vasoconstriction: A cotton- 
tipped applicator or cotton pledget is used to apply 
a topical anesthetic (eg, 1% tetracaine or 4% 
lidocaine) and a topical vasoconstrictor (eg, 1% 
ephedrine, 1% phenylephrine, or 0.05% 
oxymetazoline) to the entire nasal mucosa. If a 
bleeding site isobserved, press the vasoconstrictor 
applicator directly to that site. 

E. Visualization of bleeding site 

1. The nasal speculum should be used to localize 
active bleeding. Kiesselbach's plexus and the 
inferior turbinate are the most frequent sites of 
bleeding. 

2. Posterior bleeding may be located by applying 
suction; when the suction tip is at the site of 
bleeding, blood will no longer well up. 

V. Management of hemorrhage 

A. Anterior septal hemorrhage can sometimes be 
stopped by nose pinching alone and by having the 
patient sit upright. If bleeding continues, application 
of the topical anesthetic and vasoconstrictor may 
stop it. 

B. Cauterization 

1 . Bleeding sites that can be visualized should be 
cauterized with silver nitrate sticks; they work 
best in a dry field. 

2. The septum should not be cauterized bilaterally 
at the same level because cartilaginous 
necrosis and septal perforation may result. 

C. Hemostatic agents. Application of a hemostatic 
material (eg, microfibrillar collagen or oxidized 
regenerated cellulose) to the bleeding site may be 
useful. These products do not require removal 
because they dissolve in the nose after several 
days. The patient should lubricate the nose 
regularly with saline nasal spray and to use 
bacitracin ointment tid. 

D. Packing. Gauze packing or a sponge pack applied 
to the anterior portion of the nose may be used as 
a tamponade for uncontrolled bleeding sites. These 
packs stay in place for 5 days. 

E. Anterior nasal pack 

1. The pack consists of 72 inches of half-inch 
gauze impregnated with antibiotic ointment. 
Topical anesthesia is necessary. 

2. Use the nasal speculum to open the vestibule 
vertically. With the bayonet forceps, grasp the 
gauze approximately 10 cm from the end; place 
layers along the floor of the nose all the way 
back to the nasopharynx. 

3. An oral broad-spectrum antibiotic, such as 
cephalexin (Keflex), 500 mg orally every 6 
hours, is given while the pack is in place to 
prevent secondary sinusitis or toxic shock 
syndrome. The pack is removed after 5 days. 

F. Sponge pack 

1. This dry, compressed sponge is lubricated with 
an antibiotic ointment, then placed into the nasal 
cavity. 

2. Once moistened with blood or saline, the 
sponge expands, filling the nasal cavity and 
exerting gentle pressure on the bleeding site. 

G. Posterior pack 

1. Posterior bleeding requires a posterior pack, 
and the patient should be admitted to the 
hospital. The posterior pack requires a 
sphenopalatine block and topical anesthetic. 

2. The posterior pack is made by sewing together 
two tonsil tampons with 0-silk, leaving two 8-inch 
tails, and lubricating the tampons with antibiotic 
ointment. 

3. Place a tight anterior gauze pack, and tie the 
tails around one or two dental rolls to stabilize 



the tampons. 
H. Balloon pack 

1. An effective alternative to the tampon posterior 
pack is a 14-French, 30-mL balloon Foley 
catheter. 

2. Use the same anesthetic as for the tampon 
posterior pack, and cover the balloon with 
antibiotic ointment, and insert it through the 
nostril until the tip can be seen in the 
nasopharynx. 

References: See page 112. 



Disorders of the Alimen- 
tary Tract 



Russell A. Williams, MD 
I. James Sarfeh, MD 
S.E. Wilson, MD 



Acute Abdomen 

I. Clinical evaluation of abdominal pain 

A. Onset and duration of the pain 

1. The duration, acuity, and progression of pain 
should be assessed, and the exact location of 
maximal pain at onset and at present should be 
determined. The pain should be characterized 
as diffuse or localized. The time course of the 
pain should be characterized as either constant, 
intermittent, decreasing, or increasing. 

2. Acute exacerbation of longstanding pain 
suggests a complication of chronic disease, 
such as peptic ulcer disease, inflammatory 
bowel disease, or cancer. Sudden, intense pain 
often represents an intraabdominal catastrophe 
(eg, ruptured aneurysm, mesenteric infarction, 
or intestinal perforation). Colicky abdominal pain 
of intestinal or ureteral obstruction tendsto have 
a gradual onset. 

B. Pain character 

1 . Intermittent pain is associated with spasmodic 
increases in pressure within hollow organs. 

2. Bowel ischemia initially causes diffuse crampy 
pain due to spasmodic contractions of the 
bowel. The pain becomes constant and more 
intense with bowel necrosis, causing pain out of 
proportion to physical findings. A history of 
intestinal angina can be elicited in some 
patients. 

3. Constant pain. Biliary colic from cystic or 
common bile duct obstruction usually is 
constant. Chronic pancreatitis causes constant 
pain. Constant pain also suggests parietal 
peritoneal inflammation, mucosal inflammatory 
conditions, or neoplasms. 

4. Appendicitis initially causes intermittent 
periumbilical pain. Gradually the pain becomes 
constant in the right lower quadrant as 
peritoneal inflammation develops. 

C. Associated symptoms 

1. Constitutional symptoms (eg, fatigue, weight 
loss) suggests underlying chronic disease. 

2. Gastrointestinal symptoms 

a. Anorexia, nausea and vomiting are 
commonly associated with acute abdominal 
disorders. The frequency, character, and 
timing of these symptoms in relation to pain 
and time of the last flatus or stool should be 
determined. 

b. Constipation, obstipation, crampy pain 
and distention usually predominate in distal 
small-bowel and colonic obstruction. 
Paralytic ileus causes constipation and 
distention. 

c. Diarrhea is suggestive of gastroenteritis or 
colitis but may also be seen in partial small- 
bowel obstruction or fecal impaction. 

d. Small amounts of bleeding may 
accompany esophagitis, diverticulitis, 
inflammatory bowel disease, and left colon 
cancer. Right colon cancers usually present 
with occult blood loss. Severe abdominal 
pain accompanied by melena or 
hematochezia suggests ischemic bowel. 

e. Jaundice with abdominal pain usually is 
caused by biliary stones. Obstruction of the 
common bile duct by cancer may also cause 
pain and jaundice. 

3. Urinary symptoms. Urinary tract infections may 
cause pain in the lower abdomen (cystitis) or 
flanks (pyelonephritis). Urinary tract infections 
are characterized by dysuria, frequency, and 
cloudy urine. 

4. Recent menstrual and sexual history should 
be determined in women with acute abdominal 
pain. 

a. Menstrual cycle. Lower abdominal pain and 
a missed or irregular menses in a young 
woman suggests ectopic pregnancy. Pelvic 
inflammatory disease tendsto cause bilateral 
lower abdominal pain. Ovarian torsion may 
cause intense, acute pain and vomiting. 
Chronic pain at the onset of menses 
suggests endometriosis. 



b. Pregnancy. Ectopic pregnancy occurs in the 
first trimester. Threatened abortion, ovarian 
torsion, or degeneration of a uterine fibroid 
also may cause acute pain in women. 

D. Medications 

1. Nonsteroidal anti-inflammatory drugs 
predispose to ulcer disease. 

2. Antibiotic therapy may obscure the signs of 
peritonitis. Patients with abdominal pain and 
diarrhea who have received antibiotics may 
have pseudomembranous colitis. 

3. Anticoagulants. Warfarin therapy predisposes 
to retroperitoneal or intramural intestinal 
hemorrhage. 

4. Thiazide diuretics may rarely cause 
pancreatitis. 

E. Surgical history. Small bowel obstruction is often 
caused by postoperative adhesions. 

II. Physical examination 

A. General appearance. Peritonitis is suggested by 
shallow, rapid breathing and the patient often will 
lie still with knees flexed to minimize peritoneal 
stimulation. Patients may be pale or diaphoretic. 
Cachexia may indicate malignancy or chronic 
illness. 

B. Fever suggests an inflammatory or infectious 
etiology. Tachycardia and tachypnea may be 
caused by pain, hypovolemia, or sepsis. 
Hypothermia and hypotension often suggest an 
infectious process. Pneumonia and myocardial 
infarction may occasionally cause pain that is felt in 
the abdomen. 

C. Abdominal examination 

1. Inspection. Surgical scars should be noted. 
Distention suggests obstruction, ileus, or 
ascites. Venous engorgement of the abdominal 
wall suggests portal hypertension. Masses or 
peristaltic waves may be visible. 
Hemoperitoneum may cause bluish 
discoloration of the umbilicus (Cullen's sign). 
Retroperitoneal bleeding (eg, from hemorrhagic 
pancreatitis) can cause flank ecchymoses 
(Turner's sign). 

2. Auscultation. Borborygmi may be heard with 
obstruction. A quiet, rigid tender abdomen may 
occur with generalized peritonitis. A tender, 
pulsatile mass suggests an aortic aneurysm 
rupture. 

3. Palpation 

a. Palpation should be gently started at a point 
remote from the pain. Muscle spasm, 
tympany or dullness, masses and hernias 
should be sought. 

b. Peritoneal signs. Rigidity is caused by reflex 
spasm of the abdominal wall musculature 
from underlying inflamed parietal peritoneum. 
Stretch and release of inflamed parietal 
peritoneum causes rebound tenderness. 

c. Common signs 

(1) Murphy's sign. Inspiratory arrest from 
palpation in the right upper quadrant 
occurs when an inflamed gallbladder 
descends to meet the examiner's 
fingers. 

(2) Obturator sign. Suprapubic tenderness 
on internal rotation of the hip joint with 
the knee and hip flexed results from 
inflammation adjacent to the obturator 
internus muscle. 

(3) Iliopsoas sign. Extension of the hip 
elicits tenderness in inflammatory 
disorders of the retroperitoneum. 

(4) Rovsing's sign. Referred, rebound 
tenderness in the left lower quadrant 
suggests appendicitis. 

D. Rectal and pelvic examination 

1. Digital examination of the rectum may detect 
cancer, fecal impaction, or pelvic appendicitis. 
Stool should be checked for gross or occult 
blood. 

2. Pelvic examination. Vaginal discharge should 
be noted and cultured. Masses and tenderness 
should be sought bimanually. Adnexal or 
cervical motion tenderness indicate pelvic 
inflammatory disease. 

III. Laboratory evaluation 

A. Leukocytosis or a left shift on differential cell 
count are non-specific findings for infection. 
Leukopenia may be present in sepsis. The 
hematocrit can detect anemia due to occult blood 
loss from cancer. The hematocrit may be elevated 
with plasma volume deficits. 

B. Electrolytes. Metabolic alkalosis occurs after 
persistent vomiting. Metabolic acidosis occurs with 
severe hypovolemia or sepsis. 



C. Urinalysis. Bacteriuria, pyuria, or positive 
leukocyte esterase suggest urinary tract infection. 
Hematuria suggests urolithiasis. 

D. Liver function tests. High transaminases with mild 
to moderate elevations of alkaline phosphatase 
and bilirubin suggests acute hepatitis. High alkaline 
phosphatase and bilirubin and mild elevations of 
transaminases suggests biliary obstruction. 

E. Pancreatic enzymes. Elevated amylase and 
lipase indicates acute pancreatitis. 
Hyperamylasemia also may occur in bowel 
infarction and perforated ulcer. 

F. Serum beta-human chorionic gonadotropin is 
required in women of childbearing age with 
abdominal pain to exclude ectopic gestation. 

IV. Radiography 

A. Plain abdominal films 

1 . Acute abdomen series includes an upright PA 
chest, plain abdominal film ("flat plate"), upright 
film, and a left lateral decubitus view of the 
abdomen. 

2. Bowel obstruction 

a. Small bowel obstruction may cause 
multiple air-fluid levels with dilated loops of 
small intestine, associated with minimal 
colonic gas. 

b. Colonic obstruction causes colonic dilation 
which can be distinguished from small 
intestine by the presence of haustral 
markings and absence of valvulae 
conniventes. 

3. Free air is seen on the upright chest x-ray 
under the hemidiaphragms. Intestinal 
perforation is the most common cause of free 
air. A recent laparotomy may also cause free 
air. 

4. Stones and calcifications. Ninety percent of 
urinary stones are radiopaque. Only 15% of 
gallstones are visible on plain film. A fecalith in 
the right lower quadrant may suggest 
appendicitis. Vascular calcification may be 
visible in abdominal aneurysm. 

B. Ultrasonography is useful for evaluation of biliary 
colic, cholecystitis, or female reproductive system 
disorders. 

C. Computed tomography with or without oral 
and/or rectal contrast may help in evaluating the 
acute abdomen in the following situations: 

1. Unobtainable or highly atypical history or 
physical examination 

2. History of intraabdominal cancer 

3. Abdominal pain and fever in the immediate 
postiaparotomy period 

4. Acute pain superimposed on a history of chronic 
abdominal complaints 

5. A stable patient with suspected leaking 
abdominal aneurysm 

References: See page 112. 



Appendicitis 

About 10% of the population will develop acute 
appendicitis during their lifetime. The disorder most 
commonly develops in the teens and twenties. 
Appendicitis is caused by appendiceal obstruction, 
mucosal ischemia, infection, and perforation. 

I. Diagnosis of appendicitis 

A. Clinical presentation. Early appendicitis is 
characterized by progressive midabdominal 
discomfort, unrelieved by the passage of stool or 
flatus. Ninety percent of patients are anorexic, 70% 
have nausea and vomiting, and 1 0% have diarrhea. 
The pain migrates to the right lower quadrant after 
4-6 hours. Peritoneal irritation is associated with 
pain on movement. 

B. Physical examination 

1. Mild fever and tachycardia are common in 
appendicitis. 

2. Abdominal palpation should begin away from the 
right lower quadrant. Point tenderness over the 
right lower quadrant is the most definitive 
finding. Pain in the right lower quadrant during 
palpation of the left lower quadrant (Rovsing's 
sign) indicates peritoneal irritation. The degree 
of direct tenderness and rebound tenderness 
should be assessed. The degree of muscular 
resistance to palpation reflects the severity of 
inflammation. Cutaneous hyperesthesia often 
overlies the region of maximal tenderness. 

3. Psoas sign. With the patient lying on the left 
side, slow extension of the right hip causes local 
irritation and pain. A positive psoas sign 
indicates retroperitoneal inflammation. 



4. Obturator sign. With the patient supine, passive 
internally rotation of the flexed right hip causes 
hypogastric pain. 

5. Rectal examination should evaluate the pres- 
ence of localized tenderness or an inflammatory 
mass in the pelvis. 

6. Pelvic examination, in women, should be 
completed to assess cervical motion tenderness 
and to evaluate the presence of adnexal 
tenderness. 

7. The appendix usually is found at McBurney's 
point (two-thirds of the distance from the 
umbilicus to the anterior superior iliac spine). 

8. Diarrhea, urinary frequency, pyuria, or 
microscopic hematuria may suggest a retrocecal 
appendix, causing irritation of adjacent 
structures. 

C. Laboratory evaluation 

1. Leukocyte count greater than 11,000 cells/ul 
with polymorphonuclear cell predominance is 
common in children and young adults. 

2. Urinalysis is abnormal in 25% of patients with 
appendicitis. Pyuria, albuminuria, and hematuria 
are common. Bacteria suggest urinary tract 
infection. Hematuria suggests urolithiasis. 

3. Serum pregnancy test should be performed in 
women of childbearing age. A positive test 
suggests an ectopic pregnancy. 

D. Radiologic evaluation 

1. Abdominal x-rays. An appendicolith can be 
seen in only one-third of children and one-fifth of 
adults with appendicitis. An appendiceal mass 
can indent the cecum, and tissue edema can 
cause loss of peritoneal fat planes around the 
psoas muscle and kidney. 

2. Ultrasonography. Findings associated with 
appendicitis include wall thickening, luminal 
distention, lack of compressibility, abscess 
formation, and free intraperitoneal fluid. 

II. Differential diagnosis 

A. Gastrointestinal diseases 

1. Gastroenteritis is characterized by nausea, 
emesis prior to the onset of abdominal pain, 
malaise, fever, and poorly localized abdominal 
pain and tenderness. The WBC count is less 
frequently elevated. 

2. Meckel's diverticulitis may mimic appendicitis. 

3. Perforated peptic ulcer disease, diverticulitis, 
and cholecystitis can present similarly to 
appendicitis. 

B. Urologic diseases 

1. Pyelonephritis is associated with high fever, 
rigors, and costovertebral pain and tenderness. 
Diagnosis is confirmed by urinalysis. 

2. Ureteral colic. Renal stones cause flank pain 
radiating into the groin. Tenderness is usually 
minimal and hematuria is present. The 
intravenous pyelogram is diagnostic. 

C. Gynecologic diseases 

1. Pelvic inflammatory disease (PID). The onset 
of pain in PID usually occurs within 7 days of 
menstruation. Cervical motion tenderness, a 
white vaginal discharge, and bilateral adnexal 
tenderness suggest PID. Ultrasound can help 
distinguish PID from appendicitis 

2. Ectopic pregnancy. A pregnancy test should be 
performed in all female patients of childbearing 
age presenting with abdominal pain. 
Ultrasonography is diagnostic. 

3. Ovarian cysts can cause sudden pain by 
enlarging or rupturing. The cysts are detected by 
transvaginal ultrasonography. 

4. Ovarian torsion. The ischemic ovary often can 
be palpated on bimanual pelvic examination. 
The diagnosis is confirmed by ultrasonography. 

References: See page 112. 



Appendectomy Surgical Technique 

I. Preoperative preparation 

A. Intravenous isotonic fluid replacement should be 
initiated to achieve good urinary output and to 
correct electrolyte abnormalities. Nasogastric 
suction should be initiated if the patient is vomiting 
or if peritonitis is present. 

B. Fever is treated with acetaminophen. Broad- 
spectrum antibiotic coverage is initiated 
preoperatively. Antibiotic therapy should cover 
gram-negative and anaerobic organisms (Cefotan 
orZosyn). 

II. Surgical technique 

A. After induction of anesthesia, place an incision 
over any appendiceal mass if palpable. If no mass 
is present, make a transverse skin incision over 



McBurney's point, located two thirds of the way 
between the umbilicus and anterior superior iliac 
spine. A transverse incision allows easy extension 
medially for greater exposure. Diffuse peritonitis 
should be explored through a midline incision. 

B. Incise the subcutaneous tissues in the line of the 
transverse incision, and incise the external oblique 
aponeurosis in the direction of its muscle fibers. 
Spread the muscle with a Peon hemostat. 

C. Incise the internal oblique fascia and spread the 
incision in the direction of its fibers. Sharply incise 
the transversus abdominis muscle, transversalis 
fascia, and peritoneum. Note the presence and 
characteristics of peritoneal fluid, and send 
purulent fluid for Gram's stain and aerobic and 
anaerobic culture. 

D. Identify the base of the cecum by the converging 
taeniae coli, and raise the cecum, exposing the 
base of the appendix. Hook an index finger around 
the appendix, and gently break down any adhe- 
sions to adjacent tissues. Use gauze packing to 
isolate the inflamed appendix, and stabilize the 
appendix with a Babcock forceps. 

E. Apply two clamps to the mesoappendix, then divide 
the mesoappendix between the clamps, then firmly 
ligate belowthe clamps with 000 silk or polyglycolic 
acid sutures. Apply an encircling purse-string 
suture of 000 silk at the end of the cecum about 0.8 
cm from the base of the appendix. Place a 
hemostat at the proximal base of the appendix, and 
crush the appendix. Remove the hemostat and 
reapply it to the appendix distal to the crush. Use 
an chromic catgut suture to ligate the crushed 
area belowthe hemostat. 

F. Transect the appendix against the clamp. Invertthe 
stump into the cecum with the purse-string suture, 
and tie the purse-string suture, burying the stump. 
Irrigate the peritoneum with normal saline, and 
examine the mesoappendix and abdominal wall for 
hemostasis. Close the peritoneum with continuous 
000 catgut suture. 

G. Close the internal oblique and transversus 
abdominis with interrupted O chromic catgut. Close 
the external oblique as a separate layer. Close the 
skin and subcutaneous tissues. A soft rubber 
Penrose drain should be placed if perforation has 
occurred. It should be brought out through a stab 
incision in the lateral abdominal wall or through the 
lateral end of the incision. 

H. If the appendix is normal on inspection (5-20% of 
explorations), it should be removed, and alternative 
diagnoses should be investigated. The cecum, 
sigmoid colon, and ileum should be inspected, and 
mesenteric lymphadenopathy should be sought. 
Ovaries and fallopian tubes should be inspected for 
PID, ruptured cysts, or ectopic pregnancy. Bilious 
peritoneal fluid suggests perforation of a peptic 
ulcer or the gallbladder. 
III. Intravenous antibiotics 

A. Antibiotic prophylaxis should include coverage for 
bowel flora, including aerobes and anaerobes. 
Cefotetan (Cefotan), 1 gm IV q12h, or 
piperacillin/tazobactam (Zosyn), 4.5 gm IV q6h, 
should be given before the operation and 
discontinued after two doses postoperatively. 

B. If perforation has occurred, IV antibiotics should be 
continued for 5-10 days. Check culture on the third 
postoperative day and change antimicrobials if a 
resistant organism is present. 

References: See page 112. 



Hernias 

A hernia is an abnormal opening in the abdominal wall, 
with or without protrusion of an intraabdominal structure. 
A hernia develops in 5% of men during their lifetime. The 
most common groin hernia in males or females is the 
indirect inguinal hernia. Femoral hernias are more 
common in females than in males. 

I. Inguinal hernias 

A. Indirect hernia sacs pass through the internal 
inguinal ring lateral to the inferior epigastric vessels 
and lie within the spermatic cord. Two-thirds of 
inguinal hernias are indirect 

B. Direct hernias occur when viscera protrude 
through a weak area in the posterior inguinal wall. 
The base of the hernia sac lies medial to the inferior 
epigastric vessels, through Hesselbach's triangle, 
which is formed by the inferior epigastric artery, the 
lateral edge of the rectus sheath, and the inguinal 
ligament. 

C. Combined (pantaloon) hernias occur when direct 
and indirect hernias occur simultaneously. 



. Sliding hernias occur when part of the wall of the 
sac is formed by a viscera (bladder, colon). 
Richter's hernias occur when part of the bowel 
(rather than the entire circumference) becomes 
trapped. Only a "knuckle" of bowel enters the hernia 
sac. 

. Incarcerated hernias cannot be reduced into the 
abdominal cavity. Strangulated hernias are hernias 
with incarcerated contents and a compromised 
blood supply; intense pain indicates intestinal 
ischemia. 
Inguinal anatomy 

1. Layers of abdominal wall: Skin, subcutaneous 
fat, Scarpa's fascia, external oblique, internal 
oblique, transversus abdominous, transversalis 
fascia, peritoneum. 

2. Hesselbach's triangle: A triangle formed by the 
lateral edge of rectus sheath, the inferior 
epigastric vessels, and the inguinal ligament. 

3. Inguinal ligament: Ligament running from 
anterior superior iliac spine to the pubic tubercle. 

4. Lacunar ligament: Reflection of inguinal 
ligament from the pubic tubercle onto the 
iliopectineal line of the pubic ramus. 

5. Cooper's ligament: Strong, fibrous band 
located on the iliopectineal line of the superior 
public ramus. 

6. External inguinal ring: Opening in the external 
oblique aponeurosis; the ring contains the 
ilioinguinal nerve and spermatic cord or round 
ligament. 

7. Internal ring: Bordered superiorly by internal 
oblique muscle and inferomedially by the inferior 
epigastric vessels and the transversalis fascia. 

8. Processus vaginalis: A diverticulum of 
peritoneum which descends with the testicle and 
lies adjacent to the spermatic cord. The 
processus vaginalis may enlarge to become the 
sac of an indirect inguinal hernia. 

9. Femoral canal: Formed by the borders of the 
inguinal ligament, lacunar ligament, Cooper's 
ligament, and femoral sheath. 

. Clinical evaluation 

1. Inguinal hernias usually present as an 
intermittent mass in the groin. The symptoms 
can usually be reproduced by a purposeful 
Valsalva maneuver. A bowel obstruction may 
rarely be the first manifestation of a hernia. 

2. Physical examination. An inguinal bulge with a 
smooth, rounded surface is usually palpable. 
The bulge may become largerwith straining. The 
hernia sac can be assessed by invaginating the 
hemiscrotum with an index finger passed 
through the external inguinal ring. 

3. Radiologic evaluation. X-ray studies are not 
usually needed. Ultrasonography or CT scanning 
may be necessary to evaluate small hernias, 
particularly in the obese patient. 

. Differential diagnosis. Inguinal hernias are 
distinguished from femoral hernias by the fact that 
femoral hernias originate below the inguinal 
ligament. Inguinal adenopathy, lipomas, dilatation 
of the saphenous vein, and psoas abscesses may 
present as inguinal masses. 
Treatment 

1. Preoperative evaluation and preparation. 
Hernias should be treated surgically. If 
incarceration or strangulation has occurred, 
broad-spectrum antibiotics and nasogastric 
suction should be initiated. 

2. Reduction. In uncomplicated cases, the hernia 
should be reduced by placing the patient in 
Trendelenburg's position or by gentle pressure 
applied over the hernia. Strangulated hernias 
should not be reduced because reduction may 
cause peritoneal contamination by a necrotic 
bowel loop and release of vasoactive agents 
from infarcted tissue. 

3. Surgical repair 

a. Indirect inguinal hernias. The aponeurosis 
of the external oblique muscle should be 
opened, then the cremaster muscle is 
opened, and the contents of the cord 
identified. The hernia sac is separated from 
the cord structures and transected. The neck 
should then be ligated, and the posterior 
abdominal wall should be repaired. 

b. Direct inguinal hernias. The external 
oblique should be opened and the cord 
structures should be separated from the 
hernia sac, then the sac should be inverted. 
The posterior abdominal wall is repaired by 
approximating the inferior arch of the 
transversus muscle (conjoint tendon) to the 
iliopubic tract (Bassini repair). 



c. Lichtenstein (Tension-Free) Repair. A 
mesh plug or patch is often used to produce 
a "tension free" repair. 

II. Femoral hernias 

A. Femoral hernias account for 5% of all hernias, and 
84% of femoral hernias occur in women. 
Incarceration or strangulation occur in 25% of 
femoral hernias. 

B. In femoral hernias, the abdominal viscera and 
peritoneum protrude through the femoral ring into 
the upper thigh. The femoral ring is limited medially 
by the lacunar ligament of Gimbernat, laterally by 
the femoral vein, anteriorly and proximally by the 
inguinal ligament, and posteriorly and distally by 
Cooper's ligament. 

C. Clinical evaluation 

1. Femoral hernias may present as a tender groin 
mass, and small-bowel obstruction may 
sometimes occur. 

2. Physical examination. The hernia sac 
manifests clinically as a mass in the upper thigh, 
curving craniad over the inguinal region. It may 
appear while the patient is standing or straining 
and may disappear in the supine position. 

D. Treatment. A Cooper's ligament repair (McVey) 
through the inguinal approach is recommended. 

III. Abdominal wall hernias 

A. Incisional hernias occur at sites of previous 
incisions. Hernias occur after 14% of abdominal 
operations. 

B. Umbilical hernias are congenital defects. Most 
newborn umbilical hernias close spontaneously by 
the second year of life. Patients with ascites have a 
high incidence of umbilical hernias. 

C. Epigastric hernias occur in the linea alba above 
the umbilicus. 

D. Spigelian hernias protrude near the termination of 
the transversus abdominis muscle at the lateral 
edge of the rectus abdominis muscle. 

E. Lumbar hernias occur superior to the iliac crest or 
below the last rib. 

F. Obturator hernias pass through the obturator 
foramen and present with bowel obstruction and 
focal tenderness on rectal examination. 



Inguinal Hernia Repair Technique 

I. Indirect hernia 

A. Prep and draped the skin of the abdomen, inguinal 
region, upper thigh, and external genitalia. Place 
the incision 1 cm above and parallel to the inguinal 
ligament. Begin the incision at a point just above, 
and medial to, the pubic tubercle, and extend it to 
a point two-thirds the distance to the anterior iliac 
spine. Incise the subcutaneous fat in the length of 
the incision down to the external oblique 
aponeurosis. Clear the external oblique muscle of 
overlying fat and identify the external inguinal ring. 
Incise the aponeurosis of the external oblique, 
beginning laterally and splitting the aponeurosis in 
the direction of its fibers, taking care not to injure 
the underlying ilioinguinal nerve. Expose the 
inguinal canal in which the spermatic cord and the 
indirect inguinal hernia are located. 

B. Use blunt dissection to mobilize the spermatic cord 
and the associated hernia up to the level of the 
pubic tubercle. Lift these structures up from the 
floor of the inguinal canal, and encircle with a 
Penrose drain. Retract the drain anteriorly, and 
free the remainder of the cord from the floor of the 
canal. Use sharp and blunt dissection to incise the 
anterior muscular and fascial investments of the 
cord. 

C. Sharply incise the internal spermatic fascia, and 
locate the hernial sac; identify the spermatic artery, 
venous plexus, and vas deferens before opening 
the sac. 

D. Incise the indirect hernial sac anteriorly along its 
long axis. Place an index finger inside the hernial 
sac, and separate the sac from surrounding cord 
structures, using sharp and blunt dissection. Carry 
the dissection proximally to the internal inguinal 
ring. 

E. Close the sac with a circumferential purse-string 
suture on an atraumatic, gastrointestinal needle. 
Tie this suture, being careful that no abdominal 
organs are within the purse string. Reinforce this 
ligation by placing another transfixion suture 
through the sac 1 mm distal to the purse-string 
suture. Transect the sac a few millimeters below 
the second suture, and cut both sutures and allow 
sac to retract into the retroperitoneum. Remove the 
sac. Inspect the floor of the inguinal canal. If there 
is only minimal dilation of the internal ring, the 



hernia repair can be completed by placing a few 
interrupted sutures at the medial border of the 
internal ring and completing a Bassini repair. 

F. If the hernia is moderately sized, use a 
modification of the Bassini repair. Place a series of 
interrupted sutures in the transversalis fascia, 
beginning medially at the level of the pubic 
tubercle. Carry the sutures laterally as far as the 
medial border of the internal ring. Incorporate the 
posterior fibers of the conjoint tendon or use the 
posterior fascia of the transversalis abdominous 
muscle with some of the muscular fibers of the 
internal oblique muscle. 

G. Begin suturing medially at Cooper's ligament, and 
transition at the femoral sheath to Poupart's liga- 
ment. Place the sutures 1 cm apart, and use 
nonabsorbable monofilament 00 suture. Tie the 
sutures to reinforce the floor of the inguinal canal. 

H. Replace the ilioinguinal nerve and the spermatic 
cord in the inguinal canal, and reapproximate the 
external oblique aponeurosis over the cord with 
interrupted, nonabsorbable, 000 sutures. 

I. Close the subcutaneous tissues with interrupted, 
fine, absorbable sutures in Scarpa's fascia, and 
close the skin with staples or subcuticular sutures. 
Apply a sterile dressing. 

II. Repair of direct inguinal hernias 

A. The skin incision is the same as for repair of the 
indirect inguinal hernia. The direct inguinal hernia 
appears as a diffuse bulge in the area of 
Hesselbach's triangle, appreciated by palpating 
with a fingertip. Reduce the direct inguinal hernia 
with a series of interrupted, inverting, 00, 
nonabsorbable sutures placed in the redundant 
preperitoneal tissue. 

B. The fascial defect in Hesselbach's triangle is 
repaired with a Cooper's ligament or modified 
McVay-type repair. Sharp and blunt dissection of 
the floor of the inferior portion of the inguinal canal 
should be used to expose the lacunar ligament and 
Cooper's ligament. Dissect laterally along Cooper's 
ligament as far as the medial aspect of the femoral 
vein. A relaxing incision should be made in the 
deep portion of the anterior rectus sheath, then 
grasp the medial and superior edge of the defect in 
Hesselbach's triangle with Allis clamps. 

C. A series of interrupted sutures is placed, beginning 
medially at the pubic tubercle, and carried laterally 
as far as the femoral vein. Check the appropriate 
snugness of the deep inguinal ring. When all 
sutures are placed, tie the sutures medial to lateral. 
Replace the cord and the ilioinguinal nerve in the 
bed of the inguinal canal, and reapproximate the 
external oblique aponeurosis overthese structures. 

III. Lichtenstein (Tension-Free) Repair 

A. One of the most commonly performed open 
herniorrhaphy techniques is the tension-free 
(Lichtenstein) repair. A tension-free hernioplasty 
performed with mesh reinforcement of the inguinal 
floor significantly decreases the recurrence rate. 

B. The Lichtenstein repair is routinely performed in an 
outpatient setting with local anesthesia. A Marlex 
mesh patch is sutured to the aponeurotic tissue 
overlying the pubic bone, with continuation of this 
suture along the edge of the inguinal (Poupart's) 
ligament to a point lateral to the internal inguinal 
ring. 

C. The lateral edge of the mesh is slit to allow 
passage of the spermatic cord between the split 
limbs of the mesh. The cephalad edge of the mesh 
is sutured to the conjoined tendon, with the internal 
oblique edge overlapped by 2 cm. The two tails of 
the lateral aspect of the mesh are sutured together, 
incorporating the margin of the inguinal ligament. 

References: See page 112. 



Upper Gastrointestinal Bleeding 

When bleeding is believed to be caused by a source 
proximal to the ligament of Treitz or the source of 
bleeding is indeterminant, flexible upper gastrointestinal 
endoscopy is indicated after initial resuscitation and 
stabilization. 

I. Clinical evaluation 

A. Initial evaluation of upper Gl bleeding should 
estimate the severity, duration, location, and cause 
of bleeding. A history of bleeding occurring after 
forceful vomiting suggests Mallory-Weiss 
Syndrome. 

B. Abdominal pain, melena, hematochezia (bright red 
blood per rectum), history of peptic ulcer, cirrhosis 
or prior bleeding episodes may be present. 

C. Precipitating factors. Use of aspirin, nonsteroidal 
anti-inflammatory drugs, alcohol, or anticoagulants 
should be sought. 

II. Physical examination 

A. General: Pallor and shallow, rapid respirations 
may be present; tachycardia indicates a 1 0% blood 
volume loss. Postural hypotension (increase in 
pulse of 20 and a systolic blood pressure fall of 10- 
15 mmHg), indicates a 20-30% loss. 

B. Skin: Delayed capillary refill and stigmata of liver 
disease (jaundice, spider angiomas, parotid gland 
hypertrophy) should be sought. 

C. Abdomen: Scars, tenderness, masses, hepato- 
megaly, and dilated abdominal veins should be 
evaluated. Stool occult blood should be checked. 

III. Laboratory evaluation: CBC, SMA 12, liver function 
tests, amylase, INR/PTT, type and cross for pRBC, 
ECG. 

IV. Differential diagnosis of upper bleeding: Peptic 
ulcer, gastritis, esophageal varices, Mallory-Weiss 
tear, esophagitis, swallowed blood from epistaxis, 
malignancy (esophageal, gastric), angiodysplasias, 
aorto-enteric fistula, hematobilia. 

V. Management of upper gastrointestinal bleeding 

A. If the bleeding appears to have stopped or has 
significantly slowed, medical therapy with H2 
blockers and saline lavage is usually all that is 
required. 

B. Two 14- to16-gauge IV lines should be placed. 
Normal saline solution should be infused until 
blood is ready, then transfuse 2-6 units of pRBCs 
as fast as possible. 

C. A large bore nasogastric tube should be placed, 
followed by lavage with 2 L of room temperature 
tap water. The tube should then be connected to 
low intermittent suction, and the lavage should be 
repeated hourly. The NG tube may be removed 
when bleeding is no longer active. 

D. Oxygen is administered by nasal cannula. Urine 
output should be monitored. 

E. Serial hematocrits should be checked and 
maintained greater than 30%. Coagulopathy 
should be assessed and corrected with fresh 
frozen plasma, vitamin K, cryoprecipitate, and 
platelets. 

F. Definitive diagnosis requires upper endoscopy, at 
which time electrocoagulation, banding, and/or 
local injection of vasoconstrictors at bleeding sites 
may be completed. Surgical consultation should 
be requested in unstable patients or patients who 
require more than 6 units of pRBCs. 



Clinical Indicators of Gastrointestinal Bleeding 
and Probable Source 


Clinical 
Indicator 


Probability of 
Upper 

Gastrointestin 
al source 


Probability of 
Lower 

Gastrointestin 
al Source 


Hematemesis 


Almost certain 


Rare 


Melena 


Probable 


Possible 


Hematochezia 


Possible 


Probable 


Blood-streaked 
stool 


Rare 


Almost certain 


Occult blood in 
stool 


Possible 


Possible 



VI. Peptic Ulcer Disease 

A. Peptic ulcer disease is the commonest cause of 
upper gastrointestinal bleeding, responsible for 27- 
40% of all upper gastrointestinal bleeding episodes. 
Duodenal ulcer is more frequent than gastric ulcer. 
Three fourths of all peptic ulcer hemorrhages 
subside spontaneously. 

B. Upper gastrointestinal endoscopy is the most 
effective diagnostic technique for peptic ulcer 
disease. Endoscopic therapy is the method of 
choice for controlling active ulcer hemorrhage. 

C. Proton-pump inhibitor administration is effective 
in decreasing rebleeding rateswith bleeding ulcers. 
Therapy consists of intravenous omeprazole. 

1. Omeprazole (Prilosec) dosage is 80 mg IV, 
followed by continuous infusion with 8 mg/hrfor 
72 hrs and 20-mg per day PO. 

2. Twice daily dosing of oral proton pump inhibitors 
may be a reasonable alternative when 
intravenous formulations are not available. Oral 
omeprazole (Prilosec) for duodenal ulcer: 20 mg 
qd for 4-8 weeks. Gastric ulcers: 20 mg bid. 
Lansoprazole (Prevacid), 15 mg qd. 
Esomeprazole (Nexium) 20-40 mg qd. 

D. Indications for surgical operation include (1) 
severe hemorrhage unresponsive to initial 
resuscitative measures; (2) failure of endoscopic or 
other nonsurgical therapies; and (3) perforation, 
obstruction, or suspicion of malignancy. 

E. Duodenal ulcer hemorrhage. Suture ligation of 
the ulcer-associated bleeding artery combined with 
a vagotomy is indicated for duodenal ulcer 
hemorrhage that does not respond to medical 
therapy. Truncal vagotomy and pyloroplasty is 
widely used because it is rapidly and easily 
accomplished. 

F. Gastric ulcer hemorrhage is most often managed 
by truncal vagotomy and pyloroplasty with wedge 
excision of ulcer. 

G. Transcatheter angiographic embolization of the 
bleeding artery responsible for ulcer hemorrhage is 
recommended in patients who fail endoscopic 
attempts at control and who are poor surgical 
candidates. 

VII. Hemorrhagic Gastritis 

A. The diffuse mucosal inflammation of gastritis rarely 
manifest as severe or life-threatening hemorrhage. 
Hemorrhagic gastritis accounts for 4% of upper 
gastrointestinal hemorrhage. The bleeding is 
usually miid and self-limited. When coagulopathy 
accompanies cirrhosis and portal hypertension, 
however, gastric mucosal bleeding can be brisk 
and refractory. 

B. Endoscopic therapy can be effective for multiple 
punctate bleeding sites, but when diffuse mucosal 
hemorrhage is present, selective intra-arterial 
infusion of vasopressin may control bleeding. For 
the rare case in which surgical intervention is 
required, total gastrectomy is the most effective 
procedure. 

VIM. Mallory-Weiss syndrome 

A. This disorder is defined as a mucosal tear at the 
gastroesophageal junction following forceful 
retching and vomiting. 

B. Treatment is supportive, and the majority of 
patients stop bleeding spontaneously. Endoscopic 
coagulation or operative suturing may rarely be 
necessary. 

References: See page 112. 



Esophageal Varices 



Esophageal varices eventually develop in most patients 
with cirrhosis, but variceal bleeding occurs in only one 
third of them. The initiating event in the development of 
portal hypertension is increased resistance to portal 
outflow. 



Causes of Portal Hypertension 



Presinusoidal 

Extrahepatic causes 

Portal vein thrombosis 

Extrinsic compression of the portal vein 

Cavernous transformation of the portal vein 

Intrahepatic causes 

Sarcoidosis 

Primary biliary cirrhosis 

Hepatoportal sclerosis 

Schistosomiasis 
Sinusoidal: Cirrhosis, alcoholic hepatitis 
Postsinusoidal 

Budd-Chiari syndrome (hepatic vein thrombosis) 

Veno-occlusive disease 

Severe congestive heart failure 



Restrictive heart disease 



I. Pathophysiology 

A. Varices develop annually in 5% to 15% of patients 
with cirrhosis, and varices enlarge by 4% to 10% 
each year. Each episode of variceal hemorrhage 
carries a 20% to 30% risk of death. 

B. After an acute variceal hemorrhage, bleeding 
resolves spontaneously in 50% of patients. 
Bleeding is least likely to stop in patients with large 
varices and a Child-Pugh class C cirrhotic liver. 

II. Management of variceal hemorrhage 

A. Primary prophylaxis 

LAN patients with cirrhosis should undergo 
endoscopy to screen for varices every 2 to 3 
years. 

2. Propranolol (Inderal) and nadolol (Corgard) 
reduce portal pressure through beta, blockade. 
Beta-blockade reduces the risk of bleeding by 
45% and bleeding-related death by 50%. The 
beta-blocker dose is adjusted to decrease the 
resting heart rate by 25% from its baseline, but 
not to less than 55 to 60 beats/min. 

3. Propranolol (Inderal) is given at 10 to 480 mg 
daily, in divided doses, or nadolol (Corgard) 40 
to 320 mg daily in a single dose. 

B. Treatment of acute hemorrhage 

1. Variceal bleeding should be considered in any 
patient who presents with significant upper 
gastrointestinal bleeding. Signs of cirrhosis may 
include spider angiomas, palmar erythema, 
leukonychia, clubbing, parotid enlargement, and 
Dupuytren's contracture. Jaundice, lower 
extremity edema and ascites are indicative of 
decompensated liver disease. 

2. The severity of the bleeding episode can be 
assessed on the basis of orthostatic changes 
(eg, resting tachycardia, postural hypotension), 
which indicates one-third or more of blood 
volume loss. 

3. Blood should be replaced as soon as possible. 
While blood for transfusion is being made 
available, intravascular volume should be 
replenished with normal saline solution. Once 
euvolemia is established, the intravenous 
infusion should be changed to solutions with a 
lower sodium content (5% dextrose with 1/2 or 
1/4 normal saline). Blood should be transfused 
to maintain a hematocrit of at least 30%. Serial 
hematocrit estimations should be obtained 
during continued bleeding. 

4. Fresh frozen plasma is administered to patients 
who have been given massive transfusions. 
Each 3 units of PRBC should be accompanied 
by CaCL 2 1 gm IV over 30 min. Clotting factors 
should be assessed. Platelet transfusions are 
reserved for counts below 50,000/mL in an 
actively bleeding patient. 

5. If the patient's sensorium is altered because of 
hepatic encephalopathy, the risk of aspiration 
mandates endotracheal intubation. Placement of 
a large-caliber nasogastric tube (22 F or 24 F) 
permits tap water lavage for removal of blood 
and clots in preparation for endoscopy. 

6. Octreotide acetate (Sandostatin) is a 
synthetic, analogue of somatostatin, which 
causes splanchnic vasoconstriction. Octreotide 
is the drug of choice in the pharmacologic 
management of acute variceal bleeding. 
Octreotide infusion should be started with a 
loading dose of 50 micrograms, followed by an 
infusion of 50 micrograms/hr. Treatment is 
continued through the fifth hospital day. 
Definitive endoscopic therapy is performed 
shortly after hemostasis is achieved. 

7. Endoscopic therapy 

a. A sclerosant (eg, morrhuate [Scleromate]) is 
injected into each varix. Complications 
include bleeding ulcers, dysphagia due to 
strictures, and pleural effusions. 

b. Endoscopic variceal ligation with elastic 
bands is an alternative to sclerotherapy 
because of fewer complications and similar 
efficacy. 

c. If bleeding persists (or recurs within 48 hours 
of the initial episode) despite pharmacologic 
therapy and two endoscopic therapeutic 
attempts at least 24 hours apart, patients 
should be considered for salvage therapy 
with TIPS or surgical treatment (transection 
of esophageal varices and devascularization 
of the stomach, portacaval shunt, or liver 
transplantation). 

8. Transjugular intrahepatic portosystemic 



shunt (TIPS) consists of the angiographic 
creation of a shunt between hepatic and portal 
veins which is kept open by a fenestrated metal 
stent. It decompresses the portal system, 
controlling active variceal bleeding over 90% of 
the time. Complications include secondary 
bleeding, worsening encephalopathy in 20%, 
and stent thrombosis or stenosis. 
C. Secondary prophylaxis 

1.A patient who has survived an episode of 
variceal hemorrhage has an overall risk of 
rebleeding that approaches 70% at 1 year. 
2. Endoscopic sclerotherapy decreases the risk 
of rebleeding (50% versus 70%) and death 
(30% to 60% versus 50% to 75%). Endoscopic 
variceal ligation is superior to sclerotherapy. 
Banding is carried out every 2 to 3 weeks until 
obliteration. 
References: See page 112. 



Peptic Ulcer Disease 

Peptic ulcer disease is diagnosed in 500,000 patients 
each year. Patients with peptic ulcer disease should be 
treated as having an infectious illness caused by the 
bacterium Helicobacter pylori. Peptic ulcer disease due 
to H pylori infection can be cured with a combination of 
antimicrobial and antisecretory drugs. 

I. Pathophysiology 

A. Helicobacter pylori (HP), a spiral-shaped, 
flagellated organism, is the most frequent cause of 
peptic ulcer disease (PUD). Nonsteroidal anti- 
inflammatory drugs (NSAIDs) and pathologically 
high acid-secreting states (Zollinger-Ellison 
syndrome) are less common causes. More than 
90% of ulcers are associated with H. pylori. 
Eradication of the organism cures and prevents 
relapses of gastroduodenal ulcers. 

B. Complications of peptic ulcer disease include 
bleeding, duodenal or gastric perforation, and 
gastric outlet obstruction (due to inflammation or 
strictures). 

II. Clinical evaluation 

A. Symptoms of PUD include recurrent upper 
abdominal pain and discomfort. The pain of 
duodenal ulceration is often relieved by food and 
antacids and worsened when the stomach is empty 
(eg, at nighttime). In gastric ulceration, the pain 
may be exacerbated by eating. 

B. Nausea and vomiting are common in PUD. 
Hematemesis ("coffee ground" emesis) or melena 
(black tarry stools) are indicative of bleeding. 

C. Physical examination. Tenderness to deep 
palpation is often present in the epigastrium, and 
the stool is often guaiac-positive. 



Presentation of Uncomplicated Peptic Ulcer 
Disease 



Epigastric pain (burning, vague abdominal 

discomfort, nausea) 
Often nocturnal 

Occurs with hunger or hours after meals 
Usually temporarily relieved by meals or antacids 
Persistence or recurrence over months to years 
History of self-medication and intermittent relief 



D. NSAID-related gastrointestinal complications. 

NSAID use and H pylori infection are independent 
risk factors for peptic ulcer disease. The risk is 5 to 
20 times higher in persons who use NSAIDs than 
in the general population. Misoprostol (Cytotec) 
has been shown to prevent both NSAID ulcers and 
related complications. The minimum effective 
dosage is 200 micrograms twice daily; total daily 
doses of 600 micrograms or 800 micrograms are 
significantly more effective. 
III. Laboratory and diagnostic testing 

A. Alarm signs and symptoms that suggest gastric 
cancer are indications for early endoscopy or 
upper gastrointestinal radiology studies. 



Indications for early endoscopy 


Anorexia 


Presence of a mass 


Dysphagia 


Unexplained anemia 


Gastrointestinal bleeding 


Unexplained weight loss 


(gross or occult) 


Vomiting (severe) 


New-onset symptoms in 




persons >45 yr of age 





B. Noninvasive testing for H pylori 

1. In the absence of alarm symptoms for gastric 
cancer, most patients with dyspepsia should 
undergo evaluation for H pylori infection with 
serologic testing for H pylori antigens. 

2. Serologic testing to detect H pylori antibodies 
isthe preferred testing method. Serologic testing 
is highly sensitive, but it cannot be used for 
follow-up after therapy because antibody titers 
may remain elevated for a year or longer. Rapid 
office-based serologic kits have a sensitivity of 
90% and a specificity of 85%. 

3. Urea breath tests measure the carbon dioxide 
produced when H pylori urease metabolizes 
urea labeled with radioactive carbon (13C or 
14C). The 13C test does not involve a 
radioactive isotope and, unlike the 14C test, can 
be used in children and pregnant women. With 
the 13C test, exhaled breath samples are 
usually sent to a central testing facility. The 1 4C 
test, which exposes the patient to a small dose 
of radiation, can be analyzed in a hospital's 
nuclear medicine laboratory. Urea breath tests 
have a sensitivity and specificity of 90-99%. The 
urea breath test is the best method of 
confirmation of care. 

4. Stool testing for H pylori antigens has an 
accuracy for pretreatment testing of H pylori that 
is similar to that of other available tests. 

5. Biopsy-based testing performed at endoscopy 
can provide valuable information via histologic 
testing, rapid urease tests, and culture. 
Sensitivity of the tests for H pylori ranges from 
80% to 100%, and specificity exceeds 95%. 



Triple therapies for peptic ulcer disease 



BMT therapy: 

Bismuth subsalicylate (Pepto-Bismol), 2 tablets with meals 

and at bedtime for 14 days 

and 

Metronidazole (Flagyl), 250 mg with meals and at bedtime 

(total daily dose, 1 ,000 mg) for 14 days 

and 

Tetracycline, 500 mg with meals and at bedtime (total daily 

dose, 2 g) for 1 4 days 

or 

A prepackaged triple-therapy agent (Helidac), to be taken 

qid for 14 days, consists of 525 mg bismuth subsalicylate, 

250 mg metronidazole, and 500 mg tetracycline; an H 2 - 

blocker or proton pump inhibitor should be added 

(Omeprazole [Prilosec], 20 mg qd or lansoprazole [Prevacid], 

15 mg qd). 



Ranitidine bismuth citrate (Tritec), 1 tablet (400 mg) bid for 

14 days 

and 

Tetracycline, 500 mg bid for 14 daysandClarithromycin 

(Biaxin) or metronidazole (Flagyl), 500 mg bid for 14 days 



Omeprazoie (Prilosec), 20 mg bid, or lansoprazole 

(Prevacid), 30 mg bid 

and 

Clarithromycin (Biaxin), 250 or 500 mg bid for 14 days 

and 

Metronidazole (Flagyl), 500 mg bid, or amoxicillin, 1 g bid for 

14 days 

or 

A prepackaged triple-therapy agent (Prevpac), to be 

taken bid for 14 days, consists of 30 mg lansoprazole, 1 g 

amoxicillin, and 500 mg clarithromycin. 



IV. Treatment of peptic ulcer disease 

A. Combination therapy 

1 . Dual therapy is not recommended because cure 
rates for all regimens are less than 85%. 
Recommended triple therapies consist of a 
bismuth preparation or proton pump inhibitor or 
H2 receptor antagonist plus two antibiotics. 

2. The H pylori eradication rate is 96% for patients 
who take more than 60% of their medication. 

B. Confirmation of cure of H pylori infection 

1 . Confirmation of cure of H pylori infection is 
always necessary. About 75% of patients 
presumed to have uncomplicated peptic ulcer 
disease due to H pylori infection are cured after 
one course of therapy. 

2. The urea breath test is the best method for 
assessing the effectiveness of therapy. The 
stool antigen test is only slightly less accurate, 
and its use should be considered when breath 
testing is not available. 

3. Confirmation of cure must be delayed until at 
least 4 to 6 weeks after completion of 
antimicrobial therapy. Treatment with proton 
pump inhibitors must be discontinued at least 1 
week before urea breath testing to confirm cure. 
H2-receptor antagonists have no effect on the 
urea breath test and need not be discontinued 



before confirmation testing. 
C. Treatment of NSAID-related ulcers 

1 . When the ulcer is caused by NSAID use, healing 
of the ulcer is greatly facilitated by discontinuing 
the NSAID. Acid antisecretory therapy with an 
H2-blocker or proton pump inhibitor speeds 
ulcer healing. Proton pump inhibitors are more 
effective in inhibiting gastric acid production and 
are often used to heal ulcers in patients who 
require continuing NSAID treatment. 

2. If serologic or endoscopic testing for H pylori is 
positive, antibiotic treatment is necessary. 

3. Acute H 2 -blocker therapy 

a. Ranitidine (Zantac), 150 mg bid or 300 mg 
qhs. 

b. Famotidine (Pepcid), 20 mg bid or 40 mg 
qhs. 

c. Nizatidine (Axid Pulvules), 150 mg bid or 
300 mg qhs. 

d. Cimetidine (Tagamet), 400 mg bid or 800 
mg qhs. 

4. Proton pump inhibitors 

a. Omeprazole (Prilosec), 20 mg qd. 

b. Lansoprazole (Prevacid), 15 mg before 
breakfast qd. 

c. Esomeprazole (Nexium) 20-40 mg qd. 
V. Surgical treatment of peptic ulcer disease 

A. Indications for surgery include exsanguinating 
hemorrhage, >5 units transfusion in 24 hours, 
rebleeding during same hospitalization, 
intractability, perforation, gastric outlet obstruction, 
and endoscopic signs of rebleeding. 

B, Unstable patients should receive a truncal 
vagotomy, oversewing of bleeding ulcer bed, and 
pyloroplasty. 

References: See page 112. 



Lower Gastrointestinal Bleeding 

H.L. Daneschvar, WID 
S.E. Wilson, MD 

The spontaneous remission rates for lower 
gastrointestinal bleeding is 80 percent. No source of 
bleeding can be identified in 12 percent of patients, and 
bleeding is recurrent in 25 percent. Bleeding has usually 
ceased by the time the patient presents to the emergency 



Clinical evaluation 

A. The severity of blood loss and hemodynamic status 
should be assessed immediately. Initial 
management consists of resuscitation with 
crystalloid solutions (lactated Ringers) and blood 
products if necessary. 

B. The duration and quantity of bleeding should be 
assessed; however, the duration of bleeding is 
often underestimated. 

C. Risk factors that may have contributed to the 
bleeding include nonsteroidal anti-inflammatory 
drugs, anticoagulants, colonic diverticulitis, renal 
failure, coagulopathy, colonic polyps, and 
hemorrhoids. Patients may have a prior history of 
hemorrhoids, diverticulosis, inflammatory bowel 
disease, peptic ulcer, gastritis, cirrhosis, or 
esophageal varices. 

D. Hematochezia. Bright red or maroon output per 
rectum suggests a lower Gl source; however, 12 to 
20% of patients with an upper Gl bleed may have 
hematochezia as a result of rapid blood loss. 

E. Melena. Sticky, black, foul-smelling stools suggest 
a source proximal to the ligament of Treitz, but 
Melena can also result from bleeding in the small 
intestine or proximal colon. 

F. Clinical findings 

1. Abdominal pain may result from ischemic 
bowel, inflammatory bowel disease, or a 
ruptured aneurysm. 

2. Painless massive bleeding suggests vascular 
bleeding from diverticula, angiodysplasia, or 
hemorrhoids. 

3. Bloody diarrhea suggests inflammatory bowel 
disease or an infectious origin. 

4. Bleeding with rectal pain is seen with anal 
fissures, hemorrhoids, and rectal ulcers. 

5. Chronic constipation suggests hemorrhoidal 
bleeding. Newonset of constipation orthin stools 
suggests a left sided colonic malignancy. 

6. Blood on the toilet paper or dripping into the 
toilet water suggests a perianal source of 
bleeding, such as hemorrhoids or an anal 
fissure. 

7. Blood coating the outside of stools suggests a 
lesion in the anal canal. 



8. Blood streaking or mixed in with the stool may 
results from polyps or a malignancy in the 
descending colon. 

9. Maroon colored stools often indicate small 
bowel and proximal colon bleeding. 

II. Physical examination 

A. Postural hypotension indicates a 20% blood 
volume loss, whereas, overt signs of shock (pallor, 
hypotension, tachycardia) indicates a 30 to 40 
percent blood loss. 

B. The skin may be cool and pale with delayed refill if 
bleeding has been significant. 

C. Stigmata of liver disease, including jaundice, 
caput medusae, gynecomastia and palmar 
erythema, should be sought because patients with 
these findings frequently have Gl bleeding. 

III. Differential diagnosis of lower Gl bleeding 

A. Angiodysplasia and diverticular disease of the 
right colon accounts for the vast majority of 
episodes of acute lower Gl bleeding. Most acute 
lower Gl bleeding originates from the colon 
however 1 5 to 20 percent of episodes arise from 
the small intestine and the upper Gl tract. 

B. Elderly patients. Diverticulosisand angiodysplasia 
are the most common causes of lower Gl bleeding. 

C. Younger patients. Hemorrhoids, anal fissures and 
inflammatory bowel disease are most common 
causes of lower Gl bleeding. 



Clinical Indicators of Gastrointestinal Bleeding 
and Probable Source 


Clinical Indicator 


Probability of 
Upper 

Gastrointestinal 
source 


Probability of 
Lower 

Gastrointestinal 
Source 


Hematemesis 


Almost certain 


Rare 


Melena 


Probable 


Possible 


Hematochezia 


Possible 


Probable 


Blood-streaked 
stool 


Rare 


Almost certain 


Occult blood in 
stool 


Possible 


Possible 



IV. Diagnosis and management of lower 
gastrointestinal bleeding 

A. Rapid clinical evaluation and resuscitation 

should precede diagnostic studies. Intravenous 
fluids (1 to 2 liters) should be infused over 10- 20 
minutes to restore intravascular volume, and blood 
should be transfused if there is rapid ongoing blood 
loss or if hypotension or tachycardia are present. 
Coagulopathy is corrected with fresh frozen plasma, 
platelets, and cryoprecipitate. 

B. When small amounts of bright red blood are passed 
per rectum, then lower Gl tract can be assumed to 
be the source. In patients with large volume maroon 
stools, nasogastric tube aspiration should be 
performed to exclude massive upper 
gastrointestinal hemorrhage. 

C. If the nasogastric aspirate contains no blood then 
anoscopy and sigmoidoscopy should be performed 
to determine weather a colonic mucosal 
abnormality (ischemic or infectious colitis) or 
hemorrhoids might be the cause of bleeding. 

D. Colonoscopy in a patient with massive lower Gl 
bleeding is often nondiagnostic, but it can detect 
ulcerative colitis, antibiotic-associated colitis, or 
ischemic colon. 

E. Polyethylene glycol-electrolyte solution (CoLyte 
or GoLytely) should be administered by means of a 
nasogastric tube (Four liters of solution is given 
over a 2-3 hour period), allowing for diagnostic and 
therapeutic colonoscopy. 

V. Definitive management of lower gastrointestinal 
bleeding 

A. Colonoscopy 

1. Colonoscopy is the procedure of choice for 
diagnosing colonic causes of Gl bleeding. It 
should be performed after adequate preparation 
of the bowel. If the bowel cannot be adequately 
prepared because of persistent, acute bleeding, 
a bleeding scan or angiography is preferable. 

2. If colonoscopy fails to reveal the source of the 
bleeding, the patient should be observed 
because, in 80% of cases, bleeding ceases 
spontaneously. 

B. Radionuclide scan or bleeding scan. 
Technetium- labeled (tagged) red blood cell 
bleeding scans can detect bleeding sites when 
bleeding is intermittent. Localization may not he a 



precise enough to allow segmental colon resection. 

C. Angiography. Selective mesenteric angiography 
detects arterial bleeding that occurs at rates of 0.5 
mL/per minute or faster. Diverticular bleeding 
causes pooling of contrast medium within a 
diverticulum. Bleeding angiodysplastic lesions 
appear as abnormal vasculature. When active 
bleeding is seen with diverticular disease or 
angiodysplasia, selective arterial infusion of 
vasopressin may be effective. 

D. Surgery 

1. If bleeding continues and no source can be 
found, surgical intervention is usually warranted. 
Surgical resection may be indicated for patients 
with recurrent diverticular bleeding, or for 
patients who have had persistent bleeding from 
colonic angiodysplasia and have required blood 
transfusions. 

2. Surgical management of lower gastrointestinal 
bleeding is ideally undertaken with a secure 
knowledge of the location and cause of the 
bleeding lesion. A segmental bowel resection to 
include the lesion and followed by a primary 
anastomosis is usually safe and appropriate in 
all but the most unstable patients. 

VI.Diverticulosis 

A. Diverticulosis of the colon is present in more than 
50% of the population by age 60 years. Bleeding 
from diverticula is relatively rare, affecting only 4% 
to 1 7% of patients at risk. 

B. In most cases, bleeding ceases spontaneously, but 
in 10% to 20% of cases, the bleeding continues. 
The risk of rebleeding after an episode of bleeding 
is 25%. Right-sided colonic diverticula occur less 
frequently than left-sided or sigmoid diverticula but 
are responsible for a disproportionate incidence of 
diverticular bleeding. 

C. Operative management of diverticular bleeding is 
indicated when bleeding continues and is not 
amenable to angiographic or endoscopic therapy. 
It also should be considered in patients with 
recurrent bleeding in the same colonic segment. 
The operation usually consists of a segmental 
bowel resection (usually a right colectomy or 
sigmoid colectomy) followed by a primary 
anastomosis. 

VII. Arteriovenous malformations 

A. AVMs or angiodysplasias are vascular lesions that 
occur primarily in the distal ileum, cecum, and 
ascending colon of elderly patients. The 
arteriographic criteria for identification of an AVM 
include a cluster of small arteries, visualization of a 
vascular tuft, and early and prolonged filling of the 
draining vein. 

B. The typical pattern of bleeding of an AVM is 
recurrent and episodic, with most individual 
bleeding episodes being self-limited. Anemia is 
frequent, and continued massive bleeding is 
distinctly uncommon. After nondiagnostic 
colonoscopy, enteroscopy should be considered. 

C. Endoscopic therapy for AVMs may include heater 
probe, laser, bipolar electrocoagulation, or argon 
beam coagulation. Operative management is 
usually reserved for patients with continued 
bleeding, anemia, repetitive transfusion 
requirements, and failure of endoscopic 
management. Surgical management consists of 
segmental bowel resection with primary 
anastomosis. 

VIII. Inflammatory bowel disease 

A. Ulcerative colitis and, less frequently, Crohn'scolitis 
or enteritis may present with major or massive 
lower gastrointestinal bleeding. Infectious colitis can 
also manifest with bleeding, although it is rarely 
massive. 

B. When the bleeding is minor to moderate, therapy 
directed at the inflammatory condition is 
appropriate. When the bleeding is major and 
causes hemodynamic instability, surgical 
intervention is usually required. When operative 
intervention is indicated, the patient is explored 
through a midline laparotomy, and a total abdominal 
colectomy with end ileostomy and oversewing of 
the distal rectal stump is the preferred procedure. 



IX. Tumors of the colon and rectum 

A. Colon and rectal tumors account for 5% to 10% of 
all hospitalizations for lower gastrointestinal 
bleeding. Visible bleeding from a benign colonic or 
rectal polyp is distinctly unusual. Major or massive 
hemorrhage rarely is caused by a colorectal 
neoplasm; however, chronic bleeding is common. 
When the neoplasm is in the right colon, bleeding is 
often occult and manifests as weakness or anemia. 

B. More distal neoplasms are often initially confused 
with hemorrhoidal bleeding. For this reason, the 
treatment of hemorrhoids should always be 
preceded by flexible sigmoidoscopy in patients 
older than age 40 or 50 years. In younger patients, 
treatment of hemorrhoids without further 
investigation may be appropriate if there are no risk 
factors for neoplasm, there is a consistent clinical 
history, and there is anoscopic evidence of recent 
bleeding from enlarged internal hemorrhoids. 

X. Anorectal disease 

A. When bleeding occurs only with bowel movements 
and is visible on the toilet tissue or the surface of 
the stool, it is designated outlet bleeding. Outlet 
bleeding is most often associated with internal 
hemorrhoids or anal fissures. 

B. Anal fissures are most commonly seen in young 
patients and are associated with severe pain during 
and after defecation. Other benign anorectal 
bleeding sources are proctitis secondary to 
inflammatory bowel disease, infection, or radiation 
injury. Additionally, stercoral ulcers can develop in 
patients with chronic constipation. 

C. Surgery for anorectal problems is typically 
undertaken only after failure of conservative 
medical therapy with high-fiber diets, stool 
softeners, and/or hemorrhoidectomy. 

XI. Ischemic colitis 

A. Ischemic colitis is seen in elderly patients with 
known vascular disease. The abdomen pain may 
be postprandial and associated with bloody 
diarrhea or rectal bleeding. Severe blood loss is 
unusual but can occur. 

B. Abdominal films may reveal "thumb-printing" 
caused by submucosal edema. Colonoscopy 
reveals a well-demarcated area of hyperemia, 
edema and mucosal ulcerations. The splenic 
flexure and descending colon are the most common 
sites. Most episodes resolve spontaneously, 
however, vascular bypass or resection may be 
required. 

References: See page 112. 



Anorectal Disorders 

I. Hemorrhoids 

A. Hemorrhoids are dilated veins located beneath the 
lining of the anal canal. Internal hemorrhoids are 
located in the upper anal canal. External 
hemorrhoids are located in the lower anal canal. 

B. The most common symptom of internal hemor- 
rhoids is painless rectal bleeding, which is usually 
bright red and ranges from a few drops to a spatter- 
ing stream at the end of defecation. If internal 
hemorrhoids remain prolapsed, a dull aching may 
occur. Blood and mucus stains may appear on 
underwear, and itching in the perianal region is 
common. 



Classification of Internal Hemorrhoids 


Gr 

ad 
e 


Description 


Symptoms 


1 


Non-prolapsing 


Minimal bleeding 


2 


Prolapse with straining, 
reduce when spontan- 
eously prolapsed 


Bleeding, discomfort, 
pruritus 


3 


Prolapse with straining, 
manual reduction 
required when 
prolapsed 


Bleeding, discomfort, 
pruritus 


4 


Cannot be reduced 
when prolapsed 


Bleeding, discomfort, 
pruritus 



C. Management of internal hemorrhoids 

1. Grade 1 and uncomplicated grade 2 
hemorrhoids are treated with dietary 
modification (increased fiber and fluids). 

2. Symptomatic grade 2 and grade 3 
hemorrhoids. Treatment consists of 



hemorrhoid banding with an anoscope. Major 
complications are rare and consist of excessive 
pain, bleeding, and infection. Surgical 
hemorrhoidectomy may sometimes be 
necessary. 
3. Grade 4 hemorrhoids require surgical 
hemorrhoidectomy. 
D. External hemorrhoids 

1 . External hemorrhoids occur most often in young 
and middle-aged adults, becoming symptomatic 
only when they become thrombosed. 

2. External hemorrhoids are characterized by rapid 
onset of constant burning or throbbing pain, 
accompanying a new rectal lump. Bluish skin- 
covered lumps are visible at the anal verge. 

3. Management of external hemorrhoids 

a. If patients are seen in the first 48 hours, the 
entire lesion can be excised in the office. Local 
anesthetic is infiltrated, and the thrombus and 
overlying skin are excised with scissors. The 
resulting wound heals by secondary intention. 

b. If thrombosis occurred more than 48 hours 
prior, spontaneous resolution should be 
permitted to occur. 

II. Anal fissures 

A. An anal fissure is a longitudinal tear in the distal 
anal canal, usually in the posterior or anterior 
midline. Patients with anal fissures complain of 
perirectal pain which is sharp, searing or burning 
and is associated with defecation. Bleeding from 
anal fissures is bright red and not mixed with the 
stool. 

B. Anal fissures may be associated with secondary 
changes such as a sentinel tag, hypertrophied anal 
papilla, induration of the edge of the fissure, and 
anal stenosis. Crohn's disease should be 
considered if the patient has multiple fissures, or 
whose fissure is not in the midline. 

C. Anal fissures are caused by spasm of the internal 
anal sphincter. Risk factors include a low-fiber diet 
and previous anal surgery. 

D. Treatment of anal fissures 

1. High-fiber foods, warm sitz baths, stool 
softeners (if necessary), and daily application of 
1% hydrocortisone cream to the fissure should 
be initiated. These simple measures may heal 
acute anal fissures within 3 weeks in 90% of 
patients. 

2. Lateral partial internal sphincterotomy is 
indicated when 4 weeks of medical therapy fails. 
The procedure consists of surgical division of a 
portion of the internal sphincter, and it is highly 
effective. Adverse effects include incontinence 
to flatus and stool. 

III. Levator ani syndrome and proctalgia fugax 

A. Levator ani syndrome refers to chronic or recurrent 
rectal pain, with episodes lasting 20 minutes or 
longer. Proctalgia fugax is characterized by anal or 
rectal pain, lasting for seconds to minutes and then 
disappearing for days to months. 

B. Levator ani syndrome and proctalgia fugax are 
more common in patients under age 45, and 
psychological factors are not always present. 

C. Levator ani syndrome is caused by chronic tension 
of the levator muscle. Proctalgia fugax is caused 
by rectal muscle spasm. Stressful events may 
trigger attacks of proctalgia fugax and levator ani 
syndrome. 

D. Diagnosis and clinical features 

1. Levator ani syndrome is characterized by a 
vague, indefinite rectal discomfort or pain. The 
pain is felt high in the rectum and is sometimes 
associated with a sensation of pressure. 

2. Proctalgia fugax causes pain that is brief and 
self limited. Patients with proctalgia fugax 
complain of sudden onset of intense, sharp, 
stabbing or cramping pain in the anorectum. 

3. In patients with levator ani syndrome, palpation 
of the levator muscle during digital rectal 
examination usually reproduces the pain. 

E. Treatment 

1. Levator ani syndrome. Treatment with hot 
baths, nonsteroidal anti-inflammatory drugs, 
muscle relaxants, or levator muscle massage is 
recommended. EMG-based biofeedback may 
provide improvement in pain. 

2. Proctalgia fugax. For patients with frequent 
attacks, physical modalities such as hot packs 
or direct anal pressure with a finger or closed 
fist may alleviate the pain. Diltiazem and 
clonidine may provided relief. 

IV. Pruritus ani 

A. Pruritus ani is characterized by the intense desire 
to scratch the skin around the anal orifice. It occurs 
in 1 % of the population. Pruritus ani may be related 



to fecal leakage. 

B. Patients report an escalating pattern of itching and 
scratching in the perianal region. These symptoms 
may be worse at night. Anal hygiene and dietary 
habits, fecal soiling, and associated medical 
conditions should be sought. 

C. Examination reveals perianal maceration, 
erythema, excoriation, and lichenification. A digital 
rectal examination and anoscopy should be 
performed to assess the sphincter tone and look 
for secondary causes of pruritus. Patients who fail 
to respond to 3 or 4 weeks of conservative 
treatment should undergo further investigations 
such as skin biopsy and sigmoidoscopy or 
colonoscopy. 

D. Treatment and patient education 

1. Patients should clean the perianal area with 
water following defecation, but avoid soaps and 
vigorous rubbing. Following this, the patient 
should dry the anus with a hair dryer or by 
patting gently with cotton. Between bowel 
movements a thin cotton pledget dusted with 
unscented cornstarch should be placed against 
the anus. A high fiber diet is recommended to 
regulate bowel movements and absorb excess 
liquid. All foods and beverages that exacerbate 
the itching should be eliminated. 

2. Topical medications are not recommended 
because they may cause further irritation. If 
used, a bland cream such as zinc oxide or 1% 
hydrocortisone cream should be applied 
sparingly two to three times a day. 

3. Diphenhydramine (Benadryl) or hydroxyzine 
(Vistaril) may relieve the itching and allow the 
patient to sleep. 

. Perianal abscess 

A. The anal glands, located in the base of the anal 
crypts at the level of the dentate line, are the most 
common source of perianal infection. Acute infec- 
tion causes an abscess, and chronic infection 
results in a fistula. 

B. The most common symptoms of perianal abscess 
are swelling and pain. Fevers and chills may occur. 
Perianal abscess is common in diabetic and 
immunosuppressed patients, and there is often a 
history of chronic constipation. A tender mass with 
fluctuant characteristics or induration is apparent 
on rectal exam. 

C. Management of perianal abscess. Perianal 
abscesses are treated with incision and drainage 
using a local anesthetic. Large abscesses require 
regional or general anesthesia. A cruciate incision 
is made close to the anal verge and the corners 
are excised to create an elliptical opening which 
promotes drainage. An antibiotic, such as Zosyn, 
Timentin, or Cefotetan, is administered. 

D. About half of patients with anorectal abscesses will 
develop a fistula tract between the anal glands and 
the perianal mucosa, known as a fistula-in-ano. 
This complication manifests as either incomplete 
healing of the drainage site or recurrence. Healing 
of a fistula-in-ano requires a surgical fistulotomy. 



Fistula-in-Ano 

A fistula-in-ano develops when an anorectal abscess 
forms a fistula between the anal canal and the perianal 
skin. The fistula may develop after an anorectal abscess 
has been drained operatively, or the fistula may develop 
spontaneously. 

I. Clinical evaluation 

A. The fistula is characterized by persistent purulent 
or feculent drainage, soiling the underwear. 

B. The fistula orifice can be seen just outside the anal 
verge. Complex fistulae may have multiple tracts 
with multiple orifices. 

II. Treatment of fistula-in-ano 

A. Fistulae will not resolvewithoutdefinitivetreatment. 
The more common type of fistula, located at the 
anorectal junction, has an external opening where 
it can be drained operatively. The entire 
epithelialized tract must be found and obliterated. 

B. Goodsall's rule predicts the course of fistulae that 
exit the skin within 3 cm of the anal verge. Anterior 
fistulae go straight toward the anorectal junction; 
posterior fistulae curve toward the posterior midline 
and enter the anorectal junction. 

C. A pilonidal cyst-sinus (coccygeal region) can be 
difficult to distinguish from a fistula-in-ano. Probing 
the tract under anesthesia usually will reveal its 
origin. 

D. Fistulotomy. For fistulae that do not cross both 
internal and external sphincters, the tract should be 



unroofed and curetted. The lesion should then be 
allowed to heal by secondary intention. 
. Fistulectomy. Deeper fistulae should be treated 
by coring out the epithelialized tract to its origin. 
The fistula may recur. 

Seton procedure. Complex fistulae or fistulae that 
traverse the sphincter can be treated by looping a 
heavy suture through the entire tract under tension. 
The suture should be tightened weekly until it 
"cuts" gradually to the surface. The tract will heal 
gradually behind the suture. 



Colorectal Cancer 

Charles Theuer, MD 

Colorectal cancer is the second most common solid 
malignancy in adults and the second leading cause of 
cancer death in the US. 

I. Clinical evaluation of colorectal cancer 

A. Flexible sigmoidoscopy is indicated for screening 
of asymptomatic, healthy adults over age 50. All 
adults with anemia or guaiac positive stools should 
be evaluated for colorectal cancer; older adults 
(>40) should be evaluated even if other sources of 
bleeding have been found. Hemorrhoids and 
cancer can coexist. 

B. Flexible sigmoidoscopy plus air contrast barium 
enema is adequate to evaluate the colon when the 
source of bleeding is thought to be benign 
anorectal disease. Total colonoscopy should be 
performed for any adult with gross or occult rectal 
bleeding and no apparent anorectal source. 

C. Left-sided or rectal lesions are characterized by 
blood streaked stools, change in caliber or 
consistency of stools, obstipation, alternating 
diarrhea and constipation, and tenesmus. 

D. Right-sided lesions are characterized by a triad of 
iron deficiency anemia, right lower quadrant mass, 
and weakness. Cancers occasionally present as a 
large bowel obstruction, perforation or abscess. 

II. Laboratory evaluation 

A. Complete blood count with indices will often reveal 
a hypochromic, microcytic anemia. Liver function 
tests may sometimes be elevated in metastatic 
disease. 

B. Carcinoembryonic antigen (CEA) may be 
elevated in colorectal cancer, but it is a nonspecific 
test which may also be elevated in other 
malignancies, inflammatory bowel disease, 
cigarette smokers, and some normal persons. CEA 
is valuable in monitoring the response to treatment 
and as a marker for recurrence or metastases, 
requiring adjuvant therapy. It should be measured 
prior to resection of the tumor and at intervals 
postoperatively. 

C. Colorectal cancer is detected by colonoscopy with 
biopsies. Barium enema may complement 
colonoscopy since BE shows the exact anatomic 
location of the tumor. A chest X-ray should be done 
to search for metastases to the lungs. A CT scan 
should be done in cases where liver function test 
are elevated. 

III. Management of colorectal carcinoma 

A. Surgical resection is indicated for colorectal 
adenocarcinoma, regardless of stage. Resection of 
the primary lesion prevents obstruction or 
perforation. 

B. Extremely advanced rectal lesions, which are not 
resectable, may be candidates for palliative 
radiation and a diverting colostomy. 

C. The extent of resection is determined by the 
relationship of the lesion to the lymphatic drainage 
and blood supply of the colon. 

1. Cecum or right colon. Right hemicolectomy. 

2. Hepatic flexure. Extended right hemicolectomy. 

3. Mid-transverse colon: Transverse colectomy 
or extended left or right hemicolectomy. 

4. Splenic flexure or left colon. Left 
hemicolectomy. 

5. Sigmoid colon. Sigmoid colectomy. 

6. Upper or middle rectum. Low anterior 
rectosigmoid resection with primary 
anastomosis. 

7. Lower rectum. Abdominoperineal resection 
with permanent, end-colostomy or local 
excision. 

D. Preoperative bowel preparation 

1. Mechanical cleansing of the lumen, followed by 
decontamination with nonabsorbable oral 
antibiotics decreases the chance of infectious 
complications and allows for primary 



anastomosis. Fully obstructed patients cannot 
be prepped and must have a temporary 
colostomy. 

2. Polyethylene glycol solution (CoLyte or 
GoLYTELY) is usually administered as 4 liters 
over 4 hours on the day before surgery. Oral 
phospho-soda (given as two 11/2 ounce doses 
in 8 ounces of water) can be substituted for 
polyethylene glycol in patients with normal renal 
function. The Nichols-Condon prep consists of 
1 g neomycin sulfate and 1 g erythromycin base 
PO at 2:00, 3:00 and 1 1 :00 pm the day before 
operation. Cefotetan is given 1-2 gm IV 30 
minutes before operation. 

3. Patients with middle and lower rectal tumors 
should be staged with endoanal ultrasound. 
Tumors that invade through the muscularis 
propria (T3) or involve lymph nodes (N1) should 
be offered neoadjuvant therapy with radiation 
therapy and 5-fluorouracil. 

E. Adjuvant chemotherapy is recommended for 
advanced colon lesions with the addition of pelvic 
radiation for advanced rectal tumors. Adjuvant 
therapy is reserved for locally advanced lesions 
(B2) or those with metastases to regional lymph 
nodes or distant organs (C1 , C2, D). 

F. Pathologic staging of the tumor is done 
postoperatively by histologic examination of the 
surgical specimen. 

IV. Staging of colorectal carcinoma 

A. Astler-Coller modification of Dukes' 
Classification 

Stage A: Limited to mucosa and submucosa. 
Nodes negative. 
Stage B1: Extends into, but not through, 

muscularis propria; nodes negative. 
Stage B2: Extends through muscularis propria; 
nodes negative. 

Stage C1 : Same as B1 , except nodes positive. 
Stage C2: Same as B2, except nodes positive. 

V. Management of obstructing carcinomas of the left 
colon 

A. Correct fluid deficits and electrolyte abnormalities. 
Nasogastric suction is useful, but it is not adequate 
to decompress the acutely obstructed colon. 

B. The Hartmann procedure is indicated for distal 
descending and sigmoid colon lesions. This 
procedure consists of resection of the obstructing 
cancer and formation of an end-colostomy and 
blind rectal pouch. The colostomy can be taken 
down and anastomosed to the rectal pouch at a 
later date. 

C. Primary resection with temporary end-colostomy 
and mucous fistula should be done for lesions of 
the transverse and proximal descending colon. 
This procedure consists of resection of the 
obstructing cancer and creation of a functioning 
end-colostomy and a defunctionalized distal limb 
with separate stomas. The colostomy can be taken 
down and continuity restored at a later date. 

D. An emergency decompressive loop colostomy can 
be considered for acutely ill patients. After four to 
six weeks, a hemicolectomy can be completed. A 
primary anastomosis may be done in selected 
patients with a prepared bowel. 

VI. Management of obstructing carcinomas of the 
ascending colon. Correct fluid deficits, electrolyte 
abnormalities, and initiate nasogastric suction. A right 
hemicolectomy with primary anastomosis of the 
terminal ileum to the transverse colon can be 
performed on most patients. A temporary ileostomy is 
rarely needed. 

References: See page 112. 



Mesenteric Ischemia 

Mesenteric ischemia is classified as acute mesenteric 
ischemia (AMI) and chronic mesenteric ischemia (CMI). 
AMI is subdivided into occlusive and nonocclusive 
mesenteric ischemia. Occlusive mesenteric ischemia 
results from either thrombotic or embolic arterial or 
venous occlusion. 

Approximately 80% of cases of AMI are occlusive in 
etiology, with arterial emboli or thromboses in 65% of 
cases and venousthrombosis in 15%. Arterial occlusions 
result from emboli in 75% of patients and in situ 
thrombosis cause the remaining 25%. Nonocclusive 
mesenteric ischemia is caused by low perfusion states 
and is responsible for 20% of AMI. 

I. Clinical evaluation 

A. Mesenteric arterial embolism 

1. The median age of patients presenting with 
mesenteric arterial embolism is 70 years. The 



overwhelming majority of emboli lodge in the 
superior mesenteric artery (SMA). Emboli 
originating in the left atrium or ventricle are the 
most common cause of SMA embolism. 

2. Risk factors include advanced age, coronary 
artery disease, cardiac valvular disease, history 
of dysrhythmias, atrial fibrillation, post- 
myocardial infarction mural thrombi, history of 
thromboembolic events, aortic surgery, 
aortography, coronary angiography, and aortic 
dissection. A previous history of peripheral 
emboli is present in 20%. 

3. The disorder usually presents as sudden onset 
of severe poorly localized periumbilical pain, 
associated with nausea, vomiting, and frequent 
bowel movements. Pain is usually out of propor- 
tion to the physical findings and may be the only 
presenting symptom. 

4. The abdomen may be soft with only mild 
tenderness. Absent bowel sounds, abdominal 
distension or guarding are indicative of severe 
disease. 

5. Blood in the rectum is present in 16% of 
patients, and occult blood is present in 25% of 
patients. Peritoneal signs develop when the 
ischemic process becomes transmural. 

B. Mesenteric arterial thrombosis 

1. Thrombosis usually occurs in the area of 
atherosclerotic narrowing in the proximal SMA. 
The proximal jejunum through the distal 
transverse colon becomes ischemic. 

2. SMA thrombosis usually occurs in patients with 
chronic, severe, visceral atherosclerosis. A 
history of abdominal pain after meals is present 
in 20-50% of patients. Patients are often elderly, 
with coronary artery disease, severe peripheral 
vascular disease, or hypertension. 

3. SMA thrombosis presents with gradual onset of 
abdominal pain and distension. A history of 
postprandial abdominal pain and weight loss is 
present in half of cases. Pain is usually out of 
proportion to the physical findings, and nausea 
and vomiting are common. 

4. Signs of peripheral vascular disease, such as 
carotid, femoral or abdominal bruits, or 
decreased peripheral pulses are frequent. 
Abdominal distension, absent bowel sounds, 
guarding, rebound and localized tenderness, 
and rigidity indicate advanced bowel necrosis. 

II. Diagnostic evaluation of acute mesenteric 
ischemia 

A. Leukocyte count is elevated in most cases of 
mesenteric ischemia. In patients with SMA emboli, 
42% have a metabolic acidosis. The serum 
amylase is elevated in half of patients. 

B. Plain radiography. Abdominal and chest x-rays 
help to exclude the presence of free air or bowel 
obstruction. In rare instances, plain films of the 
abdomen reveal signs of ischemic bowel such as 
pneumatosis intestinalis, portal venous gas, or a 
thickened bowel wall with thumbprinting. However, 
plain films will be normal in the majority of cases. 

C. Angiography isthe gold standard for the diagnosis 
of AMI and is also used for therapeutic infusion of 
the vasodilator, papaverine. After obtaining plain 
abdominal films to rule out the presence of free air 
or obstruction, angiography must be obtained, 
especially in those patients in whom there is a 
strong clinical suspicion for AMI. 

III. Emergency management 

A. Stabilization and initial management 

1. Patients with significant hypotension require 
rapid fluid resuscitation, and vasopressors may 
be used. 

2. If hemoglobin is low, blood should be given. 
Patients who appear acutely ill should receive 
parenteral antibiotics to cover for gram-negative 
enteric bacteria as well as anaerobes after 
blood cultures are drawn. 

B. Papaverine 

1. Intraarterial infusion of papaverine into the 
superior mesenteric artery will increase 
mesenteric perfusion by relieving mesenteric 
vasoconstriction. 

2. Papaverine is started at angiography and 
continued postoperatively if laparotomy is 
performed. The dosing is 60 mg IV bolus, 
followed by a 30-60 mg/h continuous infusion at 
a concentration of 1 mg/mL. Papaverine 
improves survival by 20-50%. 

C. Acute mesenteric infarction with embolism 

1. Once embolism is confirmed at angiography, 
papaverine infusion is started, then laparotomy 
should be performed to evaluate bowel viability. 
Surgical intervention may involve arteriotomy 



with embolectomy and bowel resection if 
nonviable necrotic bowel is found. Postoperative 
anticoagulation is recommended for all patients. 
2. Patients without peritoneal signs with minor 
emboli, who achieve pain relief with vasodilator 
infusion, may be managed nonoperatively with 
repeated angiograms. 
D. Acute mesenteric infarction with thrombosis 

1. Acute mesenteric ischemia secondary to 
thrombosis is treated initially with a papaverine 
infusion started at angiography. Patients without 
peritoneal signs with minor thrombi may be 
treated with papaverine only. 

2. Patients with major thrombi with good collateral 
vasculature, without peritoneal signs, may be 
observed in the hospital without a papaverine 
infusion. Patients with peritoneal signs and 
documented thrombosis require laparotomy. 

References: See page 112. 



Intestinal Obstruction 

I. Clinical evaluation 

A. Intestinal obstruction is characterized by nausea, 
vomiting, cramps, and obstipation. Suspected 
intestinal obstruction requires immediate surgical 
consultation. 

B. Small-bowel obstruction. Auscultation may reveal 
high-pitched rushes or tinkles that coincide with 
episodes of cramping. Pain usually is epigastric or 
periumbilical. 

1. Proximal obstruction. Frequent non-bilious 
vomiting is prominent if the obstruction is 
proximal to the ampulla of Vater. Colicky pain 
occurs at frequent intervals (2-5 minutes). 
Obstipation may not occur until late, and 
distention is minimal or absent. 

2. Distal obstruction. Vomiting is bilious and less 
frequent. The vomiting may be feculent if the 
obstruction has been long-standing. Colicky pain 
occurs at intervals of 10 minutes or more, and it 
is less intense. A dull ache may persist between 
cramps. Distention increases gradually. 

C. Colonic obstruction 

1 . Colonic obstruction is caused by colon cancer in 
60-70% of cases, and diverticulitis and volvulus 
account for 30%. Obstruction is more common 
in the left colon than the right. 

2. Milder attacks of pain often occur in the weeks 
preceding the acute episode. Colic is perceived 
in the lower abdomen or suprapubically, and 
obstipation and distention are characteristic. 
Nausea is common, and vomiting may occur. 

3. Tenderness is usually mild in uncomplicated 
colonic obstruction. Rectal exam or 
sigmoidoscopy may detect an obstructing lesion. 

D. Colonic pseudo-obstruction (Ogilvie's 
syndrome) may occur in the elderly, bedridden or 
institutionalized individual, often after recent 
surgery. 

E. Strangulated obstruction is characterized by 
constant pain, fever, tachycardia, peritoneal signs, 
a tender abdominal mass, and leukocytosis. 

F. Laboratory evaluation of intestinal obstruction 

1. Hypokalemic alkalosis is the most common 
metabolic abnormality resulting from vomiting 
and fluid loss. Elevated BUN and creatinine 
suggests significant hypovolemia. 
Hypochloremic acidosis with increased anion 
gap may occur with strangulated obstruction. 

2. Leukocyte count frequently is normal in 
uncomplicated obstruction; however, 
leukocytosis suggests strangulation. 

3. Serum amylase may be elevated with bowel 
infarction. 

G. Radiography 

1. Plain films 

a. Small-bowel obstruction. Plain radiographs 
may demonstrate multiple air-fluid levels 
with dilated loops of small intestine, but no 
colonic gas. Proximal jejunal obstruction may 
not cause dilatation. Distal obstruction is 
characterized by a ladder pattern of dilated 
loops of bowel. 

b. Colonic obstruction. Obstructive lesions 
usually are located in the left colon and 
rectum and cause distention of the proximal 
colon. Dilated colon has a peripheral location 
within the abdomen, and haustral markings 
are present and valvulae conniventes are 
absent. 

2. Contrast studies 

a. Barium contrast enema. In colonic 
obstruction, a single-column contrast study 



with a water-soluble contrast enema should 
be performed. Contrast enema is 
contraindicated in obstruction from acute 
diverticulitis or in the presence of toxic 
megacolon, 
b. Upper Gl series. In acute small bowel 
obstruction, plain films are usually sufficient. 
An upper Gl series may be useful in partial 
small bowel obstruction. 
H. Endoscopy 

1 . Upper endoscopy is the best test in obstruction 
of the gastric outlet or duodenum. 

2. Colonoscopy can confirm the diagnosis of 
colon obstruction by cancer. Non-strangulated 
volvulus can often be reduced endoscopically, 
and elective resection can be completed at a 
later time. 

References: See page 112. 



Acute Pancreatitis 

Blanding U. Jones, MD and Russell A. Williams, MD 

The incidence of acute pancreatitis ranges from 54 to 
238 episodes per 1 million per year. Patients with mild 
pancreatitis respond well to conservative therapy, but 
those with severe pancreatitis may have a progressively 
downhill course to respiratory failure, sepsis, and death 
(less than 10%). 

I. Etiology 

A. Alcohol-induced pancreatitis. Consumption of 
large quantities of alcohol may cause acute 
pancreatitis. 

B. Cholelithiasis. Common bile duct or pancreatic 
duct obstruction by a stone may cause acute 
pancreatitis. (90% of all cases of pancreatitis occur 
secondary to alcohol consumption or cholelithiasis). 

C. Idiopathic pancreatitis. The cause of pancreatitis 
cannot be determined in 10 percent of patients. 

D. Hypertriglyceridemia. Elevation of serum 
triglycerides (>l,000mg/dL) has been linked with 
acute pancreatitis. 

E. Pancreatic duct disruption. In younger patients, 
a malformation of the pancreatic ducts (eg, 
pancreatic divisum) with subsequent obstruction is 
often the cause of pancreatitis. In older patients 
without an apparent underlying etiology, cancerous 
lesions of the ampulla of Vater, pancreas or 
duodenum must be ruled out as possible causes of 
obstructive pancreatitis. 

F. Iatrogenic pancreatitis. Radiocontrast studies of 
the hepatobiliary system (eg, cholangiogram, 
ERCP) can cause acute pancreatitis in 2-3% of 
patients undergoing studies. 

G. Trauma. Blunt or penetrating trauma of any kind to 
the peri-pancreatic or peri-hepatic regions may 
induce acute pancreatitis. Extensive surgical 
manipulation can also induce pancreatitis during 
laparotomy. 



Causes of Acute Pancreatitis 


Alcoholism 


Infections 


Cholelithiasis 


Microlithiasis 


Drugs 


Pancreas divisum 


Hypertriglyceridemia 


Trauma 


Idiopathic causes 




Medications Associated with Acute Pancreatitis 


Asparaginase (Elspar) 


Mercaptopurine 


Azathioprine (Imuran) 


(Purinethol) 


Didanosine (Videx) 


Pentamidine 


Estrogens 


Sulfonamides 


Ethacrynic acid 


Tetracyclines 


(Edecrin) 


Thiazide diuretics 


Furosemide (Lasix) 


Valproic acid 




(Depakote) 



Pathophysiology. Acute pancreatitis results when an 

initiating event causes the extrusion of zymogen 

granules, from pancreatic acinar cells, into the 

interstitium of the pancreas. Zymogen particles cause 

the activation of trypsinogen into trypsin. Trypsin 

causes auto-digestion of pancreatic tissues. 

. Clinical presentation 

A. Signs and symptoms. Pancreatitis usually 

presents with mid-epigastric pain that radiates to 

the back, associated with nausea and vomiting. 

The pain is sudden in onset, progressively 

increases in intensity, and becomes constant. The 



severity of pain often causes the patient to move 
continuously in search of a more comfortable 
position. 
B. Physical examination 

1. Patients with acute pancreatitis often appear 
very ill. Findings that suggest severe 
pancreatitis include hypotension and 
tachypnea with decreased basilar breath 
sounds. Flank ecchymoses (Grey Tuner's 
Sign) or periumbilical ecchymoses (Cullen's 
sign) may be indicative of hemorrhagic 
pancreatitis. 

2. Abdominal distension and tenderness in the 
epigastrium are common. Fever and 
tachycardia are often present. Guarding, 
rebound tenderness, and hypoactive or absent 
bowel sounds indicate peritoneal irritation. 
Deep palpation of abdominal organs should be 
avoided in the setting of suspected 
pancreatitis. 

IV. Laboratory testing 

A. Leukocytosis. An elevated WBC with a left shift 
and elevated hematocrit (indicating 
hemoconcentration) and hyperglycemia are 
common. Pre-renal azotemia may result from 
dehydration. Hypoalbuminemia, hyper- 
triglyceridemia, hypocalcemia, hyperbilirubinemia, 
and mild elevations of transaminases and alkaline 
phosphatase are common. 

B. Elevated amylase. An elevated amylase level 
often confirms the clinical diagnosis of 
pancreatitis. 

C. Elevated lipase. Lipase measurements are more 
specific for pancreatitis than amylase levels, but 
less sensitive. Hyperlipasemia may also occur in 
patients with renal failure, perforated ulcer 
disease, bowel infarction and bowel obstruction. 

D. Abdominal Radiographs may reveal non-specific 
findings of pancreatitis, such as "sentinel loops" 
(dilated loops of small bowel in the vicinity of the 
pancreas), ileus and, pancreatic calcifications. 

E. Ultrasonography demonstrates the entire 
pancreas in only 20 percent of patients with acute 
pancreatitis. Its greatest utility is in evaluation of 
patients with possible gallstone disease. 

F. Helical high resolution computed tomography 
is the imaging modality of choice in acute 
pancreatitis. CT findings will be normal in 14-29% 
of patients with mild pancreatitis. Pancreatic 
necrosis, pseudocysts and abscesses are readily 
detected by CT. 



Selected Conditions Other Than Pancreatitis 


Associated with Amylase Elevation 


Carcinoma of the 


Acute alcoholism 


pancreas 


Diabetic ketoacidosis 


Common bile duct 


Lung cancer 


obstruction 


Ovarian neoplasm 


Post-ERCP 


Renal failure 


Mesenteric infarction 


Ruptured ectopic pregnancy 


Pancreatic trauma 


Salivary gland infection 


Perforated vise us 


Macroamylasemia 


Renal failure 





Prognosis. Ranson's criteria is used to determine 
prognosis in acute pancreatitis. Patients with two or 
fewer risk factors have a mortality rate of less than 1 
percent, those with three or four risk-factors a 
mortality rate of 16 percent, five or six risk factors, a 
mortality rate of 40 percent, and seven or eight risk 
factors, a mortality rate approaching 100 percent. 



Ranson's Criteria for Acute Pancreatitis 



At admission 



During initial 48 hours 



1 . Age >55 years 

2. WBC>16,000/mm 3 

3. Blood glucose >200 
mg/dL 

4. Serum LDH >350 
IU/L 

5. AST >250 U/L 



1 . Hematocrit drop >1 0% 

2. BUN rise >5 mg/dL 

3. Arterial p0 2 <60 mm Hg 

4. Base deficit >4 mEq/L 

5. Serum calcium <8.0 mg/dL 

6. Estimated fluid 
sequestration >6 L 



VI. Treatment of pancreatitis 

A. Expectant management. Most cases of acute 
pancreatitis will improve within three to seven 
days. Management consists of prevention of 
complications of severe pancreatitis. 

B. NPO and bowel rest. Patients should take 
nothing by mouth. Total parenteral nutrition should 
be instituted for those patients fasting for more 
than five days. A nasogastric tube is warranted if 
vomiting or ileus. 



C. IV fluid resuscitation. Vigorous intravenous 
hydration is necessary. A decrease in urine output 
to less than 30 mL per hour is an indication of 
inadequate fluid replacement. 

D. Pain control. Morphine is discouraged because it 
may cause Oddi's sphincter spasm, which may 
exacerbate the pancreatitis. Meperidine 
(Demerol), 25-100 mg IV/IM q4-6h, is favored. 
Ketorolac (Toradol), 60 mg IM/IV, then 15-30 mg 
IM/IV q6h, is also used. 

E. Antibiotics. Routine use of antibiotics is not 
recommended in most cases of acute pancreatitis. 
In cases of infectious pancreatitis, treatment with 
cefoxitin (1-2 g IV q6h), cefotetan (1-2 g IV q12h), 
imipenem (1 .0 gm IV q6h), or ampicillin/sulbactam 
(1 .5-3.0 g IV q6h) may be appropriate. 

F. Alcohol withdrawal prophylaxis. Alcoholics may 
require alcohol withdrawal prophylaxis with 
lorazepam (Ativan) 1-2mg IM/IV q4-6h as needed 
x 3 days, thiamine 100mg IM/IV qd x 3 days, folic 
acid 1 mg IM/IV qd x 3 days, multivitamin qd. 

G. Octreotide. Somatostatin is also a potent inhibitor 
of pancreatic exocrine secretion. Octreotide is a 
somatostatin analogue, which has been effective 
in reducing mortality from bile-induced 
pancreatitis. Clinical trials, however, have failed to 
document a significant reduction in mortality 

H. Blood sugar monitoring and insulin 
administration. Serum glucose levels should be 
monitored. 
VII. Complications 

A. Chronic pancreatitis 

B. Severe hemorrhagic pancreatitis 

C. Pancreatic pseudocysts 

D. Infectious pancreatitis with development of sepsis 
(occurs in up to 5% of all patients with 
pancreatitis) 

E. Portal vein thrombosis 
References: See page 112. 



Acute Cholecystitis 

Russell A. Williams, MD 

Acute cholecystitis is a bacterial inflammation of the 
gallbladder which may cause severe peritonitis. 
Gallstones are present in the gallbladder in about 95% of 
cases. The incidence of acute calculous cholecystitis is 
higher in females, with a female-to-male ratio of 3:1 up to 
the age of 50 and a ratio of 1 .5:1 thereafter. 

I. Pathophysiology. Patients who have symptoms 
from gallstones have an elective cholecystectomy to 
avoid acute cholecystitis and its complications. Acute 
calculous cholecystitis is caused by obstruction of the 
cystic duct by a stone. Positive bacterial cultures of 
bile or gallbladder wall are found in 50% to 75% of 
cases. 

II. Clinical evaluation 

A. Persistent pain in the area of the gallbladder is 
present in almost every case. Frequently, the pain 
develops after ingestion of a meal. The pain is 
usually in the right upper quadrant, the 
epigastrium, or both. 

B. The pain often radiates toward the tip of the 
scapula. Pain in the right shoulder is present when 
the diaphragm is irritated by the inflammation. 

C. Nausea and vomiting occur in 60% to 70% of 
patients. 

III. Physical examination 

A. Fever is present in about 80% of patients. The 
most common and reliable finding on physical 
examination is tenderness in the right upper 
quadrant, the epigastrium, or both. About half of 
all patients have muscle rigidity in the right upper 
quadrant, and about one fourth have rebound 
tenderness. 

B. Murphy's sign, consisting of inspiratory arrest 
during deep palpation of the right upper quadrant, 
is not a consistent finding but is almost 
pathognomonic when present. A mass in the 
region of the gallbladder is palpable in about 40%. 

IV. Laboratory evaluation and imaging studies 

A. White blood cell count is elevated in 85% of 
cases. One half have elevation of the serum 
bilirubin, and the serum amylase is increased in 
one third. 



B. Radionuclide scan (HIDA scan). The specific 
test for acute cholecystitis is the HIDA scan. 
Normally, the scan outlines the liver and the 
extrahepatic biliary tract, including the gallbladder, 
and shows the nuclide flowing into the upper small 
intestine. In acute cholecystitis, the gallbladder is 
not seen on the scan. Radionuclide has a 
sensitivity of almost 100% and a specificity of 
95%. 

C. Ultrasound. Calculi within the gallbladder can be 
accurately detected by ultrasonography, but this 
test is not specificfor acute calculous cholecystitis. 
A thickened gallbladder wall and pericholecystic 
fluid are sometimes present. 

V. Differential diagnosis. Acute appendicitis, 
perforated or penetrating duodenal ulcer, acute or 
perforated gastric ulcer, and acute pancreatitis. In 
approximately 15% of cases of acute cholecystitis, 
the serum amylase is elevated, suggesting the 
possibility of acute pancreatitis. 

VI. Treatment 

A. Patients suspected of having acute cholecystitis 
should be hospitalized. Intravenous crystalloids 
should be given to restore intravascular volume. 
Preoperative management should include 
administration of an antibiotic that is effective 
against gram-positive and -negative aerobes and 
anaerobes. Those present most frequently are 
Escherichia coli, Klebsiella species, Streptococcus 
faecalis, Clostridium welchii, Proteus species, 
Enterobacter species, and anaerobic 
Streptococcus species. 

B. A second-generation cephalosporin is 
recommended for most cases of acute 
cholecystitis and reservation of the triple drug 
combination for patients who are seriously ill with 
sepsis. Antibiotic therapy should be initiated as 
soon as the diagnosis is made and should be 
continued for 24 hours postoperatively, unless 
peritonitis is severe, in which case it should 
continue for 7 days. 

1. Ampicillin 1-3.0 gm IVq6h OR 

2. Ampicillin-sulbactam (Unasyn) 1 .5-3.0 gm IV 
q6h AND 

3. Gentamicin, 1 .5-2 mg/kg, then 2-5 mg/kg/d IV. 

4. Cefoxitin (Mefoxin) 1-2 gm IV q6-8h. 

5. Ticarcillin/clavulanate (Timentin) 3.1 g IV q4- 
6h. 

6. Piperacillin/tazobactam (Zosyn) 4.5 gm IV q6h. 

7. Meperidine (Demerol) 50-100 mg IV/IM q4-6h 
prn pain. 

C. The definitive treatment of acute cholecystitis is 
early laparoscopic cholecystectomy. Operative 
cholangiography is routinely performed unless the 
extent of inflammation makes it unsafe. 



Laparoscopic Cholecystectomy 
Procedure 

I. Advantages of laparoscopic surgery: Usually less 
postoperative pain, reduced recovery time; several 
small puncture wounds instead of a large surgical 
incision, and early return to work. 

II. Contraindications to laparoscopic 
cholecystectomy: Adhesions, extreme gallbladder 
scarring, severe acute inflammation, and bleeding. 

III. Technique 

A. Preoperative antibiotic therapy with cefoxitin 
(Mefoxin) 1-2 gm IV q6h is usually used routinely. 
The procedure is performed with the patient under 
general or epidural anesthesia. 

B. The stomach is decompressed with a nasogastric 
tube to facilitate exposure. With the patient in the 
supine position, a 2-cm incision is made superior 
or inferior to the umbilicus. Using S-shaped 
retractors, the fascia is identified and grasped with 
a small Kocher or Allis clamp. The fascia is 
elevated and incised to allow for easy admission 
of a finger to confirm entrance into the abdominal 
cavity and sweep away any adhesions. A U-stitch 
is placed using an absorbable suture. The Hasson 
cannula is inserted and secured with the suture 
used for the U-stitch. 

C. After setting the insufflator to an insufflation 
pressure of 12 mm Hg, C0 2 is instilled at a low 
flow (1 L/min) into the abdominal cavity through 
the Hasson cannula. Approximately 1 L C0 2 is 
instilled at a low flow rate, and then the flow rate is 
adjusted to the maximum (20 L/min). The 
endoscope is inserted, and the abdominal and 
pelvic cavities are inspected. 

D. The patient is placed in reverse Trendelenburg 
position to allow the colon and omentum to fall 



inferiorly. After the pelvis and upper abdomen are 
visually inspected, a 10-mm cannula is inserted 
two thirds of the way between the umbilicus and 
the xiphisternum just to the right of the midline. A 
5-mm cannula is inserted 3 cm inferior to the 
costal margin in the midclavicular line, and a 
second 5-mm cannula is inserted 4 cm inferior to 
the costal margin in the midaxillary line. All three 
cannulas are inserted using a trocar under direct 
endoscopic vision. The umbilical cannula is used 
for the endoscope and C02 inflow, and the 
epigastric port is used for dissection. Through the 
most lateral right subcostal cannula, a grasper 
retracts the dome of the gallbladder over the liver 
toward the right diaphragm. Through the other 
subcostal cannula, a grasper retracts the neck of 
the gallbladder laterally and anteriorly. 

E. Adhesions are dissected off of the gallbladder, and 
dissection is begun at the neck of the gallbladder 
and proceeds along the cystic duct. After the cystic 
duct and artery have been identified by removal of 
the peritoneum overlying these structures, a 
titanium clip is placed at the junction of the neck 
and cystic duct. A cholangiogram is performed by 
partially transecting the cystic duct using scissors. 
The cholangiocatheter is inserted into the 
cholecystodochotomy, and a cholangiogram is 
performed using 30% Renografin. 

F. If the cholangiogram is normal, the catheter is 
removed and the cystic duct is secured just inferior 
to the ductotomy with two titanium clips and 
divided. The cystic artery is clipped and divided. 
The infundibulum and neck of the gallbladder are 
rotated medially or laterally, and the peritoneal 
reflection onto the gallbladder is incised using the 
hook cautery. The gallbladder is dissected from its 
bed, and before the last attachments at the dome 
are divided, the gallbladder bed is irrigated and 
inspected for bleeding and bile leaks. The stumps 
of the cystic duct and artery are inspected for 
bleeding and bile leaks. 

G. When hemostasis is attained, the remaining 
attachments between the gallbladder and the liver 
are divided and the gallbladder is positioned just 
superior to the liver. The laparoscope and CO, 
insufflation tubing are transferred to the epigastric 
cannula, and the extraction sack is passed under 
direct visualization through the umbilical cannula. 
The sack is opened, and the gallbladder placed in 
the bag and extracted. 

H. The umbilical incision is closed under direct 
visualization by tying the U-stitch. The subcostal 
cannulas are removed under direct visualization. 
The epigastric cannula is positioned over the liver 
away from the omentum, CO, insufflation stopped, 
and residual CO, allowed to escape from the 
abdomen through the cannula. The cannula is 
removed, and the incisions are closed with a 
subcuticular stitch and sterile strips. Dressings are 
placed overthe incisions, and the nasogastric tube 
and Foley catheter are removed. The patient may 
be discharged after observation. Most patients can 
be discharged within a few hours. 



Open Cholecystectomy Procedure 

A. After induction of anesthesia place a nasogastric 
tube to decompress the stomach. The most 
commonly used incision is Kocher's right 
subcostal. Place incision 4 cm below and parallel 
to the costal margin, and extend it from the midline 
to the anterior axillary line. Open the anterior 
rectus sheath with a knife in the line of the incision. 
Divide the rectus muscle with cautery, and open 
the peritoneum between forceps. 

B. Systematically explore the peritoneal cavity and 
note the appearance of the hiatus, stomach, 
duodenum, liver, pancreas, intestines, and 
kidneys. Palpate the gallbladder from the ampulla 
towards the fundus, then palpate the common 
duct, noting any dilation or foreign bodies. 
Carefully palpate the colon for neoplasms. 

C. Grasp the gallbladder with a Rochester-Pean 
clamp near the fundus. Hold forceps in one hand, 
and introduce the right hand over the right lobe of 
the liver, permitting the liver to descend. Divide 
any adhesions to the omentum, colon or 
duodenum, and place a pack over these 
structures. Retract the structures inferiorly with a 
broad-bladed Deaver's retractor. 

D. Inspect the anatomy of the biliary tree by carefully 
dividing the peritoneum covering the anterior 
aspect of the cystic duct, and continue dissecting 
into the anterior layer of the lesser omentum 



overlying the common bile duct. Bluntly dissect 
with a dissector (Kitner), exposing Charcot's 
triangle bounded by the cystic duct, common bile 
duct and inferior border of the liver. The cystic 
artery should be seen in this triangle. Carefully 
observe the arrangement of the duct system and 
arterial supply. Do not divide any structure until the 
anatomy has been identified, including the cystic 
duct and common bile duct. 
Pass a ligature around the cystic duct with a right- 
angle clamp, and make a loose knot near the 
common duct. Partially divide the cystic duct below 
the infundibulum, and place a small polyethylene 
catheter attached to a syringe filled with saline into 
the cystic duct for 1-2 cm. Tighten the ligature 
holding the catheter in position. 
Attach a second syringe containing contrast 
material to the catheter, and remove all 
instruments. Place a sterile sheet, and slowly 
inject 10-15 cc of diluted dye into the common 
duct. An operative cholangiogram should be 
performed to detect stones and evaluate the duct 
system. 

Palpate the lower end of common bile duct, 
pancreas, and the foramen of Winslow. Palpate 
the ampulla, checking for stones or tumor. Hold 
the forceps on the gallbladder in the left hand, and 
clear the cystic artery of soft tissue with a pledget 
held in forceps. Follow the artery to the 
gallbladder, and clamp it with a right angle clamp. 
Divide and ligate the artery close to the edge of 
the gallbladder, using clips or 000 silk. 
Reaffirm the junction of the cystic duct with the 
common bile, then completely divide the exposed 
cystic duct, leaving a stump of 5 mm. 
Incise the peritoneum anteriorly over the 
gallbladder with a scalpel. Elevate the peritoneum 
from the gallbladder, and separate the gallbladder 
gently with sharp and blunt dissection. Tissue 
strands containing vessels should be cauterized 
before division. 

Inspect the gallbladder bed for bleeding and 
cauterize and/or ligate any bleeding areas. Control 
any persistent oozing from the bed with a small 
pack of hemostatic gauze. 
Irrigate the site with saline. If there is excessive 
fluid present, place a soft rubber Penrose drain or 
closed suction drain in the area of the dissection, 
and bring it out through a separate stab wound in 
the right upper quadrant. Inspect the operative 
field, including the ligatures on the arteries and the 
cystic duct. Approximate the peritoneum with 
continuous nonabsorbable suture. 
Irrigate the wound with saline and approximate the 
rectus fascia and fascia of the oblique muscles 
with interrupted, nonabsorbable sutures. Irrigate 
the subcutaneous space with saline, and close the 
skin with staples, or absorbable subcuticular 
sutures. 



Choledocholithiasis 

Choledocholithiasis results when gallstones pass from 
the gallbladder through the cystic duct into the common 
duct. 

I. Clinical evaluation 

A. Patients with choledocholithiasis generally present 
with jaundice . The patient may have pain or 
symptoms from associated biliary colic or acute 
cholecystitis. 

B. Physical examination. Icterus is typical unless 
there is associated acute cholecystitis. 
Ultrasonography can demonstrate gallstones in the 
gallbladder and in the common bile duct in 20-50% 
of patients with choledocholithiasis. 

C. The diagnosis depends on demonstrating enlarged 
common bile and intrahepatic ducts associated with 
abnormal liver function tests. 

D. Alkaline phosphatase and bilirubin are usually 
elevated. 

E. Ultrasound may reveal a dilated common bile duct, 
and stones may be seen. Frequently, gallstones in 
the lower common bile duct cannot be 
demonstrated by ultrasonography because of 
overlying bowel gas. 

F. Endoscopic retrograde cholangiopan- 
creatography (ERCP) or percutaneous trans- 
hepatic cholangiography (PTC) are often used to 
confirm the diagnosis. Theses tests can opacify the 
biliary tree and demonstrate intraductal stones. 

II. Management of choledocholithiasis 

A. All jaundiced patients and those known to have 
many, large, or intrahepatic stones should have 



preoperative ERCP to rule out malignancy and 

retrieve the stones. 

Preoperative ERCP with sphincterotomy should be 

performed in all patients who are at high risk of 

common bile duct stones or in whom common bile 

duct stones have been demonstrated. 

Laparoscopic cholecystectomy may be completed 

later. 

If ERCP is unsuccessful at clearing the common 

bile duct, the patient may require a laparoscopic or 

open cholecystectomy. 



Disorders of the Breast 

John A. Butler, MD 

Breast Cancer Screening and 
Diagnosis 



Breast cancer is the second most commonly diagnosed 
cancer among women, after skin cancer. Approximately 
182,800 new cases of invasive breast cancer are 
diagnosed in the United States per year. The incidence 
of breast cancer increases with age. White women are 
more likely to develop breast cancer than black women. 
The incidence of breast cancer in white women is about 
113 cases per 100,000 women and in black women, 100 
cases per 100,000. 

I. Risk Factors 



Risk Factors for Breast Cancer 


Age greater than 50 years 


Age greater than 30 at 


Prior history of breast 


first birth 


cancer 


Obesity 


Family history 


High socioeconomic 


Early menarche, before 


status 


age 12 


Atypical hyperplasia on 


Late menopause, after 


biopsy 


age 50 


Ionizing radiation 


Nulliparity 


exposure 



A. Family history is highly significant in a first-degree 
relative (ie, mother, sister, daughter), especially if 
the cancer has been diagnosed premenopausally. 
Women who have premenopausal first-degree 
relatives with breast cancer have a three- to 
fourfold increased risk of breast cancer. Having 
several second-degree relatives with breast cancer 
may further increase the risk of breast cancer. Most 
women with breast cancer have no identifiable risk 
factors. 

B. Approximately 8 percent of all cases of breast 
cancer are hereditary. About one-half of these 
cases are attributed to mutations in the BRCA1 and 
BRCA2 genes. Hereditary breast cancer commonly 
occurs in premenopausal women. Screening tests 
are available that detect BRCA mutations. 

II. Diagnosis and evaluation 

A. Clinical evaluation of a breast mass should 
assess duration of the lesion, associated pain, 
relationship to the menstrual cycle or exogenous 
hormone use, and change in size since discovery. 
The presence of nipple discharge and its character 
(bloody or tea-colored, unilateral or bilateral, 
spontaneous or expressed) should be assessed. 

B. Menstrual history. The date of last menstrual 
period, age of menarche, age of menopause or 
surgical removal of the ovaries, previous 
pregnancies should be determined. 

C. History of previous breast biopsies, cyst 
aspiration, dates and results of previous 
mammograms should be determined. 

D. Family history should document breast cancer in 
relatives and the age at which family members 
were diagnosed. 

III. Physical examination 

A. The breasts should be inspected for asymmetry, 
deformity, skin retraction, erythema, peau d'orange 
(breast edema), and nipple retraction, 
discoloration, or inversion. 

B. Palpation 

1 . The breasts should be palpated while the patient 
is sitting and then supine with the ipsilateral arm 
extended. The entire breast should be palpated 
systematically. The mass should be evaluated 
for size, shape, texture, tenderness, fixation to 
skin or chest wall. 

2. A mass that is suspicious for breast cancer is 
usually solitary, discrete and hard. In some 
instances, it is fixed to the skin or the muscle. A 
suspicious mass is usually unilateral and 
nontender. Sometimes, an area of thickening 
may represent cancer. Breast cancer is rarely 
bilateral. The nipples should be expressed for 
discharge. 

3. The axillae should be palpated for adenopathy, 
with an assessment of size of the lymph nodes, 
number, and fixation. 

IV. Mammography. Screening mammograms are 
recommended every year for asymptomatic women 
40 years and older. Unfortunately, only 60 percent of 
cancers are diagnosed at a local stage. 



Screening for Breast Cancer in Women 


Age 


American Cancer Society 
guidelines 


20 to 39 years 


Clinical breast examination every 

three years 

Monthly self-examination of breasts 


Age 40 years 
and older 


Annual mammogram 

Annual clinical breast examination 

Monthly self-examination of breasts 



V. Methods of breast biopsy 

A. Palpable masses. Fine-needle aspiration biopsy 
(FNAB) has a sensitivity ranging from 90-98%. 
Nondiagnostic aspirates require surgical biopsy. 

1. The skin is prepped with alcohol and the lesion 
is immobilized with the nonoperating hand. A 10 
mL syringe, with a 14 gauge needle, is 
introduced in to the central portion of the mass 
at a 90° angle. When the needle enters the 
mass, suction is applied by retracting the 
plunger, and the needle is advanced. The 
needle is directed into different areas of the 
mass while maintaining suction on the syringe. 

2. Suction is slowly released before the needle is 
withdrawn from the mass. The contents of the 
needle are placed onto glass slides for 
pathologic examination. 

3. Excisional biopsy is done when needle biopsies 
are negative but the mass is clinically suspected 
of malignancy. 

B. Stereotactic core needle biopsy. Using a 
computer-driven stereotactic unit, the lesion is 
localized in three dimensions, and an automated 
biopsy needle obtains samples. The sensitivity and 
specificity of this technique are 95-100% and 94- 
98%, respectively. 

C. Nonpalpable lesions 

1. Needle localized biopsy 

a. Under mammographic guidance, a needle 
and hookwire are placed into the breast 
parenchyma adjacent to the lesion. The 
patient is taken to the operating room along 
with mammograms for an excisional breast 
biopsy. 

b. The skin and underlying tissues are infiltrated 
with 1% lidocaine with epinephrine. For 
lesions located within 5 cm of the nipple, a 
periareolar incision may be used or use a 
curved incision located over the mass and 
parallel to the areola. Incise the skin and 
subcutaneous fat, then palpate the lesion and 
excise the mass. 

c. After removal of the specimen, a specimen x- 
ray is performed to confirm that the lesion 
has been removed. The specimen can then 
be sent fresh for pathologic analysis. 

d. Close the subcutaneous tissues with a 4-0 
chromic catgut suture, and close the skin with 
4-0 subcuticular suture. 

D. Ultrasonography. Screening is useful to 
differentiate between solid and cystic breast 
masses when a palpable mass is not well seen on 
a mammogram. Ultrasonography is especially 
helpful in young women with dense breast tissue 
when a palpable mass is not visualized on a 
mammogram. Ultrasonography is not used for 
routine screening because microcalcifications are 
not visualized and the yield of carcinomas is 
negligible. 

References: See page 112. 



Breast Cysts 

I. Clinical evaluation 

A. A breast cyst is palpable as a smooth, mobile, well- 
defined mass. If the cyst is tense, the texture may 
be very firm, resembling a cancer. Aspiration will 
determine whether the lesion is solid or cystic. 
Breast cyst fluid may vary from straw-colored to 
dark green. Cytology is not routinely necessary. 
The cyst should be aspirated completely. 

B. If a mass remains after drainage or if the fluid is 
bloody, excisional biopsy is indicated. If no pal- 
pable mass is felt after drainage, the patient should 
be reexamined in 3-4 weeks to determine whether 
the cyst recurs. Recurrent cysts can be re- 
aspirated. Repeated recurrence of the cyst requires 
an open biopsy to exclude intracystic tumor. 

C. Nonpalpable cysts. If the cyst wall is seen clearly 



seen on ultrasound and there is no interior debris 
or intracystic tumor, these simple cysts do not need 
to be aspirated. Any irregularity of the cyst wall or 
debris within the cyst requires a needle localized 
biopsy. 



Fibroadenomas 

I. Clinical evaluation 

A. Fibroadenomasfrequently present in young women 
as firm, smooth, lobulated masses that are highly 
mobile. They have a benign appearance on 
mammography and are solid by ultrasound. 

B. A tissue diagnosis can be obtained by fine needle 
aspiration biopsy or excisional biopsy. 

II. Management of fibroadenomas 

A. Fibroadenomas may be followed conservatively 
after the diagnosis has been made. If the mass 
grows, it should be excised. 

B. Large fibroadenomas (>2.5 cm) should usually be 
excised. Often fibroadenomas will grow in the 
presence of hormonal stimulation, such as 
pregnancy. 

References: See page 112. 



Breast Cancer 

The initial management of the breast cancer patient 
consists of assigning a clinical stage based on 
examination. The stage may be altered once the final 
pathology of the tumor has been determined. Staging of 
breast cancer is based on the TNM staging system. 

I. Preoperative staging in stage I and II breast 
cancer. Preoperative workup should include a CBC, 
SMA18, and chest x-ray. If elevated alkaline 
phosphatase or hypercalcemia is present, a bone 
scan should be completed. Abnormal liver function 
tests should be investigated with a CT scan of the 
liver. 



American Joint Committee on Cancer TNM 


staging for breast cancer 


Stage 


Description 


Tumor 


TX 




Primary tumor not assessable 


TO 




No evidence of primary tumor 


Tis 




Carcinoma in situ 


T1 




Tumor <2 cm in greatest dimension 




T1a 


Tumor <0.5 cm in greatest dimension 




T1b 


Tumor >0.5 cm but not > 1 cm 




T1c 


Tumor >1 cm but not >2 cm 


T2 




Tumor >2 cm but <5 cm in greatest 


T3 




dimension 


T4 




Tumor >5 cm in greatest dimension 
Tumor of any size with direct extension into 




T4a 


the chest wall or skin 

Extension to chest wall (ribs, intercostal 




T4b 


muscles, or serratus anterior) 




T4c 


Peau d'orange, ulceration, or satellite skin 




T4d 


nodules 

T4a plus b 

Inflammatory breast cancer 


Regional lym 


)h nodes 


NX 




Regional lymph nodes not assessable 


NO 




No regional lymph node involvement 


N1 




Metastasis to movable ipsilaterai axillary 


N2 




lymph nodes 


N3 




Metastases to ipsilaterai axillary lymph 

nodes fixed to one another or to other 

structures 

Metastases to ipsilaterai internal mammary 

lymph nodes 


Distant metas 


tases 


MX 




Non-accessible presence of distant 


MO 




metastases 


M1 




No distant metastases 

Existent distant metastases (including 

ipsilaterai supraclavicular nodes) 



American Joint Committee on Cancer 
classification for breast cancer based on TNM 
criteria 


Stage 


Tumor 


Nodes 


Metastase 
s 





Tis 


NO 


MO 


1 


T1 


NO 


MO 


MA 


TO, 1 
T2 


N1 
NO 


MO 
MO 


MB 


T2 
T3 


N1 
NO 


MO 
MO 


IMA 


TO, 1, 2 
T3 


N2 
N1,2 


MO 
MO 


1MB 


T4 
AnyT 


Any N 
N3 


MO 
MO 


IV 


AnyT 


Any N 


M1 



II. Surgical options for stage I and II breast cancer 

A. Breast conservation therapy (BCT) has been 
shown to result in survival and local recurrence 
rates equivalent to modified radical mastectomy; 
therefore, breast conservation is the preferred 
therapy for stage I and II breast cancer. The 
technique includes lumpectomy, axillary lymph 
node dissection, and breast irradiation. 

1. Contraindications to BCT 

a. Contraindications to radiotherapy (ie, prior 
breast irradiation, ongoing pregnancy) 

b. Steroid-dependent collagen vascular disease 

c. Tumor-breast ratio that would result in an 
unacceptable cosmetic result (eg, a large 
tumor in a small breast) 

d. Diffuse, malignant microcalcifications on 
mammography 

e. Tumor greater than 5 cm in diameter 

2. Lumpectomy technique. Incisions should be 
curvilinear and parallel with the nipple. A gross 
margin of 1 cm should be removed. The 
lumpectomy specimen is given immediately to 
the pathologist for inking and for an assessment 
of the gross margins. Subcutaneous tissue is 
closed, and the skin is approximated with a 
subcuticular suture. 

3. Follow-up following BCT consists of a physical 
examination every 3-4 months for the first 3 
years, every 6 months for the next 2-3 years, 
then yearly. A SMA18 and CBC are done at 
each visit and a chest x-ray is done yearly. 

4. A posttreatment mammography of the treated 
side is done 6 months after the completion of 
radiotherapy, then every 6 months for the first 2 
years, followed by annual mammograms. Yearly 
mammography should be performed on the 
opposite breast. 

B. Modified radical mastectomy consists of a total 
mastectomy and an axillary node dissection. In 
staging axillary lymphadenectomy, levels I and II 
are removed routinely. Reconstruction of the breast 
should be offered to all patients undergoing 
mastectomy. Physical examination schedule and 
blood work are the same as for lumpectomy. The 
chest wall should be examined for of recurrence. 
Mammography of the opposite breast should con- 
tinue yearly. 

III. Locally advanced breast cancer (LABC) consists of 
T3 NO (stage MB), MIA, and 1MB breast cancer. All 
LABC patients should undergo staging with CBC, 
SMA18, bone scan, and CT scan of chest and 
abdomen. 

A. Noninflammatory LABC. Multimodality therapy 
consists of neoadjuvant chemotherapy (ie, given 
before surgery), modified radical mastectomy, 
radiotherapy to the chest wall, axilla and 
supraclavicular nodes, and further chemotherapy. 

B. Inflammatory LABC (T4d). Inflammatory breast 
cancer is characterized by erythema of the skin, 
skin edema, warmth, tenderness, and an under- 
lying tumor mass. Treatment requires aggressive 
multi-modality therapy. 

C. Follow-up. Patients should be followed closely 
because they are at higher risk of local and distant 
recurrence. 

IV.Ductal carcinoma in situ (DCIS) consist of Tis, stage 
lesions. These lesions consists of malignant ductal 



cells that have not penetrated the basement 
membrane. DCIS is a precursor of invasive ductal 
cancer. 

A. Physical examination is usually normal with 
DCIS. The most common presentation is 
suspicious microcalcifications on mammography. 
DCIS can cause a nipple discharge or a palpable 
mass. 

B. Surgical therapy 

1. BCT. Lumpectomy and adjuvant radiotherapy 
are an alternative to mastectomy in well- 
localized DCIS when negative microscopic 
margins can be obtained. 

2. Total mastectomy, including removal of the 
nipple areolar complex and breast tissue, 
results in survival rates of 98-99%. An axillary 
dissection is not done routinely because the 
chance of nodal involvement is only 1-2%. 

References: See page 112. 



Urologic Disorders 

C. Garo Gholdoian, MD 
David A. Chamberlin, MD 



Prostate Cancer 

The average age at diagnosis of prostate cancer is 73 
years. The prevalence of prostate cancer is 30% in men 
over the age of 50. One in six men will be diagnosed with 
prostate cancer during their lifetimes. Prostate cancer is 
the most common cancer in men except for skin cancer. 

I. Clinical evaluation 

A. Prostate cancer is usually asymptomatic at 
presentation, and it is usually detected by 
abnormalities on rectal examination or a high 
serum prostate specific antigen (PSA) 
concentration. 

B. Prostate cancer can cause urinary urgency, 
nocturia, frequency, and hesitancy; these 
symptoms are more likely to be caused benign 
prostatic hypertrophy (BPH) than cancer. The new 
onset of erectile dysfunction raises the suspicion of 
prostate cancer. 

C. Serum PSA elevation. A total serum PSA 
concentration >4.0 ng/mL is considered abnormal, 
and is suspicious for prostate cancer. An elevated 
serum PSA between 4 to 10 ng/mL has a positive 
predictive value (PPV) of 21 percent, and a PSA 
above 10 ng/mL has a positive predictive value of 
64 percent. Causes of an elevated PSA include 
benign prostatic hyperplasia, prostate cancer, 
prostatitis, and perineal trauma. 

D. Abnormal prostate examination. The PPV of an 
abnormal digital rectal examination for prostate 
cancer is 5 to 30 percent. All men with induration, 
asymmetry, or palpable nodularity of the prostate 
gland require further diagnostic studies to rule out 
prostate cancer. A serum PSA should be obtained 
prior to biopsy. 

II. Screening for prostate cancer 

A. Age is the most important risk factor for prostate 
cancer. Prostate cancer rarely occurs before the 
age of 45, but the incidence rises rapidly thereafter. 
Prostate cancer is more common in black men. 

B. Recommendations for screening. The American 
Cancer Society recommends that serum prostate 
specific antigen testing and digital rectal 
examination should be offered annually to men 50 
years of age and older who have a life expectancy 
of 10 years. Screening should begin at age 45 in 
patients at high-risk for prostate cancer (eg, African 
Americans and men with two or more first-degree 
relatives with prostate cancer). PSA testing is also 
recommended for men who ask their clinicians to 
make the decision about screening on their behalf. 
The American Urological Association also supports 
this policy. 

C. If the serum PSA concentration is abnormal 
(>4.0 ng/mL), the test should be repeated in four 
weeks for confirmation. The most common 
explanation for an elevated serum PSA value is 
BPH. A man who has a high serum PSA 
concentration should be treated with an antibiotic 
(trimethoprim-sulfamethoxazole ([Bactrim] 1 DS 
tablet bid for 3 weeks) because prostatitis is a 
common cause of a high PSA. The PSA 
concentration should be repeated in 4 weeks. A 
return of the PSA to normal is expected if prostatitis 
was solely responsible. 

D. Prostate biopsy is the gold standard for prostate 
cancer diagnosis. Transrectal biopsy is a simple 
office technique that requires no analgesia. In a 
standard ultrasound-guided biopsy, a specimen is 
removed with a biopsy gun from any suspicious 
areas (eg, by rectal examination or biopsy) followed 
by six to ten tissue cores from the base, midzone, 
and apical areas of the right and left lobes of the 
gland (sextant biopsies). 

III. Diagnostic and staging evaluation of prostate 
cancer 

A. The tumor-node-metastasis (TNM) system is the 
most popular method of staging prostate cancer. 
Men are usually assigned a clinical stage, or "c" 
stage, and a pathological stage, or "p" stage. The 
"c" stage is determined by the digital rectal 
examination, while the "p" stage is determined after 
a pathologist has evaluated a radical prostatectomy 
specimen. 

IV. Staging of prostate cancer 



Staging of Prostate Cancer by 2002 AJCC 


Staging System 




Clinical tumor (cT) 


Substage 


stage 






Stage cT1 


T1a 


Tumor incidental 


Clinically inapparent 




histologic finding in 


tumor neither 




five percent or less of 


palpable nor visible 




tissue resected 


by imaging 


T1b 


Tumor incidental 
histologic finding in 
more than five percent 
of tissue resected 




T1c 


Tumor identified by 
needle biopsy (eg, 
because of elevated 
PSA) 


Stage cT2 


T2a 


Tumor involves one- 


Tumor confined 




half of one lobe or less 


within the prostate 


T2b 


Tumor involves more 
than one-half of one 
lobe but not both 
lobes 




T2c 


Tumor involving both 
lobes 


Stage cT3 


T3a 


Extracapsular 
extension (Unilateral 
or bilateral) 




T3b 


Tumor invades the 
seminal vesicle(s) 


Stage cT4 




Tumor is fixed or invades adjacent structures other than 


seminal vesicles: bladder neck, 


external sphincter, 


rectum, levator muscles, and/o 


pelvic wall. 



Stage Grouping for Prostate Cancer, 2002 AJCC 
Criteria 


Stage I 


T1a 


NO 


M0 


G1" 


Stage II 


T1a 


NO 


M0 


G2, 3-4 


NO 


M0 


AnyG 


NO 


M0 


AnyG 


NO 


M0 


AnyG 


NO 


M0 


AnyG 


Stage 
III 


T3 


NO 


M0 


AnyG 


Stage 
IV 


T4 


NO 


M0 


AnyG 


AnyT 


N1 


M0 


AnyG 


AnyT 


Any N 


M1 


AnyG 


"Grade: tumor grade is assessed as follows: 

Grade 1: Well differentiated (slight anaplasia), Gleason 

score 2-4 

Grade 2: Moderately differentiated (moderate anaplasia) 

Gleason score 5-6 

Grade 3-4: Poorly differentiated/undifferentiated (marked 

anaplasia) Gleason score 7-10 



Tumor histology (Gleason score). Analysis of 
the tumor histology provides an index of prognosis 
and may guide local therapy. Gleason score of 
one represents the most well-differentiated 
appearance, and Gleason score ten represents 
the most poorly differentiated. 
Clinical staging 

1. Serum PSA. This value is not used for staging, 
but may help to predict the local extent of 
disease in men with prostate cancer. There is 
a higher likelihood of finding organ-confined 
disease when the serum PSA concentration is 
less than 4.0 ng/mL. A serum PSA 
concentration of 4.1 to 10.0 ng/mL increases 
the likelihood of finding an organ-confined 
tumor larger than 0.5 mL, but also increases 
the odds of finding extracapsular extension by 
5.1-fold. A serum PSA concentration higher 
than 10.0 ng/mL increases the likelihood of 
finding extraprostatic extension by 24 to 50- 
fold. 

2. Radionuclide bone scan. A positive 
radionuclide bone scan indicates extraprostatic 
spread and eliminates the potential for curative 
surgery. Bone scan need not be performed in 
a patient with clinical stage T1 or T2 cancer on 
physical examination, a Gleason score of six or 
less, and a serum PSA value less than 10 



ng/mL 

3. CT scan should be considered in men who are 
going to be treated with external beam 
radiation therapy, and in men who have a PSA 
>10 to 15 ng/mL or a Gleason score greater 
than six. These men have an increased 
likelihood of pelvic lymph node metastasis. 

4. Endorectal coil magnetic resonance 
imaging (MRI) of the prostate gland utilizing an 
endorectal probe can determine the likelihood 
of either seminal vesicle involvement or 
extracapsular extension in patients who are 
thought to have clinically localized prostate 
cancer. The likelihood of cure with either 
radiation therapy or radical prostatectomy is 
low in these locally advanced patients, and 
surgery is usually not recommended. 

V. Treatment for early prostate cancer (organ- 
confined) 

A. The three standard therapies for men with early 
stage (organ-confined) prostate cancer are radical 
prostatectomy (RP), radiotherapy (RT), and 
watchful waiting. Hormone therapy is reserved for 
patients with locally advanced or metastatic 
prostate cancer. 

B. Radical prostatectomy 

1. RP involves excising the entire prostate from 
the urethra and bladder, which are then 
reconnected. This treatment offers the best 
chance of long-term survival. The retropubic 
approach to RP permits pelvic lymph node 
sampling prior to prostate removal to confirm 
the presence or absence of metastases. The 
prostate is removed if the lymph nodes are free 
of disease. 

2. Fifteen-year progression-free survival rates 
have been reported to be 80 to 85 percent for 
men with organ-confined disease. 

3. Complications 

a. Incontinence. About 1.6 percent report no 
urinary control, 7 percent report frequent 
leakage, and 42 percent report occasional 
leakage. 

b. Impotence. The potency rate after surgery is 
100 percent in men in their 40s, 55 percent 
for men in their 50s, 43 percent for men in 
their 60s, and percent for men in their 70s. 

4. Perineal prostatectomy can be considered for 
men with lower grade and low volume tumors, 
such as those with Gleason score <6 and a 
serum PSA less than 10 ng/mL. The likelihood 
of extraprostatic disease is less than 5 percent 
in such patients, making pelvic lymph node 
dissection unnecessary. 

C. Radiation therapy 

1. Radiation therapy (RT) does not require 
hospitalization and normal activity can usually 
be maintained during the course of treatment. 
Cure rates with RT appear to be comparable to 
those with RP for clinically localized disease for 
the first five years. Late recurrences following 
RT ten years or more after treatment occur 
more frequently than with RP. 

D. Watchful waiting 

1 . Watchful waiting describes patients who forego 
treatment. Such patients are followed with 
serum PSA measurements and digital rectal 
examination every three months for the first 
year, and then less frequently. Definitive or 
palliative therapy is initiated if a significant 
change in the serum PSA concentration or 
DRE occurs. 

2. Men with early stage disease (TO to T2, NX, 
MO) were followed for ten years, and only 9 
percent died of prostate cancer. The survival 
rate of this "watched" group was similar to the 
survival rate of the treated group. 

E. Recommendations 

1 . Early stage (organ-confined) prostate cancer is 
a curable disease in the majority of men. 
Surgery and radiation therapy offer equivalent 
survival outcomes for the first ten years after 
therapy; beyond that time, there is a higher risk 
of recurrence with radiation. Younger men 
should undergo surgery because of the 
potential for late recurrence with radiation 
therapy. Older men may prefer radiation 
therapy. 

2. Watchful waiting is not a good option for men 
with high-risk tumors (eg, Gleason score 7 or 
higher) unlessthey have significant comorbidity 
that suggests a markedly reduced life 
expectancy. The ideal candidate for watchful 
waiting is over the age of 70 to 75 with a life 
expectancy of 10 to 15 years or less who has a 



low grade tumor (eg, Gleason score 2 to 4). 
F. Endocrine therapy of advanced prostate 
carcinoma. Treatment of stage IV prostate cancer 
involves surgical or medical castration. Total 
blockade with leuprolide (Lupron) plus flutamide 
(Eulexin) is slightly better than leuprolide alone. 
Orchiectomy is an outpatient procedure that is the 
safest and least expensive option. The incidence 
of impotence with orchiectomy is no different than 
with medical castration therapies. 
References: See page 112. 



Renal Colic 

Approximately 5% of the U.S. population will pass a 
urinary tract stone during their lifetime. 

I. Pathophysiology 

A. Calcium-containing stones are the most common 
(70%). 

B. Magnesium-ammonium-phosphate stones, also 
known as struvite stones, are almost always 
associated with urinary tract infection with urea- 
splitting bacteria, such as Proteus mirabilis. 

C. Uric acid stones are less common and are 
radiolucent, making diagnosis by plain films alone 
difficult. 

D. Cystine stones are rare and associated with 
cystinuria, a rare autosomal recessive hereditary 
disorder. 

II. Clinical evaluation 

A. Renal colic is characterized as severe colicky pain 
that is intermittent and usually in the flank or lower 
abdomen. Patients usually cannot find a 
"comfortable position," and the pain often radiates 
to the testes or groin. A history of previous stones, 
poor fluid intake, urinary tract infections, or 
hematuria is common. 

B. Obstruction located at the ureteropelvic junction 
causes pure flank pain, while upper ureteral ob- 
struction causes flank pain that radiates to the 
groin. Midureteral stones cause lower abdominal 
pain and may mimic appendicitis or diverticulitis, 
but without localized point tenderness or guarding. 
Lower ureteral stones may cause irritative voiding 
symptoms and scrotal or labial pain. 

C. Patients with nephrolithiasis generally complain of 
nausea and vomiting. They commonly have gross 
or microscopic hematuria, fever, and an increased 
white blood cell count may. Prior episodes of renal 
colic or a family history of renal stones is often 
reported. 

D. Physical examination 

1. Generally the patient is agitated, diaphoretic, 
and unable to find a comfortable position. 

2. Hypertension and tachycardia are common. 

3. Costovertebral angle tenderness is the classic 
physical finding; however, minimal abdominal 
tenderness without guarding, rebound or rigidity 
may be present. Right or left lower quadrant 
tenderness or an enlarged kidney may 
sometimes be noted. 

III. Differential diagnosis. Appendicitis, salpingitis, 
diverticulitis, pyelonephritis, ovarian torsion, prostatitis, 
ectopic pregnancy, bowel obstruction, carcinoma. 

IV. Laboratory evaluation 

A. A urinalysis with microscopic, serum electrolytes, 
BUN, creatinine, complete blood count, and urine 
culture should be obtained. An elevated white 
blood cell count may be noted. A significant 
number of white cells in the urine also suggests 
infection. 

B. A plain abdominal film may demonstrate a 
calcification along the course of the urinary tract. 
Ninety percent of all calculi are radiopaque. 
Calcifications are frequently obscured by overlying 
bowel gas. 

C. Intravenous pyelogram. IVP is the gold standard 
for the diagnosis of urolithiasis, and it allows rapid 
assessment of the degree of obstruction, location 
of the stone and any renal function impairment. 
Acute ureteral obstruction may appear as a dense 
nephrogram with a delay in excretion of contrast. 

D. Ultrasound may be useful in patients with renal 
failure or an intravenous contrast allergy. 

V. Management of renal calculi 

A. Most renal calculi will pass spontaneously, and 
only expectant management with hydration and 
analgesia is necessary. Obstruction associated 
with fever indicates urinary tract infection, and it 
requires prompt drainage with either a ureteral 
stent or percutaneous nephrostomy. 

B. Indications for admission 

1. High fever, uncontrollable pain 



2. Intractable nausea and vomiting with an inability 
to tolerate oral fluids 

3. Solitary kidney 

C. Inpatient management 

1 . Vigorous intravenous hydration and intravenous 
antibiotics are important when infection is 
suspected. Parenteral narcotics are often 
necessary. 

2. Ketorolac (Toradol), 60 mg IM/IV, then 15-30 
mg IM/IV q6h, is effective and provides a good 
alternative to narcotics. 

3. Strain all urine in an attempt to retrieve 
spontaneously passed stones for X-ray crystal- 
lographic analysis. 

4. Stones measuring 5 to 10 mm have a 
decreased likelihood of passage, and early 
elective intervention should be considered 

5. Extracorporeal shock-wave lithotripsy (ESWL) 
is the most common procedure for small renal 
or ureteral stones. Eighty percent of patients 
become stone-free after one treatment. 

6. Ureteroscopy with laser, ultrasound or 
electrohydraulic lithotripsy may be used as well. 
Open surgical stone removal is rarely 
necessary. 

D. Outpatient management 

1. Most patients with renal colic do not require 
admission. The majority of stones measuring 
less than 4 mm will pass spontaneously (90- 
95%), and 80% of these will pass within 4 
weeks. 

2. Patients should increase intake of oral fluids, 
take narcotic pain medication, and strain all 
urine. Plain abdominal films may be used to 
assess movement of the stone. 

E. Follow-up care 

1. After the stone has passed, a metabolic 
evaluation is important because 70% of patients 
will have repeat stones if not diagnosed. 

2. Evaluation may include chemistry screening, 
calcium, uric acid, phosphorous, urine cystine 
(nitroprusside test), 24 hour urine collection for 
uric acid, calcium, creatinine. 

References: See page 112. 



Urologic Emergencies 

I. Acute urinary retention 

A. Acute urinary retention is characterized by a 
sudden inability to void. It often presents with 
suprapubic pain and severe urgency. There is 
usually a history of preexisting obstructive voiding 
symptoms related to bladder outlet obstruction or 
poor detrusor function. 

B. Benign prostatic hyperplasia is the most 
common cause of acute urinary retention in men 
over the age of 50. 

1. Patients present with progressively worsening 
voiding difficulties, resulting in bladder 
overdistention and subsequent urinary 
retention. 

2. Prostate size on digital rectal examination has 
no bearing on the degree of outlet obstruction 
because minimal enlargement of the prostate 
can cause significant obstruction. 

C. Prostate cancer accounts for 25% of patients with 
acute urinary retention. Ten percent of patients 
with prostate cancer initially present with bladder 
outlet obstruction. 

D. Additional causes of acute urinary retention 
include urethral strictures, bladder neck 
contractures, bladder stones, and acute bacterial 
prostatitis. Acute urinary retention may be caused 
by prolonged obstruction, diabetes mellitus, 
neurologic disorders (spinal cord injury, herniated 
vertebral disk), and medications. 

E. Urinary retention after surgery sometimes 
temporarily develops in elderly men. Preexisting 
bladder dysfunction or outlet obstruction is usually 
present. 

F. Anticholinergic medications (antihistamines, 
antidiarrheals, antispasmodics, tricyclic 
antidepressants) can suppress bladder function. 
Sympathomimetic drugs (decongestants and diet 
pills) that cause contraction of the bladder neck 
can precipitate an increase in outlet resistance. 

G. Complications of acute urinary retention include 
postobstructive diuresis, bladder mucosal 
hemorrhage, hypotension, sepsis, renal failure, 
and autonomic bladder hyporeflexia 

H. Clinical evaluation of acute urinary retention 
1 . Retention is characterized by an inability to void 
and suprapubic discomfort. A progressive 
history of difficulty voiding and irritative voiding 



symptoms, such as frequency, nocturia or 
urgency is often noted. 

2. Some patients are incontinent as a result of 
extreme overdistention of the bladder. A past 
history of gonorrhea, trauma, underlying dis- 
eases, or medications should be sought. 

3. Palpate for a distended bladder and assess size 
and consistency of the prostate. Tenderness of 
the prostate on rectal examination suggests 
acute prostatitis; a diffusely hard or nodular 
prostate suggests carcinoma. 

4. The penis should be examined to rule out 
phimosis, paraphimosis, or meatal stenosis. A 
neurologic exam should include anal sphincter 
reflex and perineal sensation. 

5. Laboratory evaluation. Serum electrolytes, 
blood urea nitrogen (BUN), creatinine, 
urinalysis, and urine culture. 

I. Management of acute urinary retention 

1. The entire bladder contents should be drained 
with a Foley catheter. Adequate volume 
replacement is necessary to prevent 
hypotension. 

2. Lubrication with 2% lidocaine jelly (injected 
directly into the urethra with a syringe) will 
facilitate insertion of a urethral catheter. 
Medium-sized catheters (#18 to #22 French) 
should be used because they tend to be stiffer 
and easier to insert than smaller ones. 

3. In patients with large prostates, Coude' 
catheters (which have a curved tip) may be 
helpful. The curve of the Coude' catheter should 
be directed superiorly. Other methods of 
drainage include urethral sounds, filiforms with 
followers, and percutaneous suprapubic tubes. 

4. Admission to the hospital is not required for 
most patients with acute urinary retention 
unless infection or renal failure are present. 
Most patients can be managed with a Foley 
catheter and discharged home with oral 
antibiotics and a leg urine bag. 

. Testicular torsion 

A. Testicular torsion is an emergency. Delay in 
treatment may result in testicular loss. A four- to 
six-hour delay may impair normal testicular 
function. Torsion can occur at any age; however, it 
is most common in adolescents, peaking at the 
age of 15 to 16 years. 

B. Testicular torsion presents with sudden onset of 
pain and swelling in one testicle, occasionally 
associated with minor trauma. Nausea, vomiting, 
and lower abdominal or flank pain are common. A 
history of previous similar episodes with 
spontaneous resolution is common. 

C. A urinalysis is essential in differentiating testicular 
torsion from epididymitis; however, a negative 
urinalysis does not rule out epididymitis. 

D. Differential diagnosis of testicular torsion 

1 . Epididymitis due to Neisseria gonorrhoeae and 
Chlamydia trachomatis is much more common 
than torsion in adult men. 

2. Torsion of an appendix testis or appendix 
epididymis may mimic testicular torsion . Torsion 
of the appendix testis may manifest as a tender, 
pea-sized nodule at the upper pole of the 
testicle with a small blue-black dot seen through 
the scrotal skin (the blue dot sign). Management 
is conservative; however, if there is diagnostic 
uncertainty, surgical exploration is required. 

3. Other less common conditions that may present 
similarly to torsion include acute hemorrhage 
into a testicular neoplasm, orchitis, testicular 
abscess, incarcerated hernia, and testicular 
rupture. 

E. Physical examination 

1. Testicular torsion usually presents with severe 
unilateral testicular pain with an acute onset. 
The pain is associated with an extremely tender 
testicle with a transverse lie or an anterior 
epididymis that lies high in the scrotum. 

2. With testicular torsion, the testis is usually high 
in the scrotum (Brunzel's sign). The presence of 
a cremasteric reflex almost always rules out 
testicular torsion. 

3. Relief of pain by elevation of the affected testis 
(Prehn's sign) suggests epididymitis. A negative 
Prehn's sign suggests testicular torsion. 

F. Diagnostic imaging. Diagnostic testing should not 
delay surgical exploration in acute torsion. If the 
diagnosis is unclear, diagnostic tests may be 
useful. Color Doppler ultrasound is the most 
valuable diagnostic study, with nearly a 100% 
sensitivity and specificity. 

G. Management of testicular torsion 

1. Immediate detorsion is imperative for all cases 



of testicular torsion. Testicular salvage rates 
decrease to 50% at 10 hours and to 10-20% at 
24 hours. 

2. Manual detorsion can be attempted as an ur- 
gent measure by rotating the testicle medially 
about its pedicle. Surgical orchiopexy is still 
required. 

3. If an infarcted testicle is noted during surgical 
exploration, it should be removed. If the testicle 
is viable, both testicles should be fixed in the 
scrotum with nonabsorbable sutures. 

III. Priapism 

A. Priapism is defined as a prolonged penile erection. 
Most cases of priapism in adults are idiopathic. In 
children the most common causes are sickle cell 
anemia, hematologic neoplasms (leukemia), and 
trauma. 

B. Evaluation of priapism 

1 . Patients usually complain of a persistent, painful 
erection. They may have fever and voiding 
difficulties. 

2. Physical examination should include a 
neurologic evaluation and perineal inspection 
for neoplasms. Examination of the penis usually 
reveals a flaccid glans despite a rigid corpora 
cavernosa. Hematologic studies should be 
performed to rule out sickle cell anemia and 
leukemia. 

C. Treatment of priapism 

1. Early treatment reduces the risk of long-term 
impotence, which may occur in 50%. Discomfort 
can be reduced with parenteral narcotic 
analgesics and sedation. Detumescence may 
be achieved using cold compresses, ice packs, 
warm- or cold-water enemas, and prostate 
massage. 

2. If these treatments are unsuccessful, the static 
blood may be aspirated from the corpora using 
a large bore needle. Followed by irrigation of 
the corpora with saline containing a 
vasoconstricting agent (phenylephrine, 
epinephrine, or metaraminol). 

3. If this process fails to achieve detumescence, a 
shunt may be created between the affected 
corpora cavernosa and unaffected corpus 
spongiosum with a Tru-Cut biopsy needle. 
When priapism is secondary to sickle cell 
anemia, therapy also includes hydration, 
oxygen, and blood transfusion. 

References: See page 112. 



Vascular and Orthopedic 
Surgery 



Peripheral Arterial Occlusive 
Disease 



Peripheral arterial occlusive disease affects about 18 
percent of persons over 70 years of age. The disease 
presents with intermittent claudication with pain in the 
calf, thigh or buttock, which is elicited by exertion and 
relieved with a few minutes of rest. In most cases, the 
underlying etiology is atherosclerotic disease of the 
arteries. 

I. Pathophysiology 

A. The incidence of claudication rises sharply between 
ages 50 and 75 years, particularly in persons with 
coronary artery disease. This condition affects at 
least 10% of persons over 70 years of age and 2% 
of those 37-69 years of age. 

B. Risk factors. Cigarette smoking is the most 
important risk factor for PAOD. Seventy to 90% of 
patients with arterial insufficiency are smokers. 
Other risk factors include hyperlipidemia, diabetes 
mellitus, and hypertension. 

C. After five years, 4% of patients with claudication 
lose a limb and 16% have worsening claudication 
or limb-threatening ischemia. The five-year 
mortality rate for patients with claudication is 29%; 
60% of deaths result from coronary artery disease, 
15% from cerebrovascular disease, and the 
remainder result from nonatherosclerotic causes. 

II. Clinical evaluation of claudication 
A. Claudication 

1. The key clinical features of claudication are 
reproducibility of muscular pain in the thigh or 
calf aftera given level of activity and cessation of 
pain after a period of rest. 

2. Patients should be asked about the intensity of 
claudication, its location, and the distance they 
have to walk before it begins. The degree of 
functional impairment should be assessed. 

3. Aortoiliac disease is manifest by discomfort in 
the buttock and/or thigh and may result in 
impotence and reduced femoral pulses. 
Leriche's syndrome occurs when impotence is 
associated with bilateral hip or thigh 
claudication. 

4. Iliofemoral occlusive disease is characterized 
by thigh and calf claudication. Pulses are 
diminished from the groin to the foot. 

5. Femoropopliteal disease usually causes calf 
pain. Patients have normal groin pulses but 
diminished pulses distally. 

6. Tibial vessel occlusive disease may lead to 
foot claudication, rest pain, non-healing wounds, 
and gangrene. 

7. Rest pain consists of severe pain in the distal 
portion of foot due to ischemic neuritis. The pain 
is deep and unremitting, and it is exacerbated by 
elevation of the foot and is relieved by dangling 
the foot over the side of the bed. 

III. Physical examination 

A. Evaluation of the peripheral pulses should 
include the femoral, popliteal, posterior tibial, and 
dorsalis pedis arteries. Pallor on elevation of the 
extremity and rubor when the limb is dependent is 
common. 

B. Other signs of chronic arterial insufficiency include 
brittle nails, scaling skin, hair loss on the foot and 
lower leg, cold feet, cyanosis, and muscle atrophy. 
The feet should be inspected for skin breakdown or 
ulceration. 

C. Bruits may be auscultated distal to the arterial 
obstruction. Abdominal examination for a "pulsatile 
mass" should be performed because of the 
association between abdominal aortic aneurysm 
and peripheral arterial disease. 

D. Ankle-brachial index is an effective screening tool. 
The ankle-brachial index is calculated by dividing 
the ankle pressure by the brachial systolic 
pressure. 

1 . The normal ABI is above 1 .0, since the pressure 
is higher in the ankle than in the arm. 

2. An ABI below 0.9 has a 95 percent sensitivity for 
detecting angiogram-positive peripheral vascular 
disease. 

3. An ABI of 0.40 to 0.90 suggests a degree of 
arterial obstruction often associated with 
claudication. 



4. An ABI below0.4 represents advanced ischemia 

5. In patients with an abnormal ankle-brachial 
index, testing with segmental arterial pressures 
and a pulse volume recording before and after 
exercising to the point of absolute claudication 
are indicated. 



Ankle-Brachial Index Interpretation 


Normal 


>1 


Abnormal 


<0.95 


Intermittent 
claudication 


0.4-0.9 


Severe 
disease/ischemia 


less than 0.4 



E. Segmental arterial pressures. The proximal lower 
extremity pressures should be equal to or greater 
than the upper extremity pressures, and the drop in 
Doppler pressure between segments no greater 
than 20 mm Hg. These studies help predict the 
location and severity of the disease. 

F. Arteriography or magnetic resonance 
angiography is required to delineate the extent of 
the disease when intervention is anticipated. 

IV. Management 

A. Risk factor modification 

1. The goals of risk factor modification in patients 
with PAOD are the same as those in patients 
with coronary artery disease. Hypertension 
should be controlled. Beta-blockers do not 
usually worsen claudication. 

2. Lipid abnormalities must be treated. The 
target LDL cholesterol level is less than 100 mg 
per dL in patients with symptomatic vascular 
disease. 

3. Tobacco is directly toxic to the vascular 
endothelium and worsens atherosclerosis. All 
patients must abstain from tobacco use. 

B. Antiplatelet agents 

1. Aspirin should be considered for use in any 
patient with coronary artery disease, 
cerebrovascular disease or PAOD. 

2. Clopidogrel (Plavix), 75 mg qd, or ticlopidine 
(Ticlid), 250 mg bid with meals, should be 
considered in patients who are intolerant of 
aspirin therapy. Clopidogrel and ticlopidine are 
platelet inhibitors; however, clopidogrel has a 
lower risk of neutropenia. 

C. Exercise. Walking improves the symptoms of 
claudication. Patients should walk at least three 
times per week for at least 30 minutes at each 
session. Near-maximal claudication pain (absolute 
claudication distance) should be the resting point, 
and the patient should follow the program for at 
least six months. 

D. Medication 

1. Cilostazol (Pletal), 100 mg bid, is a 
phosphodiesterase inhibitor that suppresses 
platelet aggregation and acts as a direct arterial 
vasodilator. Cilostazol results in a 35 percent 
increase in the distance before claudication and 
a 41 percent increase in absolute claudication 
distance. 

2. Pentoxifylline (Trental), 400 mg tid with meals, 
provides small improvements in the initial 
claudication distance and absolute claudication 
distance. 

E. Operative and endovascular procedures 

1. Most patients with claudication respond to 
conservative therapy. Surgery is reserved for 
patients with rest pain or tissue loss. Patients 
who have intermittent calf claudication alone are 
not surgical candidates unless the claudication 
severely limits their lifestyle or occupational 
functioning. 

2. Patients with rest pain, tissue loss as a result of 
gangrene, or non-healing ulcers with an ABI less 
than 0.6 are surgical candidates. 

3. Percutaneous transluminal angioplasty hasa 
greater than 90% success rate in the treatment 
of short-segment aortoiliac occlusive disease, 
and these results may be improved with the 
placement of an intra-arterial stent. However, 
five-year patency rates are only 40-60%. 

4. Surgical bypass therapy is an effective 
treatment for claudication; however, it is 
associated with 5% morbidity and mortality rates. 
Aortobifemoral grafting has a 90% 5-year 
patency rate. Aortoiliac, femoral-femoral 
crossover, and reversed and in-situ saphenous 



vein bypass grafting from the common femoral to 

the popliteal artery have 60-70% 5-year patency 

rates. A synthetic polytetrafluoroethylene graft 

(PTFE) is indicated for above knee 

femoral-popliteal bypass, and it has a 50% 5- 

year patency rate. 

5. Axillofemoral bypass is useful for high risk, 

elderly patients who are unable to tolerate an 

aortic procedure. 

Management of the acutely threatened limb. An 

acutely occluded artery can cause limb loss within 

hours. The patient will complain of sudden onset of 

severe unrelenting rest pain. Atrial fibrillation often 

may cause acute embolic arterial occlusion. These 

patients require emergency surgical evaluation and 

immediate heparinization. 



Abdominal Aortic Aneurysms 

S.E. Wilson, MD 

Abdominal aortic aneurysms (AAAs) are the most 
common type of arterial aneurysm. Approximately 5% of 
people older than 60 years develop an abdominal aortic 
aneurysm, and the male-female ratio is 3:1. Other risk 
factors include smoking, hypertension, and a family 
history of an aneurysm. Abdominal aortic aneurysms are 
caused atherosclerosis in 90% of patients; 5% of 
aneurysms are inflammatory. 

I. Clinical evaluation 

A. Abdominal aortic aneurysms are usually 
asymptomatic. Aneurysm expansion or rupture 
may cause severe back, flank, or abdominal pain 
and shock. Distal embolization, thrombosis, and 
duodenal or ureteral compression can produce 
symptoms. 

B. Physical examination. Almost all AAAs greater 
than 5 cm are palpable as a pulsatile mass at or 
above the umbilicus. Abdominal aortic aneurysms 
range from 3 to 15 cm in diameter. 

II. Laboratory evaluation. Complete blood count, 
electrolytes and creatinine, blood urea nitrogen, 
coagulation studies, blood type and cross-matching, 
and urinalysis should be obtained. 

III. Radiologic evaluation 

A. Abdominal cross-table lateral films allow for 
estimation of aneurysm diameter. 

B. Ultrasonography and computed tomographic 
(CT) scanning demonstrate AAAs with an 
accuracy of 95% and 100%, respectively. 

IV. Elective management of abdominal aortic 
aneurysms 

A. Small aneurysms can be followed using ultrasound 
or CT scan every 6 months. 

B. Indications for repair include symptomatic 
aneurysms of any size, aneurysms exceeding 5.0 
cm, those increasing in diameter by more than 0.5 
cm per year, and saccular aneurysms. 

C. Preoperative management includes optimizing 
cardiopulmonary function and placement of a 
pulmonary artery catheter or a central venous line. 
An arterial line permits continuous BP and blood 
gas monitoring. Two peripheral venous catheters 
should be placed. 

D. Operative management. The aneurysm is 
approached through a midline abdominal incision 
and exposed by incising the retroperitoneum. 

E. The duodenum and left renal vein are dissected off 
the aorta. After heparinization, the aorta is cross- 
clamped first distal and then proximal to the 
aneurysm. Aortotomy is then made and extended 
longitudinally to the aneurysm "neck," where the 
aorta is either transected or cut in a T fashion. The 
aneurysm is opened, thrombus is removed, and 
bleeding lumbar arteries are suture ligated. Using 
a tube or bifurcation graft, the proximal 
anastomosis is performed to nonaneurysmal aorta. 
The distal anastomosis is completed at the aortic 
bifurcation (tube graft) or at the iliac or femoral 
arteries (bifurcation graft). 

F. Endovascular treatment of AAA uses a catheter 
to place a stent-graft. Early results suggest an 83% 
success rate and less than 6% mortality. 

References: See page 112. 



Orthopedic Fractures and 
Dislocations 

Harry Skinner, MD 
Michelle Schultz, MD 

I. Clinical evaluation of the injured limb 

A. Physical examination of the injured limb. 1) 

Inspection, 2) Palpation, and 3) Movement. 
Examine the bone for instability and examine the 
soft tissue for associated injury. Document 
neurologic and vascular status. 

B. Clinical features of fractures 

1 .Pain and tenderness. All fractures cause pain in 

the neurologically intact limb. 
2. Loss of function. Pain and loss of structural 

integrity of the limb cause loss of function. 
3. Deformity. Change in length, angulation, rotation 

and displacement. 
4. Attitude. The position of the fractured limb is 

sometimes diagnostic. The patient with a 

fractured clavicle usually supports the limb and 

rotates his head to the affected side. 
5.Abnormal mobility and crepitus: These signs 

should not be sought deliberately because pain 

and injury may result. 

C. Clinical features of dislocations 

1 .A dislocation occurs when the articular surfaces 
of a joint are no longer in contact. Subluxation 
(partial dislocation) is a less severe condition that 
occurs when the orientation of the surfaces is 
altered but they remain in contact. 

2.Pain and tenderness. Severe pain may be 
completely relieved when the joint is relocated. 

3. Loss of motion. Both active and passive motion 
are limited in dislocations. 

4. Loss of normal joint contour. In the anteriorly 
dislocated shoulder, the deltoid is flattened and 
the greater tuberosity of the humerus is no longer 
lateral to the acromion. 

5. Attitude. The patient carefully holds the anteriorly 
dislocated shoulder in abduction and external 
rotation. 

6. Neurologic injury. The incidence of neurologic 
injuries is much higherwith dislocations than with 
fractures. Shoulder dislocations are often 
associated with axillary nerve injury. Posterior 
dislocations of the hip can result in sciatic nerve 
contusion. Careful examination for neurologic 
status is indicated before any intervention. 

II. Clinical description of fractures 

A. Anatomic location 
I.What bone is fractured? 

2. Where on the bone is the fractured? 

a. Metaphyseal - at the flare of the bone. 

b. Diaphyseal - fracture through the diaphysis 
(proximal, middle, or distal third of shaft). 

c. Epiphyseal - fracture at the end of the bone. 
3.Salter classification of fractures (children): 

a. Type I. Fracture through physis, between the 
epiphysis and metaphysis. 

b. Type II. Fracture through physis, involving the 
metaphysis. 

c. Type III. Fracture through physis, involving the 
epiphysis. 

d. Type IV. Fracture through physis, involving the 
metaphysis and epiphysis. 

e. Type V. Crush injury to physis, between the 
epiphysis and metaphysis. 

B. Bony deformity: Describe any change in bone 
length, angulation, rotation, or displacement. 

C. Direction of the fracture line. Describe the 
radiographic direction of the fracture. 

1 .Transverse - perpendicular to the long axis of the 

bone 
2. Oblique -fracture is at an angle to the bone. 
3. Spiral - occurring secondary to torsional stress. 

D. Comminution: Fracture with more than two 
fragments. 

E. Open vs. Closed: In an open fracture the bone 
protrudes through the skin. 

F. Greenstick Fracture: one cortex is broken while 
the other remains intact 

III. Radiological evaluation of fractures. A minimum of 
two views at right angles to each other should be 
obtained. Visualize the joint above and below the 
injury and check for soft tissue swelling. Views of the 
uninjured extremity are often useful for comparison in 
children. 
IV. Management of fractures 
A. Fracture reduction 

I.The fracture must be restored to a normal 

anatomical position 
2. Muscle spasm should be relieved with traction, 



analgesics, and muscle relaxants. 
3. Bones must be in apposition, properly aligned in 

linear and rotatory directions, and set to proper 

length. 
4. Fractures should must be held in place with a 

splint or cast. 

B. Indications for operative treatment 

1 .Failure of closed methods to reduce or maintain 

reduction of the fracture. 
2. Displaced intraarticular fractures, where the 

fragments are sufficiently large to allow internal 

fixation. 
3. Multiple injuries - in a multiple trauma patient, 

operative treatment can allow early mobilization. 

Early mobilization can sometimes avoid the 

morbidity and mortality associated with prolonged 

recumbency. 

C. Upper extremity fractures and dislocations 

1. Distal radius fracture is often associated with 
fracture of the ulnar styloid. Treatment consists of 
closed reduction, casting and elevation of the 
extremity until swelling subsides. After reduction, 
check alignment with an x-ray, and rule out 
median nerve injury. 

2.Forearm shaft fracture: In adults, radius and 
ulna shaft fractures require open reduction and 
internal fixation. 

3. Humerus fracture: X-ray the entire bone and 
check for radial nerve injury (wrist drop). 
Humerus fractures are usually treated with a 
collar and cuff sling or coaptation splint 

4. Clavicle fractures. Subclavian artery and 
brachial plexus injury may occur. The fracture 
should be splinted with a figure of eight bandage 
or sling. Clavicle fracture rarely requires surgery. 

5.Anterior shoulder dislocation occurs when the 
humeral head has been forced anterior to the 
glenoid. It is usually caused by extension force 
applied to abducted arm. The patient presents 
with the arm in slight abduction, and he cannot 
bring his elbow to the side. There is a slightly 
depressed deltoid prominence and arm motion 
causes pain. Axillary nerve injury may occur, 
causing a sensory deficit over the deltoid. 
Treatment consists of reduction with gentle 
traction. 

6.Posterior shoulder dislocation occurs when the 
humeral head has been forced posterior to the 
glenoid. Dislocation may occur secondary to 
seizures or electrocution. The arm is held in 
adduction with internal rotation. Treatment 
consists of reduction with gentle traction. 

D. Lower extremity fractures and dislocations 

1. Femoral neck fractures: Osteoporotic bone 
predisposes to this intracapsular fracture. Internal 
fixation or endoprosthesis (artificial hip) are 
required. 

2. Intertrochanteric fractures usually occur in the 
elderly after a fall. The fracture is located outside 
the joint capsule. Treatment consists of internal 
fixation. 

3.Femoral shaft fracture: Early intramedullary 
nailing is recommended in adults. This fracture is 
rarely associated with a fat embolism syndrome. 

4.Patella fracture: If the bone is nondisplaced, it 
can be treated in a cast. If it is displaced more 
than 2 mm, it should be treated with internal 
fixation. Quadriceps function should be checked. 

5.Tibial shaft fracture can be treated closed or 
with internal fixation. 

6.Ankle fracture: Evaluate the stability of the ankle 
fracture by examining the joint space between the 
talus and tibial plafond. Unstable injuries require 
internal fixation. 

E. Knee injuries 

LKnee ligament testing 

a. Varus/valgus femur stress test. The 
examiner stabilizes the femur, and pressure is 
exerted outward or inward at the ankle; a tear 
of the collateral ligament is indicated by 
excess mobility. 

b. Anterior drawer test. Pull tibia anteriorly with 
the knee flexed 90 degrees to test for tear of 
anterior cruciate ligament. 

c. Posterior drawer test. Push tibia posteriorly 
with the knee flexed 90 degrees to test for tear 
of the posterior cruciate ligament. 

2. The most common ligamentous injuries are 
tearing of the medial collateral ligament by a blow 
from the lateral side of the knee, and tear of the 
anterior cruciate ligament by twisting on a planted 
foot. Brace immobilization is usually sufficient for 
medial collateral ligament tears. 

3. Dislocation of the knee often results in multiple 
ligament injury. Popliteal artery trauma should be 



excluded. Immobilization of the knee, followed by 
ligament reconstruction should be completed. 



Ankle Sprains 

A sprain is the most common ankle injury. Injury may 
range from minor ligamentous damage to complete tear 
or avulsion. Sprain occurs when stress is applied while 
the ankle is in an unstable position, causing the 
ligaments to overstretch. Stresses usually occur during 
running or walking over uneven surfaces. 

I. Clinical evaluation 

A. Ligaments of the lateral ankle consist of the anterior 
talofibular ligament, calcaneofibular ligament, and 
posterior talofibular ligament. 

B. Sprains may be classified as first-degree, involving 
stretching of ligamentous fibers, second-degree, 
involving a tear of some portion of the ligament with 
associated pain and swelling, and third-degree, 
implying complete ligamentous separation. 

C. An inversion injury is the most common type of 
sprain, causing damage to the lateral ligaments. 

II. Physical examination 

A. The examiner's fingertips should be used to check 
the anterior capsule and medial and lateral 
ligaments. 

B. Anterior draw sign suggests significant injury. The 
sign is elicited by grasping the distal tibia in one 
hand and the calcaneus and heel in the other and 
sliding the entire foot forward. This is done both 
with the ankle in neutral position and with 30°" of 
plantar flexion. With disruption of the anterior or 
lateral ligaments, 4 mm or more of anterior shift will 
occur. 

C. Passive inversion of the ankle may produce pain. 
Swelling occurs anterior to the lateral malleolus at 
the onset; ecchymoses are common. The talus tilts 
if the calcaneus is adducted. 

III. Radiographic evaluation 

A. X-rays are useful in cases of moderate to severe 
injury, helping to identify any associated skeletal 
injury in addition to assessing degree of 
ligamentous damage. 

B. A stress view is obtained (with local anesthesia, if 
necessary) to check for talar tilt. A tilt of more than 
15° is suggestive of lateral ligament injury; more 
than 25°' of tilt is diagnostic. 

IV. Management of ankle sprains 

A. Grade I sprains are defined as stretch of the 
ligaments without disruption. Grade I sprains should 
be treated with rest, ice, compression with an 
elastic bandage, elevation, and weight bearing as 
tolerated. 

B. Grade II sprains consist of partial tears of the ankle 
ligaments. Grade II sprains should be treated with 
rest, ice, compression with an elastic bandage, and 
elevation. A splint can be applied for a few days, 
followed by early range of motion. 

C. Grade III sprains consist of a complete tear of the 
ligaments. Treatment of grade III strains consists of 
rest, ice, compression with an elastic bandage, and 
elevation. A splint or cast can be applied for a short 
period of time, followed by early range of motion.