Spiller R, Garsed K. Postinfectious irritable bowelsyndrome. Gastroenterology. 2009;136(6):1979–1988.

Zanger P et al. Effectiveness of rifaximin in prevention of diarrhoea in individuals travelling to south and

southeast Asia: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Infect Dis.

2013;13(11):946–954.

Connor BA, Keystone JS. Editorial commentary: antibiotic self-treatment of travelers’ diarrhea: helpful or

harmful? Clin Infect Dis. 2015;60(6):847–848.

DuPont HL et al. A randomized, double-blind, placebo-controlled trial of rifaximin to prevent travelers’ diarrhea.

Ann Intern Med. 2005;142(10):805–812.

DuPont HL. Travelers’ diarrhea: contemporary approaches to therapy and prevention. Drugs. 2006;66:303.

Pennington H. Escherichia coli O157. Lancet. 2010;376(9750):1428–1435.

Karch H et al. Epidemiology and diagnosis of Shiga toxin-producing Escherichia coli infections. Diagn Microbiol

Infect Dis. 1999;34(3):229–243.

Mead PS, Griffin PM. Escherichia coli O157:H7. Lancet. 1998;352(9135):1207–1212.

Ericsson CD et al. Optimal dosing of trimethoprim-sulfamethoxazole when used with loperamide to treat

traveler’s diarrhea. Antimicrob Agents Chemother. 1992;36(12):2821–2824.

Dundas S et al. The Central Scotland Escherichia coli O157:H7 outbreak: risk factors for the hemolytic uremic

syndrome and death among hospitalized patients. Clin Infect Dis. 2001;33(7):923–931.

Davis TK et al. Treatment of Shiga toxin-producing Escherichia coli infections. Infect Dis Clin North Am.

2013;27(3):577–597.

Klein EJ et al. Shiga toxin-producing Escherichia coli in children with diarrhea: a prospective point-of-care study.

J Pediatr. 2002;141(2):172–171.

Gould LH et al. Recommendations for diagnosis of shiga toxin—producing Escherichia coli infections by clinical

laboratories. MMWR Recomm Rep. 2009;58(RR-12):1–14.

Banatvala N et al. The United States national prospective hemolytic uremic syndrome study: microbiologic,

serologic, clinical, and epidemiologic findings. J Infect Dis. 2001;183(7):1063–1070.

Besbas N et al. A classification of hemolytic uremic syndrome and thrombotic thrombocytopenic purpura and

related disorders. Kidney Int. 2006;70(3):423–431.

Ikeda K et al. Predictors for the development of haemolytic uraemic syndrome with Escherichia coli O157:H7

infections: with focus on the day of illness. Epidemiol Infect. 2000;124(3):343–349.

Tarr PI et al. Shiga-toxin-producing Escherichia coli and haemolytic uraemic syndrome. Lancet.

2005;365(9464):1073–1086.

Tserenpuntsag B et al. Hemolytic uremic syndrome risk and Escherichia coli O157:H7. Emerg Infect Dis.

2005;11(12):1955–1957.

Wong CS et al. The risk of the hemolytic–uremic syndrome after antibiotic treatment of Escherichia coli

O157:H7 infections. N EnglJ Med. 2000;342(26):1930–1936.

Slutsker L et al. A nationwide case-control study of Escherichia coli O157:H7 infection in the United States. J

Infect Dis. 1998;177(4):962–966.

Carter AO et al. A severe outbreak of Escherichia coli O157:H7–associated hemorrhagic colitis in a nursing

home. N EnglJ Med. 1987;317(24):1496–1500.

Thomas DE, Elliott EJ. Interventions for preventing diarrhea-associated hemolytic uremic syndrome: systematic

review. BMC Public Health. 2013;13:799.

Proulx F et al. Randomized, controlled trial of antibiotic therapy for Escherichia coli O157:H7 enteritis. J Pediatr.

1992;121(2):299–303.

Pavia AT et al. Hemolytic-uremic syndrome during an outbreak of Escherichia coli O157:H7 infections in

institutions for mentally retarded persons: Clinical and epidemiologic observations. J Pediatr. 1990;116(4):544–

551.

Cimolai N et al. Risk factors for the progression of Escherichia coli O157:H7 enteritis to hemolytic-uremic

syndrome. J Pediatr. 1990;116(4):589–592.

Panos GZ et al. Systematic review: are antibiotics detrimental or beneficial for the treatment of patients with

Escherichia coli O157:H7 infection? Aliment Pharmacol Ther. 2006;24(5):731–742.

Nelson JM et al. Antimicrobial and antimotility agent use in persons with shiga toxin-producing Escherichia coli

O157 infection in FoodNet Sites. Clin Infect Dis. 2011;52(9):1130–1132.

138.

139.

140.

141.

142.

143.

144.

145.

146.

147.

148.

149.

150.

151.

152.

153.

154.

155.

156.

157.

158.

159.

160.

161.

162.

163.

164.

165.

Bell BP et al. Predictors of hemolytic uremic syndrome in children during a large outbreak of Escherichia coli

O157:H7 Infections. Pediatrics. 1997;100(1):e12–e12.

Bartlett JG. Antibiotic-associated diarrhea. N EnglJ Med. 2002;346(5):334–339.

Freeman J et al. The changing epidemiology of clostridium difficile infections. Clin Microbiol Rev.

2010;23(3):529–549.

Wilcox MH et al. A case-control study of community-associated Clostridium difficile infection. J Antimicrob

Chemother. 2008;62(2):388–396.

Turco R et al. Proton pump inhibitors as a risk factor for paediatric Clostridium difficile infection. Aliment

Pharmacol Ther. 2009;31(7):754–759.

Dalton BR et al. Proton pump inhibitors increase significantly the risk of Clostridium difficile infection in a lowendemicity, non-outbreak hospitalsetting. Aliment Pharmacol Ther. 2009;29(6):626–634.

McDonald LC et al. An epidemic, toxin gene–variant strain of clostridium difficile. N Engl J Med.

2005;353(23):2433–2441.

Rao K et al. Clostridium difficile ribotype 027: relationship to age, detectability of toxins A or B in stool with

rapid testing, severe infection, and mortality. Clin Infect Dis. 2015;61(2):233–241.

Bartlett John G. The case for vancomycin as the preferred drug for treatment of clostridium difficile infection.

Clin Infect Dis. 2008;46(10):1489–1492.

Wanahita A et al. Conditions associated with leukocytosis in a tertiary care hospital, with particular attention to

the role of infection caused by clostridium difficile. Clin Infect Dis. 2002;34(12):1585–1592.

Savola KL et al. Fecal leukocyte stain has diagnostic value for outpatients but not inpatients. J Clin Microbiol.

2001;39(1):266–269.

Marra F, Ng K. Controversies around epidemiology, diagnosis and treatment of clostridium difficile infection.

Drugs. 2015;75(10):1095–1118.

Surawicz CM et al. Guidelines for diagnosis, treatment, and prevention of Clostridium difficile infections. Am J

Gastroenterol. 2013;108(4):478–498;quiz 499.

Peled N et al. Predicting clostridium difficile toxin in hospitalized patients with antibiotic associated diarrhea.

Infect Control Hosp Epidemiol. 2007;28(4):377–381.

Bartlett JG. Antibiotic-associated diarrhea. Clin Infect Dis. 1992;15(4):573–581.

Nelson R. Antibiotic treatment for Clostridium difficile-associated diarrhea in adults. Cochrane Database Syst

Rev. 2007:CD004610.

Fekety R. Guidelines for the diagnosis and treatment of Clostridium difficile-associated diarrhea and colitis. Am J

Gastroenterol. 1997;92:739.

Johnson S, Gerding DN. Clostridium difficile-associated diarrhea. Clin Infect Dis. 1998;26(5):1027–1034.

Pepin J et al. Emergence of fluoroquinolones as the predominant risk factor for clostridium difficile-associated

diarrhea: a cohort study during an epidemic in Quebec. Clin Infect Dis. 2005;41(9):1254–1260.

Al-Nassir WN et al. Comparison of clinical and microbiological response to treatment of Clostridium difficileassociated disease with metronidazole and vancomycin. Clin Infect Dis. 2008;47(1):56–62.

Cohen Stuart H et al. Clinical practice guidelines for clostridium difficile infection in adults: 2010 update by the

Society for Healthcare Epidemiology of America (SHEA) and the Infectious Diseases Society of America

(IDSA). Infect Control Hosp Epidemiol. 2010;31(5):431–455.

Pépin J et al. Outcomes of clostridium difficile-associated disease treated with metronidazole or vancomycin

before and after the emergence of NAP1/027. Am J Gastroenterol. 2007;102(12):2781–2788.

Teasly DG et al. Prospective randomized study of metronidazole versus vancomycin for clostridium-associated

diarrhea and colitis. Lancet. 1983;2:1043.

Tedesco F et al. Oral vancomycin for antibiotic-assocaited psedumembranous colitis. Lancet.

1978;312(8083):226–228.

Cohen SH et al. Clinical practice guidelines for Clostridium difficile infection in adults: 2010 update by the society

for healthcare epidemiology of America (SHEA) and the infectious diseases society of America (IDSA). Infect

Control Hosp Epidemiol. 2010;31(5):431–455.

Pelaez T et al. Reassessment of Clostridium difficile Susceptibility to Metronidazole and Vancomycin.

Antimicrob Agents Chemother. 2002;46(6):1647–1650.

Edwards DL et al. Disulfiram-like reaction associated with intravenous trimethoprim-sulfamethoxazole and

metronidazole. Clin Pharm. 1986;5(12):999–1000.

Fekety R et al. Treatment of antibiotic-associated Clostridium difficile colitis with oral vancomycin: comparison

of two dosage regimens. Am J Med. 1989;86(1):15–19.

166.

167.

168.

169.

170.

171.

172.

173.

174.

175.

176.

177.

178.

179.

180.

181.

182.

183.

184.

185.

186.

187.

188.

189.

190.

191.

192.

Keighley MR et al. Randomised controlled trial of vancomycin for pseudomembranous colitis and postoperative

diarrhoea. BMJ. 1978;2(6153):1667–1669.

Pettit NN et al. Risk factors for systemic vancomycin exposure following administration of oral vancomycin for

the treatment of Clostridium difficile infection. Pharmacotherapy. 2015;35(2):119–126.

Cornely OA et al. Clinical efficacy of fidaxomicin compared with vancomycin and metronidazole in Clostridium

difficile infections: a meta-analysis and indirect treatment comparison. J Antimicrob Chemother.

2014;69(11):2892–2900.

Louie TJ et al. Fidaxomicin versus vancomycin for Clostridium difficile infection. N Engl J Med.

2011;364(5):422–431.

Crawford T et al. Fidaxomicin: a novel macrocyclic antibiotic for the treatment of Clostridium difficile infection.

Am J Health Syst Pharm. 2012;69(11):933–943.

Bagdasarian N et al. Diagnosis and treatment of Clostridium difficile in adults: a systematic review. JAMA.

2015;313(4):398–408.

Reinke CM et al. ASHP therapeutic position statement on the preferential use of metronidazole for the

treatment of Clostridium difficile-associated disease. Am J Health Syst Pharm. 1998;55:1407.

Weiss K. Toxin-binding treatment for Clostridium difficile: a review including reports of studies with tolevamer.

Int J Antimicrob Agents. 2009;33(1):4–7.

Sullivan A. Probiotics in human infections. J Antimicrob Chemother. 2002;50(5):625–627.

Hickson M et al. Use of probiotic Lactobacillus preparation to prevent diarrhoea associated with antibiotics:

randomised double blind placebo controlled trial. BMJ. 2007;335(7610):80.

Munoz P et al. Saccharomyces cerevisiae Fungemia: an emerging infectious disease. Clin Infect Dis.

2005;40(11):1625–1634.

Jangi S, Lamont JT. Asymptomatic colonization by clostridium difficile in infants: implications for disease in later

life. J Pediatr Gastroenterol Nutr. 2010;51(1):2–7.

Kyne L et al. Asymptomatic carriage of clostridium difficile and serum levels of IgG antibody against toxin A. N

EnglJ Med. 2000;342(6):390–397.

Climo MW. Hospital-wide restriction of clindamycin: effect on the incidence of clostridium difficile-associated

diarrhea and cost. Ann Intern Med. 1998;128(12, part 1):989.

Kyne L et al. Underlying disease severity as a major risk factor for nosocomial clostridium difficile diarrhea.

Infect Control Hosp Epidemiol. 2002;23(11):653–659.

Pepin J. Vancomycin for the treatment of Clostridium difficile Infection: for whom is this expensive bullet really

magic? Clin Infect Dis. 2008;46(10):1493–1498.

Fekety R et al. Recurrent clostridium difficile diarrhea: characteristics of and risk factors for patients enrolled in

a prospective, randomized, double-blinded trial. Clin Infect Dis. 1997;24(3):324–333.

Naggie S et al. Community-associated Clostridium difficile infection: experience of a veteran affairs medical

center in southeastern USA. Infection. 2010;38(4):297–300.

Furuya-Kanamori L et al. Comorbidities, exposure to medications, and the risk of community-acquired

clostridium difficile infection: a systematic review and meta-analysis. Infect Control Hosp Epidemiol.

2015;36(2):132–141.

Johnson S. Treatment of asymptomatic clostridium difficile carriers (Fecal Excretors) with vancomycin or

metronidazole. Ann Intern Med. 1992;117(4):297.

Kazakova SV. A hospital outbreak of diarrhea due to an emerging epidemic strain of clostridium difficile. Arch

Intern Med. 2006;166(22):2518.

McDonald EG et al. Continuous proton pump inhibitor therapy and the associated risk of recurrent clostridium

difficile infection. JAMA Intern Med. 2015;175(5):784–791.

Goldstein EJ et al. Pathway to prevention of nosocomial clostridium difficile infection. Clin Infect Dis.

2015;60(Suppl 2):S148–S158.

Kyne L et al. Health care costs and mortality associated with nosocomial diarrhea due to clostridium difficile.

Clin Infect Dis. 2002;34(3):346–353.

Stevens VW et al. Excess length of stay attributable to clostridium difficile Infection (CDI) in the acute care

setting: a multistate model. Infect Control Hosp Epidemiol. 2015;36(9):1024–1030.

Nanwa N et al. The economic impact of Clostridium difficile infection: a systematic review. Am J Gastroenterol.

2015;110(4):511–519.

Dallal RM et al. Fulminant Clostridium difficile: an underappreciated and increasing cause of death and

complications. Ann Surg. 2002;235(3):363–372.

193.

194.

195.

196.

197.

198.

199.

200.

201.

202.

203.

204.

205.

206.

207.

208.

209.

210.

211.

212.

213.

214.

215.

216.

217.

218.

219.

220.

Zar FA et al. A comparison of vancomycin and metronidazole for the treatment of clostridium difficileassociated diarrhea, stratified by disease severity. Clin Infect Dis. 2007;45(3):302–307.

Rokas KE et al. The addition of intravenous metronidazole to oral vancomycin is associated with improved

mortality in critically ill patients with clostridium difficile infection. Clin Infect Dis. 2015;61(6):934–941.

Lamp KC et al. Pharmacokinetics and pharmacodynamics of the nitroimidazole antimicrobials. Clin

Pharmacokinet. 1999;36(5):353–373.

Friedenberg F et al. Intravenous metronidazole for the treatment of Clostridium difficile colitis. Dis Colon

Rectum. 2001;44(8):1176–1180.

Kleinfeld DI et al. Parenteral therapy for antibiotic-associated pseudomembranous colitis. J Infect Dis.

1988;157(2):389–389.

Bolton RP, Culshaw MA. Faecal metronidazole concentrations during oral and intravenous therapy for antibiotic

associated colitis due to Clostridium difficile. Gut. 1986;27(10):1169–1172.

Guzman R et al. Failure of parenteral metronidazole in the treatment of pseudomembranous colitis. J Infect Dis.

1988;158(5):1146–1146.

Apisarnthanarak A et al. Adjunctive intracolonic vancomycin for severe clostridium difficile colitis: case series

and review of the literature. Clin Infect Dis. 2002;35(6):690–696.

McFarland LV et al. Recurrent clostridium difficile disease: epidemiology and clinical characteristics. Infect

Control Hosp Epidemiol. 1999;20(1):43–50.

Barbut F et al. Epidemiology of recurrences or reinfections of Clostridium difficile-associated diarrhea. J Clin

Microbiol. 2000;38:2386.

Hecht JR, Olinger EJ. Clostridium difficile colitis secondary to intravenous vancomycin. Digest Dis Sci.

1989;34(1):148–149.

Saginur R et al. Colitis associated with metronidazole therapy. J Infect Dis. 1980;141(6):772–774.

Abdelfatah M et al. Factors predicting recurrence of clostridium difficile infection (CDI) in hospitalized patients:

retrospective study of more than 2000 patients. J Investig Med. 2015;63(5):747–751.

Deshpande A et al. Risk factors for recurrent Clostridium difficile infection: a systematic review and metaanalysis. Infect Control Hosp Epidemiol. 2015;36(4):452–460.

Kyne L et al. Association between antibody response to toxin A and protection against recurrent Clostridium

difficile diarrhoea. Lancet. 2001;357(9251):189–193.

McPherson S et al. Intravenous immunoglobulin for the treatment of severe, refractory, and recurrent

clostridium difficile diarrhea. Dis Colon Rectum. 2006;49(5):640–645.

Juang P et al. Clinical outcomes of intravenous immune globulin in severe clostridium difficile-associated

diarrhea. Am J Infect Control. 2007;35(2):131–137.

Pepin J et al. Management and outcomes of a first recurrence of clostridium difficile-associated disease in

Quebec, Canada. Clin Infect Dis. 2006;42(6):758–764.

McFarland LV et al. Breaking the cycle: treatment strategies for 163 cases of recurrent Clostridium difficile

disease. Am J Gastroenterology. 2002;97(7):1769–1775.

Tedesco F et al. Approach to patients with multiple relapses of antibiotic-associated pseudomembranous colitis.

Am J Gastroenterol. 1985;80:867.

Ariano RE et al. The role of anion-exchange resins in the treatment of antibiotic-associated pseudomembranous

colitis. CMAJ. 1990;142:1049.

Pruksananonda P et al. Multiple relapses of Clostridium difficile-associated diarrhea responding to an extended

course of cholestyramine. Pediatr Infect Dis J. 1989;8:175.

Buggy BP et al. Therapy of relapsing clostridium difficile associated diarrhea and colitis with the combination of

vancomycin and rifampin. J Clin Gastroenterol. 1987;9(2):155–159.

McFarland LV. A randomized placebo-controlled trial of saccharomyces boulardii in combination with standard

antibiotics for clostridium difficile disease. JAMA. 1994;271(24):1913.

Surawicz Christina M et al. The Search for a better treatment for recurrent clostridium difficile disease: use of

high dose vancomycin combined with saccharomyces boulardii. Clin Infect Dis. 2000;31(4):1012–1017.

Johnson S et al. Interruption of recurrent clostridium difficile-associated diarrhea episodes by serial therapy with

vancomycin and rifaximin. Clin Infect Dis. 2007;44(6):846–848.

Garey KW et al. Rifaximin in treatment of recurrent clostridium difficile-associated diarrhea: an uncontrolled

pilot study. J Clin Gastroenterol. 2009;43(1):91–92.

Gallagher JC et al. Clinical and economic benefits of fidaxomicin compared to vancomycin for clostridium

difficile infection. Antimicrob Agents Chemother. 2015;59(11):7007–7010.

221.

222.

223.

224.

225.

Nathwani D et al. Cost-effectiveness analysis of fidaxomicin versus vancomycin in Clostridium difficile

infection. J Antimicrob Chemother. 2014;69(11):2901–2912.

Drekonja D et al. Fecal microbiota transplantation for clostridium difficile infection: a systematic review. Ann

Intern Med. 2015;162(9):630–638.

Han S et al. Fecal microbiota transplant: treatment options for clostridium difficile infection in the intensive care

unit. J Intensive Care Med. 2016;31(9):577–586.

Aroniadis OC et al. Long-term follow-up study of fecal microbiota transplantation for severe and/or complicated

clostridium difficile infection: a multicenter experience. J Clin Gastroenterol. 2016;50(5):398–402.

Varier RU et al. Cost-effectiveness analysis of fecal microbiota transplantation for recurrent Clostridium difficile

infection. Infect Control Hosp Epidemiol. 2015;36:438–444.

p. 1468

Acute cholecystitis is the acute inflammation of the gallbladder,

presenting with fever, prolonged abdominal pain, and positive Murphy’s

sign of the right upper quadrant followed by nausea and vomiting. Acute

cholangitis is an acute inflammation of the common bile duct with

accompanying classic Charcot’s triad: fever, jaundice, and right upper

quadrant pain.

Case 70-1 (Question 1)

Empiric therapy for severe biliary tract infections may include

combination therapy with either ciprofloxacin, levofloxacin, or cefepime

plus metronidazole or monotherapy with piperacillin/tazobactam or an

antipseudomonal carbapenem (imipenem/cilastatin, meropenem,

doripenem) if multidrug-resistant gram-negative rods are suspected.

Case 70-1 (Question 3)

Spontaneous bacterial peritonitis (SBP) is largely a monomicrobial

infection commonly caused by aerobic enteric gram-negative rods, such

as Escherichia coli and Klebsiella pneumoniae.

Case 70-2 (Question 1)

Empiric therapy for SBP may include a third-generation cephalosporin

such as ceftriaxone or cefotaxime or parenteral fluoroquinolone with

activity versus Streptococcus pneumoniae. Length of therapy depends

on the reduction of polymorphonuclear leukocyte counts (<250 cells/μL)

in peritoneal fluid, generally occurring in 5 days or fewer.

Case 70-2 (Question 2)

For secondary peritonitis, empiric antimicrobial therapy for communityacquired infections is routinely initiated without obtaining Gram stain and

culture. Blood cultures may be useful, however, in health care–

associated infections for detection of gram-positive cocci, yeast, or

multidrug-resistant pathogens.

Case 70-4 (Question 2)

Therapy for secondary peritonitis should include antimicrobial coverage

directed at gram-negative bacteria and anaerobes. Therapy may include

(a) certain extended-spectrum β-lactams or β-lactamase inhibitor

combinations, (b) carbapenems, or (c) a fluoroquinolone with

metronidazole.

Case 70-4 (Question 3)

Duration of therapy for intra-abdominal infections typically ranges from

4 to 7 days. Clinical response and source control influence duration of

therapy.

Case 70-4 (Question 4)

Acute penetrating abdominal trauma requires a short course (<24 hours) Case 70-7 (Question 2)

of antimicrobial therapy. Delay of therapy or development of infection

necessitates a 5 to 7-day course of therapy.

INTRODUCTION

Despite the introduction of new antimicrobial agents and improvements in diagnostic

and surgical techniques, the treatment of intra-abdominal infections remains a

therapeutic challenge. However, advances in radiographic and interventional

techniques with timely source control, improved fluid and nutritional management,

and the selection of appropriate antimicrobial agents have decreased mortality in

intra-abdominal infections.

1

Intra-abdominal infections are those contained within the peritoneal cavity, which

extends from the undersurface of the diaphragm to the floor of the pelvis or the

retroperitoneal space. Intra-abdominal infections can present as a localized infection

(e.g., appendicitis), a diffuse inflammation throughout the peritoneum (peritonitis), or

as abscesses, which can form

p. 1469

p. 1470

anywhere within the abdomen, between bowel loops, or in solid organs such as the

liver, biliary tract, spleen, pancreas, or female pelvic organs.

Empiric antimicrobial therapy should be initiated as soon as an intra-abdominal

infection is suspected. Therapy should be selected on the basis of the suspected

pathogens; however, antimicrobial therapy alone is insufficient, especially in the

setting of diffuse peritonitis. Source control is a term used to involve all physical

measures needed to eradicate an infection, such as debridement of necrotic tissue or

drainage of an abscess or fluid collection. If adequate source control is delayed,

bacteremia, multiple organ failure, and mortality are more likely to occur.

2

Normal Gastrointestinal Flora

The stomach of fasting individuals contains very few bacteria (i.e., <100 colonyforming units [CFU]/mL) as a result of gastric motility and the bactericidal activity of

normal acidic gastric fluid.

1 The bacterial population of the stomach can be altered

by drugs or diseases that increase gastric pH or decrease gastric motility. Not

surprisingly, patients with bleeding or obstructing duodenal ulcers, gastric ulcers,

gastric carcinomas, or those receiving proton pump inhibitors or histamine-2

receptor antagonists have an increased number of oral bacteria colonizing the

stomach.

The upper small intestine (duodenum and jejunum) usually contains relatively few

bacteria and harbors mainly oral flora. The lower small intestine serves as a

transitional zone between the sparsely populated stomach and the abundant microbial

flora of the colon.

3

,

4

In the ileum, facultative gram-negative and gram-positive species, as well as

obligate anaerobes, are encountered. As the distal ileum is approached, the quantity

and variety of bacteria increase. Substantial numbers of anaerobic bacteria are

present, including Bacteroides species, as well as Escherichia coli and Enterococcus

species.

1

,

3

In the large bowel, anaerobic bacteria, particularly Bacteroides species,

predominate. In the distal colon, bacterial counts average 10

10 CFU/mLof feces, with

anaerobes outnumbering other organisms by a ratio of 1,000 to 10,000:1.

3 Among the

facultative aerobes, E. coli is the most frequently isolated species.

1

,

3 Given these

differences in regional microflora populations, it is not surprising that trauma to the

colon carries a much higher risk of intra-abdominal infection in comparison with the

stomach or jejunum5

(Fig. 70-1; Table 70-1).

Resistant flora are more common in health care–associated infections, including

Pseudomonas aeruginosa, Acinetobacter species, extended-spectrum β-lactamases

(ESBLs) and carbapenemase producing E. coli and Klebsiella species, methicillinresistant Staphylococcus aureus (MRSA), enterococci, and Candida species.

6–10

Figure 70-1 Microflora of the gastrointestinal tract.

p. 1470

p. 1471

Table 70-1

Common Pathogens in Intra-Abdominal Infection

Disease Common Pathogens Comments

Cholecystitis, cholangitis E. coli, Klebsiella pneumoniae,

Enterococcus, and anaerobes

Enterococcus species and

anaerobes are less frequently

isolated, but they have been

associated with nosocomial

infections and chronic surgical

infections, particularly in patients

receiving broad-spectrum

antimicrobials.

Primary peritonitis E. coli, K. pneumoniae,

Streptococcus pneumoniae,

occasional anaerobes

Also known as spontaneous

bacterial peritonitis presented in

cirrhotic patients. Often

monomicrobial. Anaerobes less

likely than aerobes.

Secondary peritonitis E. coli, Bacteroides fragilis,

Enterococcus, and anaerobes

Generally polymicrobial with both

aerobic and anaerobic pathogens.

Enterococcus species should be

considered in the setting of health

care–associated infections.

Chronic ambulatory peritoneal

dialysis

Staphylococcus epidermidis,

Staphylococcus aureus,

diphtheroids, gram-negative rods

Dwell time of exchange with

intraperitoneal antibiotics must be a

minimum of 6 hours.

INFECTIONS OF THE BILIARY TRACT

Cholecystitis and Cholangitis

Cholecystitis and cholangitis originate as inflammatory conditions, usually as a result

of obstruction in the gallbladder or common bile duct. The obstruction is typically

caused by the presence of stones. The biliary tract is sterile under normal conditions,

and the flow of bile, along with its bacteriostatic properties, functions to maintain the

sterility. Infection typically occurs as a secondary event to obstruction.

4

,

5

Acute cholecystitis is the acute inflammation of the gallbladder. The presence of

gallstones in greater than 90% of cases prevents the outflow of gallbladder drainage

by obstructing the neck of the gallbladder, Hartmann’s pouch, or the cystic duct.

4 The

obstruction causes an increase in intraluminal pressure, gallbladder distension, and

edema, which triggers an acute inflammatory response. The potential consequences

of this obstruction and inflammation include infection, ischemia, perforation, and

necrosis.

4

,

11 The reduction in gallbladder outflow leads to biliary stasis, which

provides an ideal environment for bacterial proliferation and subsequent infection

(Fig. 70-2).

Acute cholangitis is an acute inflammation of the common bile duct. The most

common cause of cholangitis in the United States is common bile duct obstruction as

a consequence of choledocholithiasis. Less common causes include neoplastic

obstruction, postoperative obstruction after biliary intervention, benign strictures,

and primary sclerosing cholangitis.

4 The decrease in biliary outflow results in biliary

stasis and bacterial proliferation. Infection results in a rapid rise in biliary pressure,

which facilitates the spread of bacteria into lymphatics and the bloodstream via

alterations in membrane permeability. In comparison with cholecystitis, acute

cholangitis generally carries a poorer prognosis.

4

,

12

Figure 70-2 Pathogenesis of acute cholecystitis.

CLINICAL PRESENTATION AND DIAGNOSIS

CASE 70-1

QUESTION 1: D.S., a 54-year-old man, presents to the hospital with a 1-week history of abdominal pain and

tenderness, localized to the right upper quadrant, fever of 38.9°C, and chills. He complains of nausea, with three

episodes of emesis occurring in the past 24 hours. D.S. appears jaundiced and reports dark-colored urine. His

laboratory values are the following:

White blood cell (WBC) count, 17 × 10

3

/μL

Serum creatinine (SCr), 1.1 mg/dL

Total bilirubin, 6 mg/dL

Alkaline phosphatase, 270 U/L

What evidence of cholangitis exists in D.S.? How does the presentation of cholecystitis differ from

cholangitis?

p. 1471

p. 1472

The clinical manifestations of acute cholangitis vary, but the classic presentation

involves Charcot’s triad, which consists of fever, jaundice, and right upper quadrant

abdominal pain. Fever is present in 90% of cases, and jaundice and abdominal pain

occur in 60% to 70% of cases.

4 A smaller percentage of patients, generally with

gram-negative septicemia, may present with changes in mental status and

hypotension.

4 Laboratory findings of acute cholangitis include leukocytosis, elevated

bilirubin (>2 mg/dL), and alkaline phosphatase, and mildly elevated liver

transaminases.

4

,

13 The clinical signs and symptoms of cholangitis, as exemplified by

D.S., include high fevers (38.9°C), chills, jaundice, and right upper quadrant

abdominal pain. Laboratory evidence supportive of cholangitis includes leukocytosis

with WBC of 17 × 10

3

/μL, increased bilirubin of 6 mg/dL, and elevated alkaline

phosphatase of 270 U/L.

The clinical presentation of acute cholecystitis involves fever and prolonged

constant abdominal pain typically localized to the right upper quadrant, followed by

nausea and vomiting. On physical examination, tenderness in the right upper quadrant

and a positive Murphy’s sign (inspiration is inhibited by pain on palpation) are often

present.

4

,

11 Laboratory findings include leukocytosis with increased neutrophils (left

shift) and mild elevations in transaminases. Jaundice is less common in patients with

cholecystitis (bilirubin <4 mg/dL) compared with cholangitis.

4

,

11

Diagnostic imaging for acute cholecystitis and cholangitis involves

ultrasonography. Findings for acute cholecystitis may reveal pericholecystic fluid,

distension of the gallbladder, thickening of the gallbladder wall, stones, and a

sonographic Murphy’s sign.

14 Ultrasound is less sensitive for choledocholithiasis, but

it may reveal biliary dilation with stones. Hepatobiliary scintigraphy is another

diagnostic tool used to identify obstruction of the cystic duct. After intravenous

injection with technetium hepatobiliary iminodiacetic acid (Tc-HIDA), failure to

observe gallbladder filling suggests acute cholecystitis.

6

,

14 Computed tomography

(CT) imaging may be superior in determining the extent of biliary obstruction.

4

Endoscopic retrograde cholangiopancreatography (ERCP) is an alternative mode of

imaging for cholangitis.

4

,

13

ETIOLOGY

CASE 70-1, QUESTION 2: What are the most likely organisms causing infection in D.S.? What clinical

specimens are helpful to identify the causative pathogen(s)?

The most common pathogens associated with acute cholangitis include E. coli,

Klebsiella species, and Enterobacter species. However, P. aeruginosa, skin flora

(Staphylococcus species, Streptococcus species), and oropharynx bacteria may be

implicated. Anaerobic organisms, commonly Bacteroides species, are associated

with approximately 15% of infections, particularly in elderly patients undergoing

biliary tract surgery.

4

,

15 The etiology of cholecystitis typically involves E. coli,

Klebsiella species, and Enterococcus species. Anaerobes are less commonly

isolated (Table 70-1).

4

,

11

As described above, the stasis of bile flow results in proliferation of bacteria

within the gallbladder or common bile duct. Acute cholangitis results in increased

pressure within the common bile duct with dissemination of bacteria from the biliary

tree to the bloodstream. Blood cultures may be positive in up to 40% of patients with

symptomatic acute cholangitis.

4 Acute cholecystitis may reveal positive bile cultures

in 20% to 75% of patients with symptomatic disease, but the utility of bile cultures

has yet to be determined.

3

In contrast to cholangitis, bacteremia is unlikely with

cholecystitis. The choice of antibacterial agents is largely empiric covering the

aforementioned common causative pathogens.

TREATMENT

Definitive therapy for both cholangitis and cholecystitis must involve source control,

using surgery, percutaneous drainage, or endoscopic intervention. Empiric

antibacterial therapy should be added owing to the likelihood of secondary infection

and to prevent complications. Supportive measures include fluid and electrolyte

supplementation and analgesia.

4

,

5

,

11

,

13

To decompress or drain the biliary ductal system in cholangitis, ERCP is used

with a success rate of greater than 90% in the treatment of cholangitis.

4 Alternatively,

percutaneous transhepatic biliary drainage or endoscopic sphincterotomy can be used

to drain the contents. Open surgery is a less favored option to decompress the biliary

tree as it is associated with increased mortality.

4 Acute symptomatic cholecystitis

requires removal of the gallbladder (cholecystectomy). The procedure of choice is

laparoscopic cholecystectomy within 48 to 72 hours of onset of symptoms. Early

intervention is associated with decreased morbidity and length of hospital stay

compared with delayed surgical intervention.

4

,

11

In high-risk patients, in whom the

risks outweigh the benefits of early surgery, percutaneous cholecystostomy can be

performed to drain the contents of the gallbladder. The procedure improves the

clinical symptoms in 75% to 90% of patients who cannot undergo surgery.

Cholecystectomy, however, should take place once the patient is stable enough for

surgery.

4

ETIOLOGY

CASE 70-1, QUESTION 3: Based on the most likely causative pathogen(s), what empiric antimicrobial

therapy is recommended for D.S., given a diagnosis of cholangitis?

Empiric antimicrobial therapy for acute cholangitis and cholecystitis should be

initiated after blood cultures have been collected. The empiric choice of therapy

should cover enteric gram-negative pathogens, particularly E. coli (Table 70-2).

5

,

6

In

general, the addition of anaerobic coverage is not mandatory for cholecystitis;

however, it should be considered in the setting of severe cases such as acute

cholangitis, biliary-enteric anastomosis, or health care–associated infections.

6

Empiric coverage for Enterococcus species should be considered for patients at risk

for health care–associated infections, such as immunocompromised patients, patients

who have required long-term hospitalization, patients who have received broadspectrum antimicrobial therapy, or patients with valvular heart disease or prosthetic

intravascular materials.

6 Antimicrobial therapy must be guided by drug

pharmacokinetics and pharmacodynamics, local resistance patterns, and patient

factors, such as drug allergies, renal or hepatic dysfunction, and cost.

Considering their activity versus multidrug-resistant gram-negative bacilli,

carbapenems generally should be reserved for infection caused by these pathogens.

Vancomycin may be added empirically in health care–associated biliary infections in

which ampicillin-resistant enterococci and MRSA are of concern. Linezolid or

daptomycin may be considered in cases of known vancomycin-resistant Enterococcus

species (VRE). Empiric broad-spectrum antimicrobial regimens should be narrowed,

when possible, based on culture and susceptibility findings. For mild-to-moderate

community-acquired acute cholecystitis, monotherapy with cefazolin, cefuroxime, or

ceftriaxone is recommended. Because of a wide prevalence of ampicillin–sulbactam

resistant E. coli, this agent is no longer recommended for empiric use.

6

,

15 Resistance

to fluoroquinolones is problematic with unpredictable susceptibility of E. coli to

ciprofloxacin and levofloxacin.

16 Current guidelines for complicated intra-abdominal

infections (cIAI) suggest fluoroquinolones be considered for therapy when

institutional antibiograms indicate greater than 90% susceptibility of E. coli.

6 Use of

aminoglycosides is not routinely recommended because of the need for therapeutic

drug monitoring and potential nephrotoxicity and ototoxicity.

6 Furthermore, in

comparison with other regimens, aminoglycoside-based regimens are inferior in the

treatment of intra-abdominal infections.

17 However, some argue the decreased

efficacy of aminoglycosides has been caused by inadequate dosing. An appropriate

empiric regimen for D.S. would be piperacillin–tazobactam 3.375 g intravenously

(IV) every 6 hours.

p. 1472

p. 1473

Table 70-2

Treatment Options for Intra-Abdominal Infections

5,6,66,67

Infection Agents and Regimens Agent Dosing

d

Mild-to-moderate in severity:

Community-acquired perforated or

abscessed appendicitis

Or

Acute cholecystitis

Single agent

Cefoxitin

Ertapenem

Moxifloxacin

a

Tigecycline

Combination

Cefazolin

Or

Ceftriaxone

Or

Cefotaxime

Or

Ciprofloxacin

Or

Levofloxacin

a + Metronidazole

Cefoxitin 2 g IV every 6 hours

Ertapenem 1 g IV every 24 hours

Tigecycline 100 mg initially then 50

mg IV every 12 hours

Cefazolin 1–2 g IV every 8 hours

Ceftriaxone 1–2 g IV every 12–24

hours

Cefotaxime 1–2 g IV every 6–8

hours

Ciprofloxacin 400 mg IV every 12

hours

Levofloxacin 750 mg IV every 24

hours

Moxifloxacin 400 mg IV every 24

hours

Metronidazole 500 mg IV every 8–

High risk in severity:

Community-acquired acute

cholecystitis with severe

physiologic disturbance, advanced

age, or immunocompromised

state

Or

Acute cholangitis associated with

biliary-enteric anastomosis

Or

Health care–associated intraabdominal infections

Single agent

Imipenem–cilastatin

Meropenem

Doripenem

Piperacillin–tazobactam

Combination

Cefepime

Or

Ceftazidime

Or

Ciprofloxacin

Or

Levofloxacin

a + Metronidazole

12 hours

Cefepime 1-2 g IV every 8 hours

and

Ceftazidime 1-2 g IV every 8-12

hours

Piperacillin–tazobactam 3.375 g IV

every 4–6 hours or 4.5 g IV every

6 hours

e

Imipenem–cilastatin 500 mg IV

every 6 hours or 1 g every 8 hours

Meropenem 1 g IV every 8 hours

Doripenem 500 mg IV every 8

hours

Vancomycin

b 15–20 mg/kg IV

every 8–12 hours (normal renal

function)

Aztreonam

c 1–2 g IV every 6–8

hours

Ceftazidime/avibactam 2.5 g IV

every 8 hours

or

Ceftolozane/tazobactam 1.5 g IV

every 8 hours

+

Metronidazole 500 mg IV every 8–

12 hours

aSelection of fluoroquinolones should be based on local antibiogram or susceptibility reports.

bMay be added to each regimen in the setting of health care–associated intra-abdominal infections (e.g.,

methicillin-resistant Staphylococcus aureus or ampicillin-resistant enterococcal infection).

cFor patients who are intolerant of β-lactams, aztreonam may be considered as an alternative agent for gramnegative coverage.

dnormal renal function dosing

eExtended infusion: 3.375 g IV every 8 hours over 4 hours

IV, intravenous.

BILIARY CONCENTRATIONS

CASE 70-1, QUESTION 4: The physician caring for D.S. questions the need for an antibiotic that

concentrates in the bile. Is there a benefit to using an antibiotic that is extensively excreted into bile?

Common bile duct obstruction prevents the entry of antibiotics into the bile, and

the need for high biliary concentrations of antibiotics in the treatment of cholangitis

has been debated.

18 Nagar and Berger

19 concluded that a number of antibiotics with

excellent in-vitro susceptibility, but poor biliary excretion, are still clinically

effective. Biliary antibacterial concentrations do not correlate with an improved

clinical outcome.

4 Highly biliary-excreted versus moderately biliary-excreted

antibiotics have been compared.

18 Serum concentrations are more important than

biliary levels in reducing the septic complications of biliary tract surgery.

Furthermore, biliary excretion of any antibiotic is minimal in the presence of

obstruction.

18

The treatment of D.S.’s biliary tract infection must include biliary drainage.

Piperacillin–tazobactam should be continued for 4 to 7 days.

PRIMARY PERITONITIS

Peritonitis is inflammation of the peritoneum as a result of infectious or chemical

inflammation within the peritoneal cavity.

3

,

20

Infectious peritonitis can be classified

as primary, secondary, or tertiary. Primary peritonitis involves the development of

infection in the peritoneal cavity in the absence of intra-abdominal pathology.

20

,

21

Secondary peritonitis classically results from contamination of the peritoneum with

gastrointestinal (GI) or genitourinary microorganisms as a result of the loss of

mucosal barrier integrity. Tertiary peritonitis is clinical peritonitis along with signs

of sepsis and multi-organ dysfunction persisting or recurring after the treatment of

secondary peritonitis.

3

,

20

Primary peritonitis is also known as spontaneous bacterial peritonitis (SBP) and is

most frequently identified in adults with cirrhosis and ascites. Nearly 10% to 30% of

hospitalized patients with cirrhosis and ascites have SBP.

3

,

21

,

22 Primary peritonitis is

associated with post-necrotic cirrhosis, chronic acute hepatitis, acute viral hepatitis,

congestive heart failure, metastatic malignancy, or systemic lupus erythematosus.

3

,

20

p. 1473

p. 1474

Spontaneous Bacterial Peritonitis in Cirrhotic Patients

CLINICAL MANIFESTATIONS AND DIAGNOSIS

Patients with SBP may present with fever, signs of peritonitis, including abdominal

pain, altered mental status, changes in GI motility, nausea, vomiting, diarrhea, or

ileus.

22 Fever is common, occurring in 50% to 80% of patients.

3 Some patients have

an atypical presentation or even lack symptoms.

22 The diagnosis of SBP is made on

clinical presentation and examination of the peritoneal fluid via paracentesis. The

ascitic fluid is tested for cell counts with WBC differential, and the presence of

microorganisms is based on Gram stain and culture. Ascitic fluid with an elevated

polymorphonuclear (PMN) count (≥250/μL) is diagnostic for SBP.

22

Comments

Search This Blog

Archive

Show more

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

علاقة البيبي بالفراولة بالالفا فيتو بروتين

التغيرات الخمس التي تحدث للجسم عند المشي

إحصائيات سنة 2020 | تعداد سكَان دول إفريقيا تنازليا :

ما هو الليمونير للأسنان ؟

ACUPAN 20 MG, Solution injectable

CELEPHI 200 MG, Gélule

الام الظهر

VOXCIB 200 MG, Gélule

ميبستان

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

Kana Brax Laberax

TRIPASS XR تري باس

PARANTAL 100 MG, Suppositoire بارانتال 100 مجم تحاميل

الكبد الدهني Fatty Liver

الم اسفل الظهر (الحاد) الذي يظهر بشكل مفاجئ bal-agrisi

SEDALGIC 37.5 MG / 325 MG, Comprimé pelliculé [P] سيدالجيك 37.5 مجم / 325 مجم ، قرص مغلف [P]

نمـو الدمـاغ والتطـور العقـلي لـدى الطفـل

CELEPHI 200 MG, Gélule

أخطر أنواع المخدرات فى العالم و الشرق الاوسط

Archive

Show more