lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile...

29
Quality Department Guidelines for Clinical Care 1 Michigan Medicine C. difficile Guideline December 2016 Clostridium difficile Infection Guideline Team Team Leads Tejal N Gandhi MD Infectious Diseases Krishna Rao, MD Infectious Diseases Team Members Gregory Eschenauer, PharmD Pharmacy John Y Kao, MD Gastroenterology Andrea H Kim, MD Internal Medicine Lena M Napolitano, MD Surgery F Jacob Seagull, PhD Leaning Heath Sciences David M Somand, MD Emergency Medicine Kathleen B To, MD Surgery Alison C Tribble, MD Pediatric Infectious Diseases Amanda M Valyko, MPH, CIC Infection Control Michael E Watson Jr, MD, PhD Pediatric Infectious Diseases Consultants: Meghan A. Arnold, MD Pediatric Surgery Daniel H Teitelbaum, MD Pediatric Surgery Laraine Lynn Washer, MD Infection Diseases Initial Release: December 2016 Inpatient Clinical Guidelines Oversight Megan R Mack, MD David H Wesorick, MD F Jacob Seagull, PhD Literature search service Taubman Health Sciences Library For more information: 734- 936-9771 © Regents of the University of Michigan These guidelines should not be construed as including all proper methods of care or excluding other acceptable methods of care reasonably directed to obtaining the same results. The ultimate judgment regarding any specific clinical procedure or treatment must be made by the physician in light of the circumstances presented by the patient. Clostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with a primary or recurrent episode of Clostridium difficile infection (CDI) Objectives: 1. Provide a brief overview of the epidemiology of, and risk factors for development of CDI 2. Provide guidance regarding which patients should be tested for CDI, summarize merits and limitations of available diagnostic tests, and describe the optimal approach to laboratory diagnosis 3. Review the most effective treatment strategies for patients with CDI including patients with recurrences or complications Key points for adult patients: Diagnosis Definitive diagnosis of CDI requires either the presence of toxigenic C. difficile in stool with compatible symptoms, or clinical evidence of pseudomembranous colitis (Table 2, Figure 4). Once identified, CDI should be classified according to severity ( Table 3). Although risk factors for CDI (Table 1) should guide suspicion for CDI, testing should be ordered only when indicated (Figure 1). [IC] Choice of test should be guided by a multi-step algorithm for the rapid diagnosis of CDI ( Figure 2). [IIC] Single-step PCR testing (not part of the UMHS algorithm) occurs as part of the new Biofire test panel for gastrointestinal pathogens and should not be used if CDI is suspected. [IIIB] However, if C. difficile is detected as part of this panel and the patient’s symptoms are compatible with CDI, then treatment is appropriate and additional testing is unnecessary. [IIC] Patients who are asymptomatic, actively being treated or completed treatment for CDI with clinical improvement in symptoms, or have post-infectious irritable bowel syndrome after CDI should not undergo testing for CDI. [IIIC] Treatment: (See Figure 3 and Table 4) Mild-Moderate CDI: Patient does not meet criteria for “severe” or “complicated” CDI metronidazole 500 mg PO TID for 10-14 days [IIB] OR In patients with metronidazole allergy, pregnant, nursing, or on warfarin therapy: vancomycin 125 mg PO QID for 10-14 days. [IB] Severe CDI: Patients with WBC ≥ 15K, Cr ≥ 1.5x baseline, Age ≥ 65, ANC ≤ 500, Albumin ≤ 2.5, SOT/BMT < 100 days, chronic GVHD (BMT), treatment of rejection in the preceding 2 months (SOT), Small bowel CDI, or inflammatory bowel disease vancomycin 125 mg PO QID for 10-14 days [IA] Complicated CDI: Patients with septic shock, ileus, toxic megacolon, peritonitis, or bowel perforation Triple therapy=vancomycin 500 mg PO QID, metronidazole 500 mg IV every 8 hours, and vancomycin enema every 6 hours (in patients with ileus, bowel obstruction or toxic megacolon) [IB] Consult infectious diseases Consult surgery to assist in management including possible surgical intervention (Table 4, Figure 5). Recurrent CDI: Recurrent symptoms and positive testing for toxigenic C. difficile within 8 weeks of prior episode First recurrence: Classify as “mild-moderate” “severe,” or “complicated,” and treat accordingly [IC] Second or multiple recurrences (third or more episode of CDI): Consult infectious diseases vancomycin PO (dose, need for concurrent IV metronidazole/vancomycin enemas depends on disease classification as noted above) for 10-14 days then taper to 125 mg PO BID for 7 days, 125 mg PO daily for 7 days, and then pulse with 125 mg PO once every 2-3 days for 2-8 weeks. [IIC] OR fidaxomicin 200 mg PO BID for 10 days (with approval from the infectious diseases consult service). [IIE] * Strength of recommendation: I = generally should be performed; II = may be reasonable to perform; III = generally should not be performed. Level of evidence supporting a diagnostic method or an intervention: A = systematic reviews of randomized controlled trials with or without meta-analysis, B = randomized controlled trials, C = systematic review of non-randomized controlled trials or observational studies, non-randomized controlled trials, group observation studies (cohort, cross-sectional, case-control), D = individual observation studies (case study/case series), E = expert opinion regarding benefits and harm

Transcript of lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile...

Page 1: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

Quality Department

Guidelines for Clinical Care

1 Michigan Medicine C. difficile Guideline December 2016

Clostridium difficile

Infection Guideline

Team

Team Leads

Tejal N Gandhi MD

Infectious Diseases

Krishna Rao, MD

Infectious Diseases

Team Members

Gregory Eschenauer, PharmD

Pharmacy

John Y Kao, MD Gastroenterology

Andrea H Kim, MD Internal Medicine

Lena M Napolitano, MD

Surgery

F Jacob Seagull, PhD

Leaning Heath Sciences

David M Somand, MD Emergency Medicine

Kathleen B To, MD

Surgery

Alison C Tribble, MD

Pediatric Infectious

Diseases

Amanda M Valyko, MPH,

CIC

Infection Control

Michael E Watson Jr, MD,

PhD

Pediatric Infectious Diseases

Consultants:

Meghan A. Arnold, MD Pediatric Surgery

Daniel H Teitelbaum, MD

Pediatric Surgery

Laraine Lynn Washer, MD

Infection Diseases

Initial Release:

December 2016

Inpatient Clinical

Guidelines Oversight Megan R Mack, MD

David H Wesorick, MD

F Jacob Seagull, PhD

Literature search service

Taubman Health Sciences

Library

For more information: 734- 936-9771

© Regents of the

University of Michigan These guidelines should not be

construed as including all proper

methods of care or excluding

other acceptable methods of care

reasonably directed to obtaining

the same results. The ultimate

judgment regarding any specific

clinical procedure or treatment

must be made by the physician in

light of the circumstances

presented by the patient.

Clostridium difficile Infection

in Adults and Children

Patient population: Adult and pediatric patients with a primary or recurrent episode of Clostridium difficile infection (CDI)

Objectives:

1. Provide a brief overview of the epidemiology of, and risk factors for development of CDI 2. Provide guidance regarding which patients should be tested for CDI, summarize merits and

limitations of available diagnostic tests, and describe the optimal approach to laboratory diagnosis 3. Review the most effective treatment strategies for patients with CDI including patients with

recurrences or complications

Key points for adult patients:

Diagnosis

• Definitive diagnosis of CDI requires either the presence of toxigenic C. difficile in stool with compatible symptoms, or clinical evidence of pseudomembranous colitis (Table 2, Figure 4).

• Once identified, CDI should be classified according to severity (Table 3). • Although risk factors for CDI (Table 1) should guide suspicion for CDI, testing should be ordered

only when indicated (Figure 1). [IC] • Choice of test should be guided by a multi-step algorithm for the rapid diagnosis of CDI (Figure 2).

[IIC] • Single-step PCR testing (not part of the UMHS algorithm) occurs as part of the new Biofire test

panel for gastrointestinal pathogens and should not be used if CDI is suspected. [IIIB] However, if C. difficile is detected as part of this panel and the patient’s symptoms are compatible with CDI, then treatment is appropriate and additional testing is unnecessary. [IIC]

• Patients who are asymptomatic, actively being treated or completed treatment for CDI with clinical improvement in symptoms, or have post-infectious irritable bowel syndrome after CDI should not undergo testing for CDI. [IIIC]

Treatment: (See Figure 3 and Table 4)

Mild-Moderate CDI: Patient does not meet criteria for “severe” or “complicated” CDI • metronidazole 500 mg PO TID for 10-14 days [IIB]

OR • In patients with metronidazole allergy, pregnant, nursing, or on warfarin therapy: vancomycin 125

mg PO QID for 10-14 days. [IB]

Severe CDI: Patients with WBC ≥ 15K, Cr ≥ 1.5x baseline, Age ≥ 65, ANC ≤ 500, Albumin ≤ 2.5, SOT/BMT < 100 days, chronic GVHD (BMT), treatment of rejection in the preceding 2 months (SOT), Small bowel CDI, or inflammatory bowel disease • vancomycin 125 mg PO QID for 10-14 days [IA]

Complicated CDI: Patients with septic shock, ileus, toxic megacolon, peritonitis, or bowel perforation • Triple therapy=vancomycin 500 mg PO QID, metronidazole 500 mg IV every 8 hours, and

vancomycin enema every 6 hours (in patients with ileus, bowel obstruction or toxic megacolon) [IB]

• Consult infectious diseases • Consult surgery to assist in management including possible surgical intervention (Table 4, Figure 5).

Recurrent CDI: Recurrent symptoms and positive testing for toxigenic C. difficile within 8 weeks of prior episode

First recurrence: • Classify as “mild-moderate” “severe,” or “complicated,” and treat accordingly [IC]

Second or multiple recurrences (third or more episode of CDI): • Consult infectious diseases • vancomycin PO (dose, need for concurrent IV metronidazole/vancomycin enemas depends on

disease classification as noted above) for 10-14 days then taper to 125 mg PO BID for 7 days, 125 mg PO daily for 7 days, and then pulse with 125 mg PO once every 2-3 days for 2-8 weeks. [IIC] OR

• fidaxomicin 200 mg PO BID for 10 days (with approval from the infectious diseases consult service). [IIE]

* Strength of recommendation:

I = generally should be performed; II = may be reasonable to perform; III = generally should not be performed.

Level of evidence supporting a diagnostic method or an intervention: A = systematic reviews of randomized controlled trials with or without meta-analysis, B = randomized controlled trials, C = systematic review of non-randomized controlled trials or observational studies, non-randomized controlled trials, group

observation studies (cohort, cross-sectional, case-control), D = individual observation studies (case study/case series), E =

expert opinion regarding benefits and harm

Page 2: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

2 Michigan Medicine C. difficile Guideline December 2016

Key points for pediatric patients ≤ 18 years of age:

Diagnosis • The decision to test children for CDI is complicated given a high rate of asymptomatic carriage, especially in infants <

12 months of age. Although risk factors for CDI should guide suspicion for CDI, testing should be ordered only when indicated (Figure 1). [IC] Indications and contraindications for testing pediatric patients are included in Figure 1, but testing is rarely indicated or recommended for infants < 12 months [IIC] and consultation with pediatric ID is recommended. Children from 12 months to 36 months of age may be diagnosed with CDI if no alternative etiology for diarrhea is identified and with positive diagnostic testing. [IIC]

• Definitive diagnosis of CDI requires either the presence of toxigenic C. difficile in stool with other symptoms, or clinical evidence of pseudomembranous colitis (Table 2). Choice of test should be guided by a multi-step algorithm for the rapid diagnosis of CDI (Figure 2). Single-step PCR testing (not part of the UMHS algorithm) occurs as part of the new Biofire test panel for gastrointestinal pathogens and should not be used if CDI is suspected. [IIIE] However, if C. difficile is detected as part of this panel and the patient’s symptoms are compatible with CDI, then treatment is appropriate and additional testing is unnecessary. [IIC] Patients who are asymptomatic, actively being treated or completed treatment for CDI with clinical improvement in symptoms, or have post-infectious irritable bowel syndrome after CDI should not undergo testing for CDI. [IIIC]

• Once identified, CDI should be classified according to severity (Table 3). Certain pediatric conditions are associated with severe CDI and should be treated as such (see Table 3). [IIC]

Treatment: (See Figure 3 for general strategy and Table 3 for pediatric-specific dosing recommendations)

Mild-Moderate CDI: Patient does not meet criteria for “severe” or “complicated” CDI • metronidazole 7.5 mg/kg/dose PO QID for 10 days, maximum 500 mg/dose [IIC] OR • In patients with metronidazole allergy, pregnant, nursing, or on warfarin therapy, or who fail to improve after 3-5 days

of PO metronidazole therapy: vancomycin 10 mg/kg/dose PO QID, up to maximum 125 mg/dose x 10 days. [IIC] Severe CDI: Pediatric patients with ≥ 2 lab criteria (WBC ≥ 15K, Cr ≥ 1.5x baseline, ANC ≤ 500, Albumin ≤ 2.5) OR ANY

high-risk condition with Hirschsprung’s Disease or other intestinal dysmotility disorder, neutropenia from leukemia or other

malignancy, inflammatory bowel disease, SOT/BMT < 100 days • vancomycin 10 mg/kg/dose PO QID, up to maximum 125 mg/dose x 10 days [IB]

Complicated CDI: Patients with septic shock, ICU admission within 2 days of CDI diagnosis, surgery related to CDI diagnosis, ileus, toxic megacolon, peritonitis, or bowel perforation.

• Triple therapy = vancomycin up to 500 mg PO QID, metronidazole 7.5 mg/kg/dose IV Q6H up to 500 mg/dose, and vancomycin enema 10-20 ml/kg/dose up to 1000 ml/dose every 6 hours of vancomycin 500 mg/L solution [if tolerated, 20 mL/kg/dose every 6 hours is preferred, however in patient with additional administration considerations, a minimum of 10 mL/kg/dose every 8 hours should be used] (in patients with ileus, bowel obstruction or toxic megacolon; bowel perforation is a contraindication to enema therapy) [IC]

• Consult pediatric infectious diseases • Consult pediatric surgery to assist in management including possible surgical intervention (Table 4).

Recurrent CDI: Recurrent symptoms and positive testing for toxigenic C. difficile within 8 weeks of prior episode. First recurrence: • Classify as “mild-moderate” “severe,” or “complicated,” and treat accordingly. [IC] Second or multiple recurrences (third or more episode of CDI): • Consult pediatric infectious diseases • vancomycin PO (dose, need for concurrent IV metronidazole/vancomycin enemas depends on disease classification as

noted above) for 10-14 days then taper to 125 mg PO BID for 7 days, 125 mg PO daily for 7 days, and then pulse with 125 mg PO once every 2-3 days for 2-8 weeks. [IIC]

OR • fidaxomicin 16 mg/kg/dose BID, max 200 mg per dose, for 10 days [IIC]; pediatric ID approval is required for use.

Page 3: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

3 Michigan Medicine C. difficile Guideline December 2016

Table 1. Risk Factors for CDI

Past history of CDI

Current or recent antibiotic use (highest risk within 3 months of exposure)

Advanced age (65 or older)

Severe comorbid disease(s)

Hospitalization within 30 days

Inflammatory bowel disease (IBD

Immunosuppressed state and use of immunosuppressive drugs

Acid suppressive therapy (especially, proton pump inhibitors)

Additional risk factors specific to pediatrics

Pediatric patients with history of prematurity, prolonged or frequent

hospitalizations, or history of frequent or recent antimicrobial therapy

Pediatric patients with Hirschsprung’s disease, other intestinal dysmotility disorder,

or history of abdominal surgery, including gastrostomy or jejunostomy tubes

Note: Community-associated CDI1 can be seen in low risk population or in patients without traditional risk factors.

Figure 1. Indications and Contraindications for CDI Testing

CDI: Clostridium difficile infection; IBD: inflammatory bowel disease; ID: infectious disease (service); PCR: polymerase chain

reaction; WBC: white blood cell count.

Page 4: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

4 Michigan Medicine C. difficile Guideline December 2016

Table 2. Definition of Clostridium difficile Infection (CDI)

CDI: Clostridium difficile infection; WBC: white blood cell count.

Figure 2. University of Michigan Health System Multistep Algorithm*

for the Rapid Diagnosis of C. difficile Infection

CDI: Clostridium difficile infection; EIA: enzyme immunoassay; GDH: glutamate dehydrogenase; PCR: polymerase chain

reaction.

* Adapted under Creative Commons License from Rao K, Erb-Downward JR, Walk ST, et al. The Systemic Inflammatory

Response to Clostridium difficile Infection. PLoS ONE. 2014;9(3):e92578.

Requires either of the following:

1. A positive laboratory result for presence of toxigenic C. difficile in the stool

PLUS Any of the following symptoms compatible with CDI

Diarrhea (≥3 unformed stools in a 24-hour period without an alternate explanation such

as laxative use)

Radiographic evidence of ileus without alternate explanation (especially if leukocytosis

present [WBC >15,000 cells/mm3])

Abdominal pain with radiographic evidence of bowel thickening

Radiographic evidence of toxic megacolon

OR

2. Colonoscopic or histopathologic evidence of pseudomembranous colitis.

Page 5: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

5 Michigan Medicine C. difficile Guideline December 2016

Table 3. Criteria for Severity Classification of Clostridium difficile Infection

Adult* Pediatric*

Severity Treatment Severity Treatment

Mild/ Moderate

• Diagnosis of CDI (see

Table 2)

AND

• None of the criteria in the

“severe” or “complicated”

columns below

Patient does not meet criteria

for “severe” or “complicated”

CDI

• metronidazole 500 mg PO

TID for 10-14 days

OR

Patients with metronidazole

allergy, pregnant, nursing, or

on warfarin therapy:

• vancomycin 125 mg PO

QID for 10-14 days

Mild/ Moderate

• Diagnosis of CDI (see Table 2)

AND

• None of the criteria in the

“severe” or “complicated”

columns below

Patient does not meet criteria for

“severe” or “complicated” CDI

• metronidazole 7.5 mg/kg/dose PO

QID for 10 days, maximum 500

mg/dose

OR

• Patients with metronidazole allergy,

pregnant, nursing, on warfarin

therapy, or who fail to improve after

3-5 days of PO metronidazole

therapy: vancomycin 10 mg/kg/dose

PO QID, up to maximum 125

mg/dose x 10 days

Severe (ANY of the

following)

• Age ≥ 65

• WBC ≥ 15K

• Cr ≥ 1.5x baseline

• ANC ≤ 500

• ALB ≤ 2.5

• SOT/BMT < 100 days

• Small bowel CDI

• Inflammatory Bowel

Disease

• Treatment of rejection in

the preceding 2 months

(SOT)

• Chronic GVHD (BMT)

• vancomycin 125 mg PO QID

for 10-14 days

Severe (at least 2 abnormal lab

values OR at least 1 high-risk

condition)

• WBC ≥ 15K

• Cr ≥ 1.5x baseline

• ANC ≤ 500

• ALB ≤ 2.5

OR

• SOT/BMT < 100 days

• Small bowel CDI

• Inflammatory Bowel Disease

• Hirschsprung’s Disease or other

intestinal dysmotility disorder

• Neutropenia from malignancy

• vancomycin 10 mg/kg/dose PO

QID, up to maximum 125 mg/dose

x 10 days

Complicated (ANY of the

following)

• Septic shock –Sepsis with

persistent hypotension,

requiring vasopressors to

maintain MAP ≥ 65 mm

Hg and having a serum

lactate level > 2 mmol/L

despite adequate fluid

resuscitation

• Severe sepsis –Life-

threatening organ

dysfunction caused by a

dysregulated host response

to infection. Suspected or

documented infection and

an acute increase of ≥ 2

SOFA points

• Ileus or bowel obstruction

• Toxic megacolon

• Peritonitis

• Bowel perforation

• vancomycin 500 mg PO

QID

• metronidazole 500 mg IV

every 8 hours

• vancomycin enema 500 mg

in 1000 mL of normal saline

every 6 hours (in patients

with ileus, bowel

obstruction or toxic

megacolon)

• Consult infectious diseases

and surgery to assist in

management including

possible surgical

intervention (Table 4).

Operative management

strategies for CDI may

include exploratory

laparotomy, diverting loop

ileostomy with lavage, total

or subtotal abdominal

colectomy with end

ileostomy (Figure 5).

Complicated (ANY of the

following)

• Septic shock

• Severe sepsis

• Ileus or bowel obstruction

• Toxic megacolon

• Peritonitis

• Bowel perforation

• vancomycin up to maximum 500

mg/dose PO QID

• metronidazole 7.5 mg/kg/dose IV

every 6 hours, up to maximum 500

mg/dose • vancomycin retention enema (10-20

mL/kg/dose, up to 1000 mL max, of a 500 mg/L solution in normal saline q6h instilled by appropriately sized Foley catheter inserted into rectum with balloon inflated and Foley clamped for 1 hour, in patients with ileus, bowel obstruction or toxic megacolon). Treatment naïve patients should be started at 10 mL/kg/dose and escalated as tolerated up to 20 mL/kg/dose up to a maximum of 1000 mL). Suspected or known bowel perforation is a contraindication for rectal administration.)

• Consult pediatric infectious diseases

and pediatric surgery to assist in

management

ANC: absolute neutrophil count; ALB: albumin; BMT: bone marrow transplant; CDI: Clostridium difficile infection; Cr: serum

creatinine; GVHD: graft versus host disease; SOT: solid organ transplant; WBC: white blood cell count. *Please see text

regarding treatment of second or greater recurrence of CDI and the role of prophylactic oral vancomycin with concomitant

antibiotic use for first or greater CDI episode

Page 6: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

6 Michigan Medicine C. difficile Guideline December 2016

Figure 3. Clostridium difficile Infection Treatment Algorithm Overview

Note: Doses indicated are for adult patients. For pediatric-specific dosing recommendations see Table 3.

BID: twice per day; CDI: Clostridium difficile infection; CT: computed tomography; ID: infectious disease (service); IV:

intravenous; PO: by mouth; QID: four times per day; TID: three times per day.

Page 7: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

7 Michigan Medicine C. difficile Guideline December 2016

Figure 4. Imaging of “accordion sign” and “target sign”

(Left) Accordion sign in 50-year-old woman with C. difficile colitis. Marked submucosal edema is present in the right colon

(“thumbprint” appearance on longitudinal axis, short arrows). Oral contrast material (arrowhead) is trapped within the lumen.

(image from Macari Balthazar Megibow 1999).

(Right) A 65-yo C. difficile –positive woman with CT through the mid-abdomen showing diffusely thickened colonic wall

appearing as a “target sign” (concentric circles formed by the layers of bowel wall in inflammatory disease) on axial imaging

(arrow) (image from AJR:186, May 2006).

Table 4. Indications for Surgical Consult 2-6

Surgical consultation is appropriate for C. difficile infected patients in these situations:

- Any patient with complicated CDI (see Table 3)

- Any patient with CDI and clinical deterioration attributable to CDI, including the following:

Worsening abdominal distention/pain and/or peritonitis

Bowel obstruction

Intubation

Vasopressor requirement

Mental status changes

New or worsening Acute Kidney Injury

Worsening Lactate > 5mmol/L

Persistent or worsening leukocytosis (WBC ≥35,000 cells/mm3)

Hirschsprung’s disease

- Any patient with failure to improve with standard therapy within 5 days as determined by resolving symptoms and

physical exam, resolving WBC/band count

CDI: Clostridium difficile infection; WBC: white blood cell count.

Page 8: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

8 Michigan Medicine C. difficile Guideline December 2016

Figure 5. Operative Management Strategy for CDI

Table 5. Guidance on Preemptive Isolation for Patients with Diarrhea.

Preemptive isolation should be considered if patients have diarrhea (3 or more watery stools in 12 to 24 hours) *not caused by laxative use, chemotherapy, enteral feeds or other medical causes AND AT LEAST ONE of the following:

Current or prior antibiotic use (within 30 days)

Significant abdominal pain, not caused by incisional pain, dyspepsia, or nausea

History of C. difficile

Suspect C. difficile

Place patient in room with curtain or door (NO HALL BEDS)

Place CONTACT PRECAUTIONS-D sign on door/curtain

*1L of colostomy output, >200mL of watery rectal bag output

Page 9: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

9 Michigan Medicine C. difficile Guideline December 2016

Clinical Background

Clostridium difficile is a Gram-positive bacillus that can

asymptomatically colonize the gastrointestinal tract or

cause symptomatic disease through the production of

cytotoxins TcdA and TcdB.2 Patients usually develop

Clostridium difficile infection (CDI) after exposure to

antibiotics, and the severity of CDI can range from self-

limited diarrheal illness, to a fulminant, life-threatening

colitis.7 Even among those that recover, recurrent disease is

common.8 Despite being first identified in 1978 as the

causative agent of pseudomembranous colitis9,10 and

subsequently garnering significant attention from the

medical community, attempts to control C. difficile have not

met with much success, especially in hospitals and other

acute care settings.

The past decade has seen a significant increase in the

incidence of CDI in the United States. In hospitals and

nursing homes, there are now at least 450,000 new cases

and 29,000 deaths per year.11-13 The increased burden of

disease is largely attributable to the emergence of several

strains, especially polymerase chain reaction (PCR)

ribotype 027, which led to a worldwide epidemic.14 Though

CDI occurs in all age groups, infection with ribotype 027,

as well as CDI in general, is most common in older adults.15

Thus, as the US population ages, the incidence of CDI and

adverse outcomes will likely increase.15 Ninety-two percent

of CDI-related deaths occur in adults of age 65 or older,

where CDI is the 18th leading cause of mortality.16 The risk

of recurrent CDI is 2-fold higher with each decade of life.8

The annual rate of CDI-related hospitalizations in those

aged >85 years exceeds those of all other age groups

combined.17 All of these epidemiologic trends contribute to

an estimated $1.5 billion in excess healthcare costs each

year due to CDI.18

Rationale for Recommendations

The increased incidence of CDI and its adverse outcomes

among hospitalized patients, coupled with the availability

of new therapies has complicated the management of CDI.

This underscores the need for guidelines that review the

evidence and provide recommendations for the prevention,

diagnosis, and treatment of CDI in hospitalized patients.

Clinical Problem and Management Issues

Causes and Risk Factors

Risk factors for CDI are listed in Table 1 and elaborated

below.

History of CDI. As CDI can be recurrent, a history of

CDI is predictive of CDI in patients presenting with

diarrhea.

Antibiotic Exposure. Antibiotic use is one of the most

significant yet modifiable risk factors for CDI. Number

(dose dependent risk), class, and duration of antibiotic

use (significantly higher risk if > 7 days) impact the risk

for CDI (See Antibiotic Section under Prevention and

Treatment).

Advanced Age. Patients age 65 or older are at several

fold higher risk for CDI.19 Older patients are also at

higher risk for more severe disease with complications,

especially if they have poor preadmission functional

status.

Comorbid Conditions, Severity of Disease and

Hospitalization. Presence of severe underlying

comorbidities has been associated with increased risk of

CDI. Severity of comorbid conditions upon admission,

and longer hospitalization have all been associated with

higher risk for hospital-acquired CDI. Other risk factors

for CDI also include gastrointestinal surgery and the use

of tube feeds.20

Inflammatory Bowel Disease (IBD). Patients with

inflammatory bowel disease (IBD) are at higher risk for

CDI compared to the general population. Thus, all

patients with IBD presenting with diarrhea concerning for

possible IBD flare should also undergo C. difficile

testing, even in the absence of other risk factors for CDI.

In a retrospective study, only 61% of IBD patients with

CDI had antibiotic exposure.21 The risk in IBD patients is

even higher if they have colonic involvement and they

are on immunomodulator therapy.21

Immunosuppression. Immunosuppressed states such as

leukemia, lymphoma, HIV, neutropenia, organ

transplantation, and use of immunosuppressive drugs

significantly increase the risk for CDI. In HIV-infected

patients, CDI is the most common cause for bacterial

diarrhea and risk of CDI correlates with severity of HIV

disease.22,23 Acid Suppressive Therapy. Acid suppressive therapy,

including use of proton pump inhibitors (PPIs), elevates

the risk of CDI. (For expanded explanation, see “Proton

Pump Inhibitors” section on Page 13).

Pediatric Population Risk Factors

Risk factors for acquiring CDI in pediatric populations

include a history of prematurity, prolonged or frequent

hospitalizations, history of antimicrobial therapy, solid

organ transplantation, the presence of gastrostomy or

jejunostomy tubes, and the use of proton pump inhibitors

(see Table 1). Conditions associated with acquisition of

severe CDI are shown in Table 2. Special attention to the

importance of Hirschsprung’s disease, neutropenia from

leukemia and other malignancies, and inflammatory bowel

disease as high-risk factors for severe disease in children

should be noted.24-27

Page 10: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

10 Michigan Medicine C. difficile Guideline December 2016

Risks in Community-Associated CDI.

Community-associated CDI has a different risk profile, and

is less likely to be severe CDI. Community-associated CDI

is defined by the Infectious Diseases Society of America as

symptom onset occurring in the community or within 48

hours of hospital admission with no prior hospitalization in

the past 12 weeks. Community-associated CDI affects

populations that were previously thought to be at low risk

such as younger adults without the traditional risk factors

mentioned above. In a US population-based study involving

385 patients with CDI, 41% of cases met the criteria for

community-associated CDI. Patients with community-

associated infection were younger (median age of 50 years

compared with 72 years), had lower comorbid scores, had

less exposure to antibiotics (78% vs 94%), and were less

likely to have severe infection.28

Diagnosis

Testing for C. difficile

Patients with diarrhea and risk factors for CDI should

undergo testing for C. difficile (Figure 1).

The diagnosis of C. difficile infection (CDI) is based on the

combination of both clinical findings (usually diarrhea), as

well as laboratory or histopathological findings (see Table

2) for definition of CDI). Clinical severity can range from

mild diarrhea to severe, fulminant colitis with paralytic

ileus and toxic megacolon. Patients can also have

asymptomatic carriage or colonization of Clostridium

difficile.

The following signs and symptoms suggest patients who

exhibit these features should be tested for the disease. The

most common symptom of CDI is diarrhea, defined as 3 or

more loose or unformed stool in less than 24 hours, without

an alternative cause (e.g., laxative use). The stool may

contain occult blood, but melena and hematochezia are

atypical. Fever and abdominal pain are present in about

50% of patients, and can be markers for increasing disease

severity. Leukocytosis (WBC > 15,000 cells/mm3) occurs

frequently and may actually precede diarrhea or other

clinical symptoms.

The presence of pseudomembranes on lower endoscopy is

essentially pathognomonic for CDI, and occurs in 50% of

cases. Endoscopy is not needed for diagnostics, however, as

stool studies are readily available and active CDI renders a

patient at increased risk for endoscopic complications

including perforation.

Less common signs and symptoms of CDI include

arthralgias and reactive arthritis, as well as severe protein-

wasting diarrhea with resultant hypoalbuminemia, edema

and ascites.

Contraindications to testing for C difficile

Because diarrhea occurs in the majority of patients with

CDI, individuals without diarrhea should usually not be

tested for CDI (Figure 1). The prevalence of asymptomatic

colonization with C. difficile is 10–26% among hospitalized

patients and may approach 50% among patients residing in

long-term care facilities. Test of cure should not be

performed in patients who have finished treatment for CDI

and have experienced clinical improvement in symptoms

because they can have persistent shedding of toxin for up to

6 weeks after completing treatment.29 For the same reason,

there is no indication for testing while an individual is being

actively treated. However, testing can be performed if

symptoms have not resolved following a full treatment

course. Finally, it is important to note that some individuals

may take several weeks after completing therapy for stool

consistency and frequency to become entirely normal. In

addition, some individuals may develop a post-infectious

irritable bowel syndrome after CDI, which can be difficult

to differentiate from true recurrent CDI and clinicians must

use their judgment. In this circumstance, clinicians should

refrain from repeat testing for C. difficile.

Laboratory Testing Algorithms

Two- or three-stage testing algorithms are preferable to

single-step methods.

The optimal rapid laboratory testing algorithm for presence

of toxigenic C. difficile in stool has not been established,

but there is evidence that two- or three-stage testing

algorithms are preferable to single-step methods due to

improved specificity.30

For UMHS testing algorithms See Figures 1 and 2.

When possible, testing should be limited to diarrheal stools

unless the suspicion for CDI is high and an ileus is present.

In select patients (immunocompromised, ileus, or on

empiric therapy) the sensitivity of other rapid tests can be

low and ID consultation and PCR-based testing should be

considered (Appendix A).

The gold standard for organism detection is cytotoxigenic

culture and the corresponding gold standard for toxin

detection is the cell cytotoxicity assay.31 Both methods

require considerable time and expense and are now rarely

performed by clinical laboratories.

An overview of symptoms consistent with CDI is provided

in Figure 1. To avoid false-positive results (e.g., positive C.

difficile testing in the setting of colonization), only diarrheal

stools (those that take the shape of the container) should be

submitted for testing. An exception to this is the case of

patients with suspected CDI and ileus, in which case stool

of any consistency can be considered for testing. If a patient

with ileus and suspected CDI is unable to produce stool, a

rectal swab for tcdB PCR can be sent for after consultation

with ID. If a patient has an ileostomy with higher then

baseline output (without an alternative explanation) then a

Page 11: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

11 Michigan Medicine C. difficile Guideline December 2016

stool specimen can be sent for CDI testing. In patients

where there is concern for CDI involving the rectal stump, a

rectal swab for direct tcdB PCR may be sent after

consultation with ID. (For PCR testing at UMHS, after

electronically placing an order for a C. difficile test, phone

the lab to request PCR only from the swab. See Appendix A

for available diagnostic tests for toxigenic C. difficile).

The recommended UMHS testing algorithm consists of two

initial EIA tests (GDH and Toxins A/B), with reflex to a

PCR test for tcdB gene for discordant results (Figure 2).

Data and recent guidelines support the use of multi-step

over single-step testing with EIA for toxigenic C. difficile

due to improved test characteristics, though single-step

testing via PCR also performed well.30-34 This algorithm has

been validated by our clinical laboratory and has a negative

predictive value of 99%.35

There are circumstances when false-negative results can

occur with EIA testing alone. Immunocompromised

patients with symptoms suggestive of CDI (colitis on

imaging, ileus with minimal stool production, and/or WBC

>15,000 cells/µL with diarrhea) and patients receiving

empiric therapy at the time of diagnosis are at risk for a

false-negative EIA test.36 In these patients, if PCR testing

was not performed, an ID consult and direct PCR for tcdB

on stool or via rectal swab should be considered (at UMHS,

ordered separately by phone). Finally, single-step PCR

testing (not part of the UMHS algorithm) occurs as part of

the new Biofire test panel for gastrointestinal pathogens

(FilmArray, BioFire Diagnostics Inc., Salt Lake City, UT).

This test should not be used if CDI is suspected, and

providers should follow the recommendations in Figure 2

and order multi-step testing as indicated. However, if C.

difficile is detected as part of the Biofire panel and the

patient’s symptoms are compatible with CDI, then a

separate order for multi-step testing is unnecessary and

providers should refer to our treatment algorithm (Figure 3)

and begin therapy as indicated.37

Imaging. In patients with abdominal distention and

suspicion for CDI, radiologic evaluation may serve as a

useful diagnostic adjunct. This is especially true if there is a

concern for CDI-induced ileus or toxic megacolon. Plain

film abdominal x-rays may show dilated colon or ileus

pattern. If free air is present on x-ray imaging, emergent

surgical consult is warranted.6,38

In patients who present with abdominal pain, significant

abdominal distention, or other signs of complicated CDI,

computed tomography (CT) of the abdomen and pelvis may

be considered for further evaluation. Findings on CT may

include colonic wall thickening, ascites, megacolon

(distension of the colon of >6 cm in transverse width), ileus

or perforation.4 Overall CT sensitivity for diagnosis of C.

difficile colitis is 52–85%, with specificity of 48–93%.39

The use of enteral, intravenous and rectal contrast is

preferred by UMHS Acute Care Surgery group, unless

otherwise contraindicated. Some institutions advocate a

more rigid adherence to CT scan diagnostic criteria for C.

difficile colitis of colon wall thickening of greater than 4

mm combined with any one or more findings of pericolonic

stranding, colon wall nodularity, the “accordion” sign

(alternating edematous haustral folds separated by

transverse mucosal ridges filled with oral contrast material,

simulating the appearance of an accordion40), or otherwise

unexplained ascites, with a reported sensitivity of 70% and

specificity of 93% 39 (Figure 4).

Patients who have CT findings concerning for severe or

complicated CDI, or who are critically ill with documented

severe CDI warrant early surgical consultation. Specific

findings have not been shown to reliably predict the need

for surgical intervention.39,41 However, early involvement

of a general surgeon may initiate discussions of treatment

options, including consideration for diverting loop

ileostomy for antegrade colonic irrigation. (Refer to section

on surgical management, Figure 5).

Endoscopy. Colonoscopy may be useful in patients with

persistent diarrhea despite negative C. difficile toxin or with

toxin-positive CDI refractory to antibiotics. In patients with

positive stool testing for CDI, a colonoscopy is not

necessary for diagnosis given that pseudomembranes are

present only in 50% of patient with toxin-positive CDI.42

Additional indications to perform a colonoscopy in toxin-

positive CDI patients include the assessment of CDI

severity and the management of severe colonic distension

associated with ileus. It is worth noting that a negative

flexible sigmoidoscopy does not rule out CDI as sparing of

the rectosigmoid colon is common in CDI patients with

pseudomembranes on colonoscopy.43

Colonoscopy is contraindicated, especially for diagnostic

purposes, in patients with hemodynamic instability or with

significant risk for bowel perforation (e.g., fulminant

colitis, recent bowel surgeries, bowel obstruction).

Differential Diagnosis of Diarrhea. C. difficile-toxin

negative patients with persistent diarrhea should be

evaluated further with colonoscopy with random biopsies

and esophagogastroduodenoscopy (EGD), with duodenal

biopsies for inflammatory and non-inflammatory causes of

persistent diarrhea. Inflammatory diarrhea includes

inflammatory bowel disease (ulcerative colitis and Crohn’s

disease), celiac disease, microscopic colitis (collagenous

and lymphocytic colitis), CMV (in immunocompromised

hosts), and routine enteric pathogens when patients have

exposure history or risk factors. Non-inflammatory causes

include, dietary intolerance (lactose, fructose, or rapidly

fermentable, short-chain carbohydrates [“FODMAP”44]) or

small intestinal bacterial overgrowth in patients with

significant abdominal bloating. Functional etiologies

(irritable bowel syndrome) should be considered when

workup is negative for inflammatory and non-inflammatory

diarrhea.

Disease Classification

Criteria for disease classification are summarized in Table

3, and discussed below.

Page 12: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

12 Michigan Medicine C. difficile Guideline December 2016

Though risk factors for adverse outcomes after CDI have

been identified, there is no generally accepted and validated

definition for severe CDI. Nonetheless, it is important to

classify the episode as mild-moderate, severe, or

complicated prior to initiation of CDI therapy. It is

imperative to classify CDI by disease severity and whether

the CDI episode represents a recurrence, because it

influences the choice of therapy. Recurrent CDI often

requires a longer duration of therapy, consideration of

adjunctive treatments such as antimicrobial “chasers,” or

fecal microbiota transplantation (FMT).

CDI can range from self-limited disease to severe infection,

sometimes resulting in colectomy or death.7 Even among

those that recover, recurrent disease is common.8 Although

the reasons are incompletely understood, older adults are

disproportionately affected by adverse outcomes.16

Despite these epidemiologic links, no robust, validated

predictive models for the development of adverse outcomes

from CDI or recurrence exist for clinical use. Several

prediction models for severe disease outcomes based on

initial diagnostic criteria have been proposed with variable

sensitivity/specificity. Eight different published scores for

severe disease have been compared at diagnosis to assess if

they predicted complicated CDI in a validation cohort.45

The scores used a variety of clinical variables including

age, medication use, symptoms, vital signs, physical exam

findings, laboratory parameters, and abdominal

radiographic changes. The agreement between predicted

and observed outcomes was variable (Cohen’s κ 0.18–

0.69). The “Hines VA” score had the highest κ but was only

73.7% sensitive. The classification criteria for CDI used in

this guideline are presented in Table 3. These were drafted

after considering published guidelines on CDI,1,4,37,45,46 and

several studies on predictors of adverse outcomes and

recurrence, and the expert opinion of this guideline

committee’s membership.

This guideline’s definition for recurrence follows the CDC

definition. Recurrent disease is defined as the presence of

recurrent symptoms and positive testing within 8 weeks of

initial onset (the index episode), but after the original

symptoms resolved.1 There are situations, however, when

patients will have another episode of CDI soon after this

arbitrary 8-week window has elapsed. In such cases, it may

not be unreasonable to classify these episodes as recurrent

and treat accordingly (Figure 3). This is especially

important in patients with risk factors for further

recurrences, including the need for concurrent therapy with

antimicrobials to treat condition other than CDI, age 65 or

older, and use of proton pump inhibitors.8

Special Populations

Extra-colonic CDI. Small bowel enteritis secondary to C.

difficile is rare, but usually occurs in patients with a partial

or total colectomy. The literature on this topic consists

primarily of case reports,47 making it difficult to

recommend specific treatment strategies. However,

outcomes in small bowel enteritis from CDI can be poor

and, these patients warrant aggressive therapy in most cases

(see severe and complicated CDI arms in Figure 3).

Inflammatory Bowel Disease. There is a high incidence of

CDI in IBD patients.48-51 The most likely explanation is the

shared risk factor of an altered gut microbiota, known as

dysbiosis.52 Thus, workup of patients presenting with signs

and/or symptoms of IBD flare should include testing for

CDI. IBD patients at significant risk for CDI include those

living in nursing homes, with recent or ongoing

hospitalization, with previous broad-spectrum antibiotic

use, and with a surgical pouch.53-56 Other risk factors

include increased severity of colitis and

immunosuppression (especially corticosteroids with 3-fold

risk increase).21,57-60

Pediatric population. CDI may be associated with

significant morbidity and mortality in children.24,61 In

general, the diagnosis of CDI in pediatric patients follows

the same diagnostic criteria and algorithms for adult

patients as discussed above (see Figure 3). However, the

accurate diagnosis of CDI in infants and young children

represents a special challenge given a high rate of

asymptomatic C. difficile colonization in this population.

Treatment decisions in this population should be made in

conjunction with ID consultation. Please note the following

age-specific recommendations for interpretation of testing

results:

1. Testing for CDI in infants < 12 months of age is

generally not recommended. It is recommended to

consult Pediatric ID if CDI is suspected in infants <12

months of age. Testing should be limited to those

symptomatic with frequent diarrhea and/or ileus who

meet one or more of the following conditions:

Have comorbidities associated with severe CDI

(see Table 3)

Have history of multiple antibiotic exposures

C. difficile outbreak situations

Note: Alternative etiologies for diarrhea should always

be considered even in those infants < 12 months of age

with a positive test for CDI (e.g., viral gastroenteritis,

food allergy, carbohydrate malabsorption, immune-

mediated condition, etc.).

2. Children 12 to 36 months of age may have

asymptomatic colonization. A positive test result in a

symptomatic patient indicates possible CDI.

Alternative etiologies for diarrhea should continue to

be considered even in those testing positive for CDI.

3. Children >36 months of age who are symptomatic

with diarrhea and/or ileus, with positive testing results

suggests probable CDI, especially in those with risk

factors including history of antibiotic therapy, use of

proton pump inhibitors, or comorbidities associated

with severe CDI (see Table 1 and Table 3).

Page 13: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

13 Michigan Medicine C. difficile Guideline December 2016

Asymptomatic carriage of C. difficile varies significantly by

age. A review of multiple studies of carriage in healthy

infants and young children determined colonization of

neonates up to 1 month of age occurs at an average of 37%,

between 1 to 6 months of age at an average of 30%,

between 6 to 12 months of age at an average of 14%, and

by 36 months of age the carriage rate is similar to that of

healthy, non-hospitalized adults at < 3%.62 Carriage may be

transient, and different strains of C. difficile may colonize

an individual over time as new strains are acquired from the

environment. Breastfed infants have lower C. difficile

carriage rates than do formula-fed infants (14% vs. 30%,

respectively), but these differences decrease after 12

months of age.62 Given the frequency of asymptomatic

colonization, testing for CDI should generally only be

performed in children with diarrhea and other risk factors

for CDI.63 Despite high rates of colonization and significant

amounts of detectable toxin, clinical illness with CDI is

relatively uncommon in children <36 months of age.

Hypotheses regarding the lack of clinical illness in infants

and young children include the possibility that neonates and

infants lack cellular receptors to bind and process C.

difficile toxin, preferential colonization by nontoxigenic or

less-pathogenic strains, and protective factors in breast milk

and the developing microbiota.25,62 Rates of hospitalized

infants and young children with positive CDI testing are

increasing, but again, many studies do not adequately

distinguish CDI disease from asymptomatic

colonization.25,26,64,65 Therefore, it is critical to consider

other etiologies for diarrhea in infants and young children,

even in those with positive CDI testing. Consultation with a

Pediatric Infectious Diseases specialist may be helpful

when considering the need for treatment in young age

groups.25

Pediatric Disease Classification. In pediatric patients, a

diagnosis of CDI may be considered based on a

combination of signs and symptoms, generally involving

frequent diarrhea, and evidence of C. difficile and toxin

present in stool (see age-based interpretation of testing

results above).

1. In pediatric patients CDI is classified as mild to

moderate when lab values are reassuring (e.g., WBC

<15,000 cells/mm3 and serum creatinine <1.5 times

baseline level), similar to adult classification schemes

(see Table 3).

2. In patients failing to improve after 3 to 5 days of

therapy, lab values should be repeated to determine if it

is still appropriate to maintain classification as mild to

moderate, or if the classification should be escalated to

severe.

3. In pediatric patients CDI is determined to be severe

when ≥ 2 lab values are abnormal (e.g., WBC ≥ 15,000

cells/mm3, creatinine >1.5 times baseline level, ANC ≤

500, or ALB ≤ 2.5) OR the presence of high-risk

patient factors for severe CDI exists (e.g.,

Hirschsprung’s disease, neutropenia from leukemia,

IBD). Patients with these high-risk factors should

generally be classified as, and treated for, at least

severe disease (see Table 3).

4. In pediatric patients CDI is determined to be

complicated when evidence exists of sepsis, shock,

ileus, toxic megacolon, peritonitis, bowel perforation,

or other conditions requiring ICU admission within 2

days of CDI diagnosis are present (see Table 3).

5. For pediatric patients meeting severe or complicated

clinical criteria (see Table 4), or having risk factors

associated with severe CDI disease (see Table 3),

testing should be performed as described above (see

Figure 2), and empiric therapy should be strongly

considered while awaiting results.

The majority of CDI in infants and children is of mild to

moderate disease severity criteria.26,27,66 Applying the same

criteria for adult severe disease to pediatric populations

tends to overclassify pediatric disease as severe, hence the

need for 2 or more abnormal lab criteria required to make a

severe diagnosis (or the presence of a high-risk condition).

The frequency of pediatric CDI patients meeting criteria for

severe disease is low (~8%), with similar proportions of

severe disease noted across all pediatric age groups.66

Prevention

Antibiotics and Prevention

Nearly all antimicrobial classes have been associated with

CDI. However, clindamycin and cephalosporins (especially

third-generation cephalosporins) have been consistently

associated with the highest risk of CDI. After the

emergence of the epidemic NAP1/ribotype 027 strain in

2002, fluoroquinolones, have also been associated with a

high risk of CDI.67-69

The risk of CDI is highest during antibiotic therapy and in

the first month after cessation of antibiotics (7 to 10 fold

increased risk compared to patients who did not receive

antibiotics). Risk appears to normalize 3 months after

cessation of antibiotic therapy.70 The use of >14 Defined

Daily Doses (DDDs) of antibiotics in the 3 months prior to

CDI had the strongest association with CDI (OR 8.50;95%

CI 4.56–15.9). Another study found the risk of CDI

increases with cumulative dose and number of antibiotics,

as well as days of antibiotic exposure.71 Poor clinical

outcomes in patients with CDI were independently

associated with concomitant use of non-CDI-related

antimicrobials,72 and are associated with a doubling in risk

of failure of CDI therapy.73 In addition, use of non-CDI-

related antimicrobials within 30 days of an episode of CDI

is associated with a 3-fold increase in CDI recurrence.74

Based on the results of these studies, it is imperative that

providers stop unnecessary antibiotic therapy to reduce the

risk of CDI.

Use of Prophylactic Vancomycin

Recommendations:

1. Consider use of vancomycin prophylaxis in patients that

recently had a first or greater recurrence of CDI and require

antimicrobials for a different infection.

Page 14: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

14 Michigan Medicine C. difficile Guideline December 2016

2. The dose of prophylactic vancomycin is 125 mg PO BID

to QID and the duration of prophylaxis should be at least

50% of the expected duration of antibiotic therapy for the

other infection.

There is a paucity of evidence to support the practice of

extending or using vancomycin prophylactically in patients

that recently were diagnosed with CDI and require non-CDI

antibiotics for treatment of a different infection. A recent

retrospective study showed that, for patients where the CDI

episode was a first or greater recurrence, prophylactic

vancomycin reduced the risk of recurrent CDI by nearly

50% when on other antibiotics. Potential downsides of this

strategy include selection for resistant organisms such as

vancomycin-resistant Enterococcus, onset or worsening of

antibiotic-associated diarrhea, and additional expense.

Probiotics

Probiotics are not recommended as primary prevention of

CDI in hospitalized patients. There are conflicting data on

whether probiotics are effective in preventing primary

episodes of CDI in patients receiving antibiotics, with the

largest RCT to-date including over 2,000 patients failing to

replicate possible reductions seen from meta-analyses of

smaller, more heterogeneous trials.75,76 Also, there are

concerns regarding safety in patients with certain comorbid

conditions (immunocompromised- receiving chemotherapy,

recipients of solid organ transplant or bone marrow

transplant) which are common among hospitalized patients.

The literature has noted invasive infections or poor

outcomes with Lactobacillus probiotic use in

immunocompromised patients,77 patients with severe acute

pancreatitis,78 and patients with central venous catheters.79

As such, we do not recommend the use of probiotics in

primary prevention of CDI in hospitalized patients.

Proton Pump Inhibitors (PPIs)

PPIs and acid suppression may increase the risk of CDI and

recurrence of CDI. Unnecessary use of PPIs should be

avoided and acid suppression should be minimized,

especially in patients with a history or current diagnosis of

CDI. A systematic review of case-control and cohort

studies has shown an association between PPIs and

increased risk of CDI.8 Although a causal relationship is not

clear, this finding is similar to prior systemic reviews.80 A

prospective cohort study showed that increasing levels of

acid suppression correlated with increased risk of

nosocomial CDI, with the highest risk in patients receiving

greater than once daily PPI dosing.81

Infection Control Measures

Isolation. Patients with diarrhea and a positive C. difficile

lab test must be placed in Contact Precautions Diarrheal

(CP-D). Patients are placed in either a private room or

cohorted with another patient with CDI. CP-D rooms are

bleach cleaned on a daily basis and upon discharge.

Healthcare personnel must wear an isolation gown and

gloves upon entry to the room. Healthcare personnel must

remove gowns and gloves upon exiting the room and wash

hands with soap and water. Any equipment leaving the

room is disinfected with bleach.

Preemptive Isolation (Initiation of isolation for patients

with diarrhea prior to test results). CP-D is not required, but

may be utilized at the discretion of Infection Prevention &

Epidemiology (IPE) or the provider. (At UMHS, nursing

may initiate this by working through the provider or IPE.)

See Table 5 for further guidance on preemptive isolation.

Disinfection and Hygiene. Both the patient’s immediate

environment and the broader environment have been

implicated in the spread of C. difficile. Quaternary

ammonia-based disinfectants have been shown to be

ineffective against C. difficile spores. Bleach is sporicidal

and has been shown to decrease the bioburden of C. difficile

in the healthcare setting.82-85 An approved hospital

sporicidal (such as bleach) must be used on rooms of

patients with CDI as well as any equipment leaving those

rooms. In locations where bleach is the approved sporicidal,

a bleach wipe or 1:10 bleach solution may be used.

Additionally, patients undergoing inpatient fecal microbiota

transplantation (FMT) should have their rooms cleaned

with an approved hospital sporicidal, with the same rigor as

used in terminal cleaning, prior to them returning to the

room from the endoscopy suite. In addition to using the

appropriate disinfectant, the importance of good mechanical

cleaning with the product should be emphasized to

maximize the mechanical removal of spores.

Alcohol based hand rub is not effective against C.

difficile spores.

Hands are to be cleaned with soap and water upon

exit of a CDI room to ensure mechanical removal of

the spores.86

Duration of Isolation. Patients are to remain in CP-D for

the duration of their hospitalization, as their hospital room

will remain contaminated with C. difficile spores as long as

they inhabit it. Terminal cleaning and decontamination is

not possible with the patient present; spores persist and may

continue to be transmitted. Thus, patients must remain in

CP-D and the room must be terminally cleaned at

discharge. This recommendation for isolation holds even

for FMT recipients after their room has been cleaned with

bleach following the FMT. In special circumstances,

arrangements may be made to move the patient to allow for

a terminal clean and the discontinuation of precautions.

These are determined on a case-by-case basis with IPE and

the clinical team.

Isolation Practices for Readmission. When patients with a

history of CDI are readmitted, they do not need to be placed

in CP-D unless they are readmitted with diarrhea. Patients

with a history of CDI that are readmitted with diarrhea

should be managed in CP-D until CDI has been ruled out.

Visitor/Family Recommendations. For the protection of

family members, it is recommended that family and visitors

wear an isolation gown and gloves when assisting in the

care of a patient with CDI. Family and visitors must wash

Page 15: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

15 Michigan Medicine C. difficile Guideline December 2016

hands with soap and water upon leaving the patient room.

Though family and visitors may move about the facility,

they should not utilize the unit nourishment room. There is

no published evidence that visitors contribute to the spread

of CDI in hospitals. Studies have included interventions

such as having visitors comply with wearing cover gowns

and gloves in CDI rooms, but no studies have evaluated the

sole impact of visitors on CDI transmission.

Isolation of Asymptomatic Carriers. Except in special

circumstances, asymptomatic carriers are not placed in CP-

D. At UMHS, the exception to this is the adult bone

marrow transplant unit. Adult Bone-Marrow Transplant

(BMT) patients are screened for C. difficile upon admission

and if positive, placed in CP-D for the duration of their

hospitalization. Patients are placed in CP-D, even if they

don’t currently have symptoms as many of the treatments

for which they are admitted can cause them to become

symptomatic. Because BMT patients are such a vulnerable

population, this protects non-colonized patients from

exposure. Please note this policy does not apply to UMHS

Pediatric BMT units given the high rate of asymptomatic

colonization among young children.

Medical Treatment of C. difficile Infection

The approach to antibiotic treatment for CDI is shown in

Figure 3. Details of the treatment options and

considerations are listed below.

Treatment of asymptomatic carriers is not recommended.

There is no role for prophylactic CDI therapy in

asymptomatic carriers. In a small, randomized placebo

controlled trial, only oral vancomycin (not metronidazole)

was effective in reducing C. difficile carriage. However oral

vancomycin treatment was associated with significantly

higher rates of C. difficile carriage 2 months after

treatment.87 Further study is needed to assess the role of

asymptomatic carriers in C. difficile transmission and the

need for isolation precautions. In a prospective study, 29%

of cases of hospital acquired CDI were associated with

carriers.88

Antimicrobial Treatment Based on Disease

Severity

Antimicrobial treatment for CDI is based on the severity of

the disease:

Mild-Moderate CDI: metronidazole 500 mg PO TID

Severe CDI: vancomycin 125 mg PO QID

Complicated CDI: vancomycin 500 mg PO QID,

metronidazole 500 mg IV every 8 hours, and if ileus,

small bowel obstruction, or toxic megacolon

vancomycin enema every 6 hours.

Two recent, prospective, randomized trials comparing oral

metronidazole and oral vancomycin to tolevamer, found

vancomycin was associated with clinical success even after

adjusting for disease severity.89,90 However, this study was

not designed or powered for this secondary analysis that

pooled data from 2 RCTs.89,90 Another RCT comparing

metronidazole and vancomycin, found vancomycin

treatment was associated with a higher rate of clinical cure

in patients with severe CDI (ZAR 2007). Currently, oral

metronidazole is recommended for the treatment of

mild/moderate CDI (except with ≥2 recurrences; see Figure

3), and oral vancomycin is recommended in severe CDI.37,46

Oral vancomycin is also preferred in pregnant or

breastfeeding patients, those with metronidazole allergy, or

those receiving concomitant warfarin.

The duration of therapy in the above trials was 10 days.

However, some patients may respond slowly to treatment

and IDSA guidelines endorse treatment for 10–14 days.46

Intravenous metronidazole is less efficacious than oral

metronidazole or oral vancomycin in the treatment of CDI,

and should only be utilized when oral or enteral

administration is not feasible.

There is no supportive data for the use of oral vancomycin

doses ≥ 125 mg QID in patients with mild, moderate, or

severe disease without complications. There is an absence

of data regarding the optimal treatment of complicated CDI.

Guidelines currently recommend combination therapy with

administration of intravenous metronidazole and high-dose

oral vancomycin (500mg QID) in patients with complicated

CDI.91,92 Intracolonic administration of vancomycin may be

considered in all patients with complicated CDI, but should

be given in patients with ileus, small bowel obstruction, and

toxic megacolon.92 In the absence of data, longer durations

of therapy (≥ 14 days) may be recommended in patients

with complicated CDI,4,37,46 and final treatment plan should

be formulated in discussion with the infectious diseases

consult team.93

C. difficile Enteritis (small bowel involvement). Patient

with CDI of the small bowel should be treated with oral

vancomycin 125 mg PO QID. Small bowel involvement

with C. difficile is rare but has been reported.47 The

majority of patients with C. difficile enteritis have either

surgically altered intestinal anatomy such as colectomy

with ileostomy and/or inflammatory bowel disease. In a

review of 56 patients with C. difficile enteritis the majority

of patients required ICU management and mortality was

high (32.1%).47 In patients with an ileostomy, fever and

increased ileostomy output should prompt further

evaluation for C. difficile infection. The optimal treatment

of C. difficile enteritis is unknown. However, since these

patients are often refractory to metronidazole therapy,94

often require ICU management, and have a high mortality,47

vancomycin treatment is recommended.

Recurrent C. difficile Infection. The treatment of CDI

recurrence depends on how many recurrences the patient

has experienced.

First recurrence: Classify as mild-moderate, severe, or

complicated and treat accordingly.

Second or multiple recurrences (third or more episode

of CDI):

- vancomycin PO (dose, need for concurrent IV

metronidazole/vancomycin enemas depends on

Page 16: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

16 Michigan Medicine C. difficile Guideline December 2016

disease classification as noted in the sections

immediately above) for 10–14 days then taper to

125 mg PO BID for 7 days, 125 mg PO daily for 7

days, and then pulse with 125 mg PO every 2-3

days for 2–8 weeks. Duration of taper should be

determined in conjunction with the infectious

diseases consult services.

OR

- fidaxomicin 200 mg PO BID for 10 days (with

infectious diseases consult service approval).

There is a paucity of data regarding the optimal treatment of

patients with recurrent CDI. Guidelines currently

recommend treatment concordant with severity of disease

for the first recurrence (i.e., metronidazole for mild-

moderate CDI and vancomycin for severe CDI46). Due to

the possibility of peripheral neuropathy with prolonged

exposure to metronidazole, guidelines recommend

vancomycin therapy for second (or further) recurrences.

Tapered or pulsed dosing regimens of vancomycin have

been shown to reduce the risk of further recurrences

compared to placebo in patients with multiple recurrences,

and thus are recommended in patients with a second (or

further) recurrence.4,31,46,95

In a randomized, trial of patients with either a first episode

of CDI or first recurrence, fidaxomicin was non-inferior to

vancomycin in terms of clinical cure rates.96 Recurrence

rates were lower with fidaxomicin, but only in the subset of

patients infected with non-NAP1/027 strains. Due to

uncertain cost-effectiveness (~$300/day) compared to

vancomycin (especially when vancomycin is compounded

from the intravenous formulation),97,98 the role of

fidaxomicin in CDI therapy remains undefined. At UMHS

fidaxomicin requires ID approval and has been reserved for

treatment of CDI in patients with documented recurrent

disease who have failed a recent vancomycin taper.

Fidaxomicin has not been studied in patients with

complicated CDI, and should not be utilized in this

scenario.

Numerous agents (rifaximin or fidaxomicin “chasers,”

which are provided at the end of a vancomycin taper;

tigecycline; IVIG; nitazoxanide; etc.) have been studied in

patients with refractory or recurrent CDI.99-102 The data

supporting their use is limited. Rifaximin or fidaxomicin

chasers can be considered in patients with multiple

recurrences especially in patients who choose not to

perform fecal microbiota transplantation (FMT) or who are

not candidates for FMT.99,100

To our knowledge, there is a paucity of data evaluating

extension of CDI therapy duration for patients receiving

non-CDI antibiotics.103 However, concomitant anti-

microbials in the context of CDI are associated with poor

clinical outcomes, extended time to resolution of diarrhea,

treatment failure, and recurrence of CDI.30,72,73,104 In one

study, the use of non-CDI antimicrobials within 30 days of

an episode of CDI was associated with a 3-fold increase in

CDI recurrence.74 Therefore, it may be advisable in patients

at high risk for recurrent CDI to overlap and extend anti-

CDI therapy beyond the duration of concomitant antibiotic

use. However, there are no data to support providing

secondary prophylaxis for all patients with CDI on

concomitant antibiotic therapy. Consultation with ID is

recommended to determine if the patient would benefit

from this practice.105

Probiotic Treatment

Probiotics as adjunct treatment of CDI is not recommended

in hospitalized patients. There are limited data supporting

the use of Lactobacillus-containing probiotic preparations

in the treatment of CDI, and use is not recommended.

Two randomized, double-blind, placebo-controlled trials

have shown that Saccharomyces boulardii, in conjunction

with standard treatment for CDI, significantly reduces the

number of further episodes of CDI in patients with a history

of recurrent infection.106,107 However, a recent meta-

analysis only showed a modest but non-significant

reduction in recurrent CDI from either S. boulardii or

Lactobacillus species.108 Additionally, the use of

Saccharomyces probiotic preparations has been associated

with invasive infection in patients with central venous

catheters, intestinal disease (abdominal surgery, intestinal

obstruction, ulcerative colitis, neoplasm, bowel or gastric

ulcerations), and critically ill or immunocompromised

patients.109 In the opinion of this committee, these safety

concerns preclude use of S. boulardii for hospitalized

patients with CDI.

In patients with multiple recurrences of CDI, who are not

candidates for FMT, a regimen of staggered and tapered

oral vancomycin in combination with kefir can be

considered. A prospective case series of patients with

multiple recurrences of CDI found that daily administration

of kefir (a probiotic yogurt drink) in combination with a

staggered and tapered oral vancomycin or metronidazole

regimen achieved a treatment success rate of 84%.110

Immunotherapy

Intravenous Gamma globulins (IVIG) can be considered in

patients with hypogammaglobulinemia and recurrent CDI,

but IVIG in all other patient populations is not

recommended. There are no randomized controlled trials on

the use of human IVIG. A retrospective analysis of 18

patients with severe CDI treated with IVIG and standard

therapy found no difference in clinical outcomes compared

with matched controls.111 In patients with recurrent CDI and

hypogammaglobulinemia (<450 mg/dL) in the presence of

a cancer or immune deficiency disorder, IVIG can be

considered on a case by case basis. In a randomized,

double-blind, placebo-controlled Phase II trial, addition of

two neutralizing monocolonal antibodies against C. difficile

toxins A and B to standard therapy did not impact the initial

infection course but did significantly reduce infection

recurrence. This therapy is currently being evaluated in

Phase III trials.4,31,111,112

Page 17: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

17 Michigan Medicine C. difficile Guideline December 2016

Recently, two phase-3 studies, MODIFY I and MODIFY II

evaluated the efficacy of two monoclonal antibodies,

actoxumab (ACT) and bezlotoxumab (BEZ), with activity

against TcdA and TcdB, respectively. The study arm with

ACT alone was stopped early due to lack of efficacy in an

interim analysis. The pooled analysis of the 2327 patients

that received either ACT + BEZ or BEZ alone observed

recurrent CDI in 15.4% and 16.5%, respectively, versus

26.6% in the placebo arm (P <.001). The safety profiles in

the interventional arms were similar to the placebo arm.113

Toxin-Binding Polymers and Resins

Therapy with toxin-binding polymers (tolevamer) and

resins (cholestyramine and colestipol) is not recommended.

In two multinational, randomized, controlled trials, clinical

success (defined as resolution of diarrhea and absence of

abdominal discomfort) of tolevamer (a non-antibiotic,

toxin-binding polymer) was inferior to both metronidazole

and vancomycin.89 Regarding toxin-binding resins,

colestipol was found to be no more effective than placebo at

decreasing fecal excretion of C. difficile toxin.114

Cholestyramine, another resin, binds oral vancomycin,

which may lead to decreased concentrations of the

antibiotic,115 and thus concomitant cholestyramine and oral

vancomycin administration should be avoided in patients

with CDI.

Fecal Microbiota transplantation (FMT)

Recommendations: 1. FMT is a good treatment option for multiple

recurrences of CDI (after two or more recurrences of

CDI within one year) and can be considered in patients

with CDI not responsive to standard treatment by day

5, assuming escalation of pharmacologic therapy has

already been attempted (Figure 3).

2. At UMHS, outpatient FMT is initiated in the outpatient

ID clinic and requires a referral. Inpatient FMT is

initiated through consultation of the inpatient ID and

GI consult services, though GI consult is unnecessary if

the patient is already on the GI inpatient service.

3. Conventional therapy should be pursued for the

treatment of primary CDI or first recurrence. There is

insufficient experience with FMT to recommend it as a

primary approach. More longitudinal data on patients

that have undergone FMT are needed; therefore,

judicious use of this treatment modality is warranted.

4. Conventional therapy should be pursued first in

patients with severe CDI, and FMT is contraindicated

in patients with complicated CDI. The safety and

efficacy of FMT for these patients has not been

established.

5. Caution should be exercised with use of FMT in

patients with IBD. Those who undergo FMT for CDI

may be at increased risk of IBD flare.

6. Either frozen or fresh stool can be used for FMT in the

setting of recurrent CDI.

Overview. The human gut microbiome is a diverse

community with thousands of bacterial species116 which

likely protects against invasive pathogens.117,118 The

pathogenesis of CDI is thought to require disruption of the

gut microbiota prior to the onset of symptomatic disease,119

usually through antibiotic exposure.120

Even after recovery from CDI, some patients retain a

susceptible microbiome and can have recurrent CDI.121 This

can occur either from recrudescence of the original

infection or from reinfection with a new C. difficile

strain.122 A small minority of patients (<5%) have difficulty

achieving clinical cure with conventional antibiotic therapy

and will enter a cycle with multiple recurrences,123,124 often

relapsing soon after antibiotics are stopped. Restoration of

the gut microbiome through fecal microbiota

transplantation (FMT) appears to be the most effective

treatment strategy for these patients and has gained

widespread acceptance in the medical community.125

Indications and Summary of Evidence. There is

insufficient experience with FMT to recommend it for the

treatment of primary CDI or first recurrence vs. standard

therapy with either vancomycin or metronidazole.126,127

There is also insufficient evidence for treatment of severe

CDI or complicated CDI with FMT, though several

published case reports suggest that it may be effective.128-131

Based on a wealth of data from case reports, systematic

reviews, and clinical trials,132-144 FMT appears quite

effective for recurrent CDI with cure rates exceeding 85–

90% in most studies.

Thus, the UMHS FMT protocol excludes patients with

primary CDI, first recurrence of CDI, or complicated

disease but other patients become eligible for FMT with

two or more recurrences of CDI (especially within 1 year).

Additionally, patients are eligible for FMT with CDI not

responsive to standard treatment by day 5, assuming

escalation of pharmacologic therapy has already been

attempted (Figure 3).

Often, donors are family members or close friends. Some

studies suggest that related donors are associated with a

higher resolution of CDI than unrelated donors, 93% vs.

84%, respectively. However, the results of a meta-analysis

indicated that there was no significant difference between

outcomes from related and unrelated donors.145

Furthermore, a randomized non-inferiority trial conducted

in patients with recurrent CDI found that the use of frozen

stool for FMT resulted in a rate of clinical resolution of

diarrhea that was no worse than that obtained with fresh

stool for FMT (per-protocol analysis revealing ,83.5% vs.

85.1%; difference, −1.6% [95% CI, −10.5% to ∞];

P=0.01).146

Page 18: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

18 Michigan Medicine C. difficile Guideline December 2016

Protocol. The optimal parameters for FMT, including stool

preparation, infusion amount, and infusion route, are not

known, but FMT appears highly effective regardless of

variability in these specifics.132 There are three formulations

of stool available at UMHS through the non-profit stool

bank OpenBiome (Somerville, MA): oral fecal capsules

(outpatient only), upper GI (for G- or J-tubes only;

nasogastric or Dobhoff tube routes are not available), and

lower GI (sigmoidoscopy- or colonoscopy-delivered, in the

medical procedures unit). The upper and lower GI infusions

can also utilize stool from a directed donor (related or

unrelated), selected by the patient and/or their family.

OpenBiome has a rigorous donor selection process that

entails thorough screening questionnaires and testing of

donors and donor stool in order to ensure safety. The oral

capsule formulation has specific exclusion criteria and

lower efficacy compared to lower delivery of FMT.142,147

The efficacy of capsules is 70% with one administration

and 94% with multiple administrations.148

Outpatient FMT requires the patient be seen in the

infectious diseases (ID) clinic, so the protocol is initiated

through a referral to the ID clinic and the ID provider will

coordinate the process from that point onward. Inpatient

FMT requires consults to be placed to both the ID and

gastroenterology (GI) services, if GI is not already the

primary service for the patient. The ID/GI inpatient services

will coordinate the process from that point onward.

Preparation of the recipient ideally involves cessation of all

antibiotics, including those used to treat CDI, 48 hours prior

to FMT. In recipients undergoing FMT via colonoscopy a

standard colonoscopy bowel preparation usually must also

be administered. The ID and GI physicians coordinating the

FMT will make any final determinations about exceptions

to these criteria or modifications in the FMT preparations.

Safety. FMT is generally safe and effective for most

patients with recurrent CDI in the short term (1-2 years).93

Among most immunocompromised patients, FMT also

appears safe.140 However, patients with IBD may be at

increased risk for disease flare, fever, and/or elevation in

inflammatory markers following FMT.140,149-151 Though

several case reports exist, there is insufficient data on safety

or efficacy to support the use of FMT for severe CDI.128-131

Of particular concern is a case of toxic megacolon that

resulted in death of a patient that received FMT for severe

CDI, though it is undetermined whether FMT itself or

withdrawal of antibiotics for CDI following FMT

contributed to the outcome.152

There are few data on long-term safety of FMT, but the

intestinal microbiota has been associated with colon cancer,

diabetes, obesity, and atopic disorders such as asthma.153

One study following patients for 3-68 months (mean 17

months) after FMT154 noted the development of new

conditions, including autoimmune disease, ovarian cancer,

myocardial infarction, and stroke, though a causal

mechanism cannot be inferred from these data. Thus, more

longitudinal data on patients that have undergone FMT are

needed and at present, judicious use of this treatment

modality is warranted.

Avoid Anti-Motility Agents

Anti-motility agents in the initial treatment or treatment

adjunct of CDI are not recommended as they can mask

symptoms and increase risk of toxic megacolon.46 In a

literature review of 55 patients, 31% of patients who

received anti-motility agents without initial treatment of C.

difficile developed toxic megacolon or colonic dilatation.155

No significant difference in complications was noted in

patients who received anti-motility agents along with

treatment of C. difficile.155 Further study is needed to assess

role of anti-motility agents to control diarrhea in patients

with mild-to moderate CDI who are already being treated

for CDI.

Treatment Response

Treatment response generally occurs after 3–5 days of

therapy and is characterized by a decrease in stool

frequency, improvement in stool consistency, and

improvement in laboratory, radiologic and physiologic

parameters.

For patients with mild to moderate CDI with no

improvement or worsening of symptoms after 3–5 days of

metronidazole therapy, escalation to vancomycin therapy is

indicated.

Recurrence of CDI after the initial episode occurs in is

approximately 19–25% of patients.8,132 Approximately 25-

64.7% of patients have another recurrence of CDI after first

or multiple recurrences.8,132

Surgical Treatment

In patients who have severe, complicated (fulminant) CDI

and clinical deterioration despite maximal medical therapy,

surgery may confer mortality benefit However, in advanced

fulminant CDI, surgical mortality can be up to 80% (range

19–80%).5,156

The optimal timing for surgical intervention is not well

defined, with only expert opinion supporting

recommendations. Early surgical involvement may be

warranted in patients who are critically ill from CDI.6

While not every patient requires surgical intervention,

patients with complicated CDI (Table 4) warrant early

surgical consultation.2,45 Indications for surgical

consultation and potential considerations for surgery6 are

listed in Table 4.

Delayed surgical intervention may result in increased

morbidity and mortality.2,3 Prior studies indicate higher

mortality rates if surgical intervention occurred after

evidence of end-organ failure (e.g. intubation for

respiratory failure, vasopressor therapy for hemodynamic

instability, or acute renal failure). In one study evaluating

timing of surgical intervention for fulminant pseudo-

Page 19: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

19 Michigan Medicine C. difficile Guideline December 2016

membranous colitis, patients who survived received

surgical consult at a mean of 3.2 days from admission (or

onset of symptoms for those already in the hospital), as

compared to the nonsurvivors who received surgical consult

at a mean of 5.4 days after admission or symptom onset.157

Likewise, an analysis of the National Inpatient Sample

(NIS) database suggested that surgical intervention more

than 3 days after admission for CDI may be associated with

increased mortality.5

Surgical options include total or subtotal colectomy with

end ileostomy (total abdominal colectomy, or TAC).158

Partial colectomy is contraindicated: in one

retrospective analysis, partial colectomy carried a

mortality rate of 100% when compared to TAC.159

In patients with peritonitis, prior abdominal surgeries

with adhesions, or frank perforation, exploratory

laparotomy with TAC may be warranted.

Diverting loop ileostomy with antegrade colonic lavage

may be associated with reduced mortality when

compared to a historical population of patients

undergoing total abdominal colectomy,2 and the

majority of patients undergoing the former intervention

(93%) achieved colonic preservation. This may also

allow for earlier surgical intervention on CDI patients,

before the development of end-organ failure and shock

progression. However, more research is needed before

this can be universally adopted as standard of care.

In patients who are poor surgical candidates, or who are in

refractory end-stage septic shock, with continued clinical

decline, the risk for morbidity and mortality may be

prohibitively high, and a frank discussion with the patient

and/or the patient’s family may be warranted regarding

goals of care, including the risks and benefits involved with

an aggressive attempt at surgery vs. continued nonoperative

management with maximal medical therapy.

Special Populations - Pediatric Treatment

For pediatric patients, treatment guidelines generally follow

those outlined above for adults (see Figure 3). Ideally,

discontinuation of antimicrobial agents is the first step in

treating CDI and may suffice to resolve symptoms in mild

cases. Antiperistaltic medications are not recommended for

pediatric patients as these may mask symptoms and

precipitate complications, including toxic megacolon.

1. For pediatric patients with mild to moderate disease

(see Table 3) who lack significant pediatric risk factors

associated with severe disease (see Table 3),

metronidazole (7.5 mg/kg/dose QID, orally, maximum

500 mg/dose x 10 days) is the preferred therapy for

initial treatment of first episode of CDI.

2. For pediatric patients with criteria meeting severe

disease (Table 3), including those with risk factors

associated with severe disease (see Table 3),

metronidazole allergy, pregnancy/ breastfeeding, or

without symptom resolution after 3 to 5 days of oral

metronidazole, oral vancomycin is the preferred

therapy (10 mg/kg/dose QID, up to maximum 125

mg/dose x 10 days).

3. Combination therapy with vancomycin, both oral and

by retention enema, and intravenous metronidazole, or

second-line agents, may be considered in complicated

high-risk cases with sepsis, shock, ileus, small bowel

obstruction, peritonitis, or toxic megacolon (see Triple

Therapy in Figure 3). Do not use enema if concern of

colonic perforation. Intracolonic vancomycin

administered by retention enema (10-20 mL/kg/dose,

up to 1000 ml max, of a 500 mg/L solution in normal

saline q6h instilled by appropriately-sized Foley

catheter inserted into rectum with balloon inflated and

Foley clamped for 1 hour; treatment naïve patients

should be initiated at 10 mL/kg/dose and escalated as

tolerated to a maximum of 20 mL/kg/dose or 1000

mL), and high-dose oral vancomycin (minimum

10mg/kg/dose up to 500 mg/dose QID) in combination

with IV metronidazole should be administered. For

patients receiving combination therapy including PO,

IV, and PR vancomycin or in patient at high risk for

systemic absorption, serum vancomycin concentrations

should be considered to evaluate for elevated serum

concentrations.

4. For pediatric patients with first recurrence of CDI, that

is mild to moderate, oral metronidazole is the preferred

therapy.

5. For pediatric patients with prolonged or recurrent CDI,

metronidazole should not be used for chronic therapy

due to possible neurotoxicity. Oral vancomycin is

recommended for second or multiple recurrences (10

mg/kg/dose QID, maximum 500 mg/dose, x 10 days),

and pulsed or tapered regimens may be beneficial in

difficult cases (see adult section above).

6. Criteria for optimal use of second-line agents in

recurrent CDI, including nitazoxanide, fidaxomicin,

and rifaximin, do not exist in children, and consultation

with a Pediatric Infectious Diseases specialist is

recommended if considering these options. Pediatric

dosing for fidaxomicin is 16 mg/kg/dose BID, max 200

mg per dose; Peds ID approval is required for use of

fidaxomicin.

7. Fecal microbiota transplantation (FMT) may be

considered in children <18 years old as an outpatient to

help prevent recurrence of CDI; consultation with a

Pediatric Infectious Disease specialist is recommended.

There are currently no mechanisms in place to provide

FMT to children <18 years old as an inpatient.

8. Probiotics are not routinely recommended for either

prevention or treatment of CDI in infants and children,

due to a lack of sufficient evidence in infants and

children and a theoretical risk of bacteremia due to gut

translocation.

Summary of Evidence. It should be recognized that oral

vancomycin is the only FDA-approved medication for the

treatment of CDI in children; however, metronidazole is

currently the accepted first-line medication in otherwise

healthy infants and children with mild to moderate

disease.24,27 Prospective trials evaluating the use of

metronidazole or vancomycin for longer than 10 to 14 days

in children are lacking.26 Data does not support a clear

benefit for combination therapy vs. oral vancomycin

Page 20: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

20 Michigan Medicine C. difficile Guideline December 2016

monotherapy for treatment of severe CDI.160 Experience

with FMT in pediatrics is limited, but growing evidence

suggests that FMT is effective and safe for treatment of

chronic CDI in children.161 An FMT program for inpatients

<18 years old is currently unavailable at UMHS;

consultation with Pediatric Infectious Diseases is suggested

if considering FMT as an option as an outpatient procedure

to prevent future recurrences. The American Academy of

Pediatrics (AAP) does not currently recommend probiotics

for prevention or treatment of CDI in infants and children

citing insufficient data.25 However, a recent Cochrane

Database review of 23 randomized controlled trials found

moderate quality evidence to suggest that short-term use of

probiotics in healthy, immunocompetent adults and children

was safe and effective in the prevention of CDI.162 UMHS

guidelines reflect the recommendation of the AAP and do

not recommend probiotic use for prevention or treatment of

CDI in this patient population.

Special Populations - Inflammatory Bowel Disease

In IBD patients with concomitant CDI and mild IBD flare,

antibiotics for CDI should be started without corticosteroids

(patient should be maintained on home immunosuppression

medications). This recommendation is based on evidence

suggesting worse outcomes in IBD patients with CDI on

immunosuppression,141,142 and a reduction of corticosteroids

dose is associated with a lower colectomy rate.14 However,

with a lack of symptom improvement after 48 hours of CDI

therapy or in severe IBD flare with concomitant CDI,

antibiotics and corticosteroids co-therapy is advised. This

strategy is based on clinical data indicating a more severe

IBD course (i.e., an increased colectomy rate and mortality)

in IBD patients with CDI vs IBD patients without

CDI.50,143,144 The recommended practice of IBD experts at

UMHS is to use CDI therapy and corticosteroids when IBD

patients with concomitant CDI present with a severe IBD

flare defined as >6 bloody stools/day and one or more of:

Temp>37.5, Pulse >90, Hgb<10.5, ESR>30.

Vancomycin is preferred over metronidazole for the

treatment of CDI in IBD patients based on a retrospective

study showing fewer readmissions and shorter lengths of

stay when IBD patients are treated with vancomycin-

containing regimen relative to those treated with

metronidazole alone.163

Related National/International Guidelines

The UMHS Clinical Guideline on CDI is generally

consistent with other guidelines published nationally and

internationally, including:

1. 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.

2. Guidelines for Diagnosis, Treatment, and Prevention of

Clostridium difficile Infections. Am J Gastroenterol

2013; 108:478–498.

3. European Society of Clinical Microbiology and

Infectious Diseases: update of the treatment guidance

document for Clostridium difficile infection. Clin

Microbiol Infect 2014; 20 (Suppl. 2): 1–26

Notable differences in recommendations: Our guideline

deviates from the above guidelines in the way that it

classifies "mild-to-moderate" vs. "severe" vs. "severe and

complicated" disease (see Table 3). The change in

classification criteria was undertaken to better reflect

additional factors we identified in the literature that can

predispose patients to developing complicated C. difficile or

more severe manifestations of C. difficile.

Guideline Development Methodology

Funding

The development of this guideline was funded by the

University of Michigan Health System.

Guideline Development Team and Disclosures

The multidisciplinary guideline development team

consisted of:

Primary care physicians: Andrea H Kim, MD, Internal

Medicine

Specialists: Meghan A Arnold, MD, Pediatric Surgery,

Gregory Eschenauer, PharmD, Pharmacy, Tejal N

Gandhi, MD, Infectious Diseases, John Y Kao, MD,

Gastroenterology, Lena M Napolitano, MD Surgery,

Krishna Rao, MD, Infectious Diseases, David M.

Somand, MD, Emergency Medicine, Daniel H

Teitelbaum, MD, Pediatric Surgery, Kathleen B To,

MD, Surgery, Alison C Tribble, MD, Pediatric

Infectious Diseases, Amanda M Valyko, MPH, CIC,

Infection Control, Laraine L Washer, MD Infection

Diseases, Michael E Watson, Jr., MD, PhD, Pediatric

Infectious Diseases

A guideline development methodologist: F. Jacob

Seagull, PhD, Learning Health Sciences.

Literature search services were provided by

informationists at the Taubman Health Sciences Library,

University of Michigan Medical School.

The University of Michigan Health System endorses the

Guidelines of the Association of American Medical

Colleges and the Standards of the Accreditation Council for

Continuing Medical Education that the individuals who

present educational activities disclose significant

relationships with commercial companies whose products

or services are discussed. Disclosure of a relationship is not

intended to suggest bias in the information presented, but is

made to provide readers with information that might be of

potential importance to their evaluation of the information.

No relevant personal financial relationships with

commercial entities: Meghan A Arnold, Gregory

Eschenauer, PharmD, Tejal N Gandhi MD, John Y Kao,

Page 21: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

21 Michigan Medicine C. difficile Guideline December 2016

MD, Andrea H Kim, MD, Lena M Napolitano, MD,

Krishna Rao, MD, F Jacob Seagull, PhD, David M Somand,

MD, Daniel H Teitelbaum, MD, Kathleen B To, MD,

Alison C Tribble, MD, Amanda M Valyko, Laraine Lynn

Washer, MD, Michael E Watson Jr, MD, PhD.

Strategy for Literature Search

Within the Medline (Ovid) database, the following search

strategy was used for most of the search topics, except for

the searches on suppurative thrombosis, endocarditis, and

vascular infection. The search below is identified as Main

in the search strategies document. Because the appropriate

indexing terms either do not exist or were applied

inconsistently, the main search uses keywords in addition to

MeSH terms to arrive at the following main strategy.

1. exp *Clostridium difficile/ or exp *enterocolitis/ or exp

*Clostridium Infections/

2. exp *Hirschsprung Disease/

3. limit 2 to "all child (0 to 18 years)"

4. 1 or 3

5. exp animals/ not (exp animals/ and humans/)

6. 4 not 5

7. limit 6 to (english language and yr="2013 -Current")

8. remove duplicates from 7. The searches on suppurative

thrombosis, endocarditis, and vascular infection used the

first 3 searches of the main search strategy. The MEDLINE

In-Process search was based entirely on keywords.

Results were limited to: English, and 2013 to present. The

Main search retrieved 893 references. When the search

hedges for Guidelines, Clinical Trials, and Cohort Studies

were added, the base results are as follow:

C-Diff -Guidelines, total results were 8

C-Diff -Clinical Trials, total results were 27

C-Diff -Cohort Studies, total results were 59

The search was conducted in components each keyed to a

specific causal link in a formal problem structure (available

upon request). The search was supplemented with very

recent clinical trials known to expert members of the panel.

Negative trials were specifically sought. The search was a

single cycle.

Level of evidence supporting a diagnostic method or an

intervention:

A = systematic reviews of randomized controlled trials with

or without meta-analysis,

B = randomized controlled trials,

C = systematic review of non-randomized controlled trials

or observational studies, non-randomized controlled

trials, group observation studies (cohort, cross-

sectional, case-control),

D = individual observation studies (case study/case series),

E = expert opinion regarding benefits and harm

Search details and evidence tables available at

http://www.uofmhealth.org/provider/clinical-care-

guidelines.

Recommendations

Guideline recommendations were based on prospective

randomized controlled trials if available, to the exclusion of

other data; if RCTs were not available, observational

studies were admitted to consideration. If no such data were

available for a given link in the problem formulation, expert

opinion was used to estimate effect size. The “strength of

recommendation” for key aspects of care was determined

by expert opinion.

The strength of recommendations regarding care were

categorized as:

I = Generally should be performed

II = May be reasonable to perform

III = Generally should not be performed

Review and Endorsement

Drafts of this guideline were reviewed in clinical

conferences and by distribution for comment within

departments and divisions of the University of Michigan

Medical School to which the content is most relevant:

Family Medicine, General Medicine, Department of

Surgery, Infectious Diseases Division, Gastroenterology

Division. Pediatrics and Communicable Diseases and the

Division of Pediatric Infectious Diseases. The Executive

Committee for Clinical Affairs of the University of

Michigan Hospitals and Health Centers endorsed the final

version.

References

1. McDonald LC, Coignard B, Dubberke E, et al.

Recommendations for surveillance of clostridium difficile-

associated disease. Infect Control Hosp Epidemiol.

2007;28(2):140-145.

2. Neal MD, Alverdy JC, Hall DE, Simmons RL,

Zuckerbraun BS. Diverting loop ileostomy and colonic

lavage: An alternative to total abdominal colectomy for the

treatment of severe, complicated clostridium difficile

associated disease. Ann Surg. 2011;254(3):423-7;

discussion 427-9.

3. Halabi WJ, Nguyen VQ, Carmichael JC, Pigazzi A,

Stamos MJ, Mills S. Clostridium difficile colitis in the

united states: A decade of trends, outcomes, risk factors for

colectomy, and mortality after colectomy. J Am Coll Surg.

2013;217(5):802-812.

4. Surawicz CM, Brandt LJ, Binion DG, et al. Guidelines

for diagnosis, treatment, and prevention of clostridium

difficile infections. Am J Gastroenterol. 2013;108(4):478-

98; quiz 499.

Page 22: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

22 Michigan Medicine C. difficile Guideline December 2016

5. Ferrada P, Velopulos CG, Sultan S, et al. Timing and

type of surgical treatment of clostridium difficile-associated

disease: A practice management guideline from the eastern

association for the surgery of trauma. The Journal of

Trauma and Acute Care Surgery. 2014;76(6):1484-1493.

6. To KB, Napolitano LM. Clostridium difficile infection:

Update on diagnosis, epidemiology, and treatment

strategies. Surg Infect (Larchmt). 2014;15(5):490-502.

7. Kuijper EJ, Coignard B, Tull P, ESCMID Study Group

for Clostridium difficile, EU Member States, European

Centre for Disease Prevention and Control. Emergence of

clostridium difficile-associated disease in north america and

europe. Clin Microbiol Infect. 2006;12 Suppl 6:2-18.

8. Abou Chakra CN, Pepin J, Sirard S, Valiquette L. Risk

factors for recurrence, complications and mortality in

clostridium difficile infection: A systematic review. PLoS

One. 2014;9(6):e98400.

9. Bartlett JG, Moon N, Chang TW, Taylor N, Onderdonk

AB. Role of clostridium difficile in antibiotic-associated

pseudomembranous colitis. Gastroenterology.

1978;75(5):778-782.

10. Bartlett JG, Chang TW, Gurwith M, Gorbach SL,

Onderdonk AB. Antibiotic-associated pseudomembranous

colitis due to toxin-producing clostridia. N Engl J Med.

1978;298(10):531-534.

11. Campbell RJ, Giljahn L, Machesky K, et al. Clostridium

difficile infection in ohio hospitals and nursing homes

during 2006. Infect Control Hosp Epidemiol.

2009;30(6):526-533.

12. Tabak YP, Zilberberg MD, Johannes RS, Sun X,

McDonald LC. Attributable burden of hospital-onset

clostridium difficile infection: A propensity score matching

study. Infect Control Hosp Epidemiol. 2013;34(6):588-596.

13. Lessa FC, Mu Y, Bamberg WM, et al. Burden of

clostridium difficile infection in the united states. N. Engl.

J. Med. Feb 26 2015;372(9):825-834.

14. He M, Miyajima F, Roberts P, et al. Emergence and

global spread of epidemic healthcare-associated clostridium

difficile. Nat Genet. 2013;45(1):109-113.

15. Louie TJ, Miller MA, Crook DW, et al. Effect of age on

treatment outcomes in clostridium difficile infection. J Am

Geriatr Soc. 2013;61(2):222-230.

16. Lessa FC, Gould CV, McDonald LC. Current status of

clostridium difficile infection epidemiology. Clin Infect

Dis. 2012;55 Suppl 2:S65-70.

17. Keller JM, Surawicz CM. Clostridium difficile infection

in the elderly. Clin Geriatr Med. 2014;30(1):79-93.

18. Zimlichman E, Henderson D, Tamir O, et al. Health

care-associated infections: A meta-analysis of costs and

financial impact on the US health care system. JAMA Intern

Med. 2013;173(22):2039-2046.

19. Pepin J, Valiquette L, Alary ME, et al. Clostridium

difficile-associated diarrhea in a region of quebec from

1991 to 2003: A changing pattern of disease severity.

CMAJ. 2004;171(5):466-472.

20. Bliss DZ, Johnson S, Savik K, Clabots CR, Willard K,

Gerding DN. Acquisition of clostridium difficile and

clostridium difficile-associated diarrhea in hospitalized

patients receiving tube feeding. Ann Intern Med.

1998;129(12):1012-1019.

21. Issa M, Vijayapal A, Graham MB, et al. Impact of

clostridium difficile on inflammatory bowel disease. Clin

Gastroenterol Hepatol. 2007;5(3):345-351.

22. Sanchez TH, Brooks JT, Sullivan PS, et al. Bacterial

diarrhea in persons with HIV infection, united states, 1992-

2002. Clin Infect Dis. 2005;41(11):1621-1627.

23. Haines CF, Moore RD, Bartlett JG, et al. Clostridium

difficile in a HIV-infected cohort: Incidence, risk factors,

and clinical outcomes. AIDS. 2013;27(17):2799-2807.

24. Sammons JS, Toltzis P, Zaoutis TE. Clostridium

difficile infection in children. JAMA Pediatr.

2013;167(6):567-573.

25. Schutze GE, Willoughby RE, Committee on Infectious

Diseases, American Academy of Pediatrics. Clostridium

difficile infection in infants and children. Pediatrics.

2013;131(1):196-200.

26. Kim J, Smathers SA, Prasad P, Leckerman KH, Coffin

S, Zaoutis T. Epidemiological features of clostridium

difficile-associated disease among inpatients at children's

hospitals in the united states, 2001-2006. Pediatrics.

2008;122(6):1266-1270.

27. McFarland LV, Ozen M, Dinleyici EC, Goh S.

Comparison of pediatric and adult antibiotic-associated

diarrhea and clostridium difficile infections. World J

Gastroenterol. 2016;22(11):3078-3104.

28. Khanna S, Baddour LM, Huskins WC, et al. The

epidemiology of clostridium difficile infection in children:

A population-based study. Clinical Infectious Diseases.

2013;56(10):1401-1406.

29. Fekety R, Silva J, Kauffman C, Buggy B, Deery HG.

Treatment of antibiotic-associated clostridium difficile

Page 23: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

23 Michigan Medicine C. difficile Guideline December 2016

colitis with oral vancomycin: Comparison of two dosage

regimens. Am J Med. 1989;86(1):15-19.

30. Bagdasarian N, Rao K, Malani PN. Diagnosis and

treatment of clostridium difficile in adults: A systematic

review. JAMA. 2015;313(4):398-408.

31. Debast SB, Bauer MP, Kuijper EJ, Committee.

European society of clinical microbiology and infectious

diseases: Update of the treatment guidance document for

clostridium difficile infection. Clinical Microbiology &

Infection. 2014;20(Suppl 2):1-26.

32. Bignardi GE, Hill K, Berrington A, Settle CD. Two-

stage algorithm for clostridium difficile: Glutamate-

dehydrogenase-positive toxin-negative enzyme

immunoassay results may require further testing. J Hosp

Infect. 2013;83(4):347-349.

33. Finch LS, Duncan CM. Molecular test to determine

toxigenic capabilities in GDH-positive, toxin-negative

samples: Evaluation of the portrait toxigenic C. difficile

assay. Br J Biomed Sci. 2013;70(2):62-66.

34. Walkty A, Lagace-Wiens PR, Manickam K, et al.

Evaluation of an algorithmic approach in comparison with

the illumigene assay for laboratory diagnosis of clostridium

difficile infection. J Clin Microbiol. 2013;51(4):1152-1157.

35. Brown NA, Lebar WD, Young CL, Hankerd RE,

Newton DW. Diagnosis of clostridium difficile infection:

Comparison of four methods on specimens collected in

cary-blair transport medium and tcdB PCR on fresh versus

frozen samples. Infect Dis Rep. 2011;3(1):e5.

36. Sunkesula VC, Kundrapu S, Muganda C, Sethi AK,

Donskey CJ. Does empirical clostridium difficile infection

(CDI) therapy result in false-negative CDI diagnostic test

results?. Clinical Infectious Diseases. 2013;57(4):494-500.

37. Debast SB, Bauer MP, Kuijper EJ, European Society of

Clinical Microbiology and Infectious Diseases. European

society of clinical microbiology and infectious diseases:

Update of the treatment guidance document for clostridium

difficile infection. Clin Microbiol Infect. 2014;20 Suppl

2:1-26.

38. Fujitani S, George WL, Murthy AR. Comparison of

clinical severity score indices for clostridium difficile

infection. Infect Control Hosp Epidemiol. 2011;32(3):220-

228.

39. Kirkpatrick ID, Greenberg HM. Evaluating the CT

diagnosis of clostridium difficile colitis: Should CT guide

therapy? AJR Am J Roentgenol. 2001;176(3):635-639.

40. Macari M, Balthazar EJ, Megibow AJ. The accordion

sign at CT: A nonspecific finding in patients with colonic

edema. Radiology. 1999;211(3):743-746.

41. Ash L, Baker ME, O'Malley CM,Jr, Gordon SM,

Delaney CP, Obuchowski NA. Colonic abnormalities on

CT in adult hospitalized patients with clostridium difficile

colitis: Prevalence and significance of findings. AJR Am J

Roentgenol. 2006;186(5):1393-1400.

42. Gerding DN, Olson MM, Peterson LR, et al.

Clostridium difficile-associated diarrhea and colitis in

adults. A prospective case-controlled epidemiologic study.

Arch Intern Med. 1986;146(1):95-100.

43. Tedesco FJ. Antibiotic associated pseudomembranous

colitis with negative proctosigmoidoscopy examination.

Gastroenterology. 1979;77(2):295-297.

44. Gibson PR, Shepherd SJ. Evidence-based dietary

management of functional gastrointestinal symptoms: The

FODMAP approach. J Gastroenterol Hepatol.

2010;25(2):252-258.

45. Fujitani S, George WL, Murthy AR. Comparison of

clinical severity score indices for clostridium difficile

infection. Infect Control Hosp Epidemiol. 2011;32(3):220-

228.

46. Cohen SH, Gerding DN, Johnson S, 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.

47. Kim JH, Muder RR. Clostridium difficile enteritis: A

review and pooled analysis of the cases. Anaerobe.

2011;17(2):52-55.

48. Rodemann JF, Dubberke ER, Reske KA, Seo da H,

Stone CD. Incidence of clostridium difficile infection in

inflammatory bowel disease. Clin Gastroenterol Hepatol.

2007;5(3):339-344.

49. Issa M, Ananthakrishnan AN, Binion DG. Clostridium

difficile and inflammatory bowel disease. Inflamm Bowel

Dis. 2008;14(10):1432-1442.

50. Ananthakrishnan AN, McGinley EL, Saeian K, Binion

DG. Temporal trends in disease outcomes related to

clostridium difficile infection in patients with inflammatory

bowel disease. Inflamm Bowel Dis. 2011;17(4):976-983.

51. Kelsen JR, Kim J, Latta D, et al. Recurrence rate of

clostridium difficile infection in hospitalized pediatric

patients with inflammatory bowel disease. Inflamm Bowel

Dis. 2011;17(1):50-55.

Page 24: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

24 Michigan Medicine C. difficile Guideline December 2016

52. Kostic AD, Xavier RJ, Gevers D. The microbiome in

inflammatory bowel disease: Current status and the future

ahead. Gastroenterology. 2014;146(6):1489-1499.

53. Li Y, Qian J, Queener E, Shen B. Risk factors and

outcome of PCR-detected clostridium difficile infection in

ileal pouch patients. Inflamm Bowel Dis. 2013;19(2):397-

403.

54. Shen B, Goldblum JR, Hull TL, Remzi FH, Bennett AE,

Fazio VW. Clostridium difficile-associated pouchitis. Dig

Dis Sci. 2006;51(12):2361-2364.

55. Causey MW, Spencer MP, Steele SR. Clostridium

difficile enteritis after colectomy. Am Surg.

2009;75(12):1203-1206.

56. Lundeen SJ, Otterson MF, Binion DG, Carman ET,

Peppard WJ. Clostridium difficile enteritis: An early

postoperative complication in inflammatory bowel disease

patients after colectomy. J Gastrointest Surg.

2007;11(2):138-142.

57. Ananthakrishnan AN, Issa M, Binion DG. Clostridium

difficile and inflammatory bowel disease. Gastroenterol

Clin North Am. 2009;38(4):711-728.

58. Jen MH, Saxena S, Bottle A, Aylin P, Pollok RC.

Increased health burden associated with clostridium

difficile diarrhoea in patients with inflammatory bowel

disease. Aliment Pharmacol Ther. 2011;33(12):1322-1331.

59. Jodorkovsky D, Young Y, Abreu MT. Clinical

outcomes of patients with ulcerative colitis and co-existing

clostridium difficile infection. Dig Dis Sci. 2010;55(2):415-

420.

60. Schneeweiss S, Korzenik J, Solomon DH, Canning C,

Lee J, Bressler B. Infliximab and other immunomodulating

drugs in patients with inflammatory bowel disease and the

risk of serious bacterial infections. Aliment Pharmacol

Ther. 2009;30(3):253-264.

61. Sammons JS, Localio R, Xiao R, Coffin SE, Zaoutis T.

Clostridium difficile infection is associated with increased

risk of death and prolonged hospitalization in children.

Clinical Infectious Diseases. 2013;57(1):1-8.

62. 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.

63. Leibowitz J, Soma VL, Rosen L, Ginocchio CC, Rubin

LG. Similar proportions of stool specimens from

hospitalized children with and without diarrhea test positive

for clostridium difficile. Pediatr Infect Dis J.

2015;34(3):261-266.

64. Nylund CM, Goudie A, Garza JM, Fairbrother G,

Cohen MB. Clostridium difficile infection in hospitalized

children in the united states. Arch Pediatr Adolesc Med.

2011;165(5):451-457.

65. Zilberberg MD, Tillotson GS, McDonald C.

Clostridium difficile infections among hospitalized

children, united states, 1997-2006. Emerg Infect Dis.

2010;16(4):604-609.

66. Wendt JM, Cohen JA, Mu Y, et al. Clostridium difficile

infection among children across diverse US geographic

locations. Pediatrics. 2014;133(4):651-658.

67. Owens RC,Jr, Donskey CJ, Gaynes RP, Loo VG, Muto

CA. Antimicrobial-associated risk factors for clostridium

difficile infection. Clin Infect Dis. 2008;46 Suppl 1:S19-31.

68. Slimings C, Riley TV. Antibiotics and hospital-acquired

clostridium difficile infection: Update of systematic review

and meta-analysis. J Antimicrob Chemother.

2014;69(4):881-891.

69. Goorhuis A, Debast SB, Dutilh JC, et al. Type-specific

risk factors and outcome in an outbreak with 2 different

clostridium difficile types simultaneously in 1 hospital. Clin

Infect Dis. 2011;53(9):860-869.

70. Hensgens MP, Goorhuis A, Dekkers OM, Kuijper EJ.

Time interval of increased risk for clostridium difficile

infection after exposure to antibiotics. J Antimicrob

Chemother. 2012;67(3):742-748.

71. Stevens V, Dumyati G, Fine LS, Fisher SG, van

Wijngaarden E. Cumulative antibiotic exposures over time

and the risk of clostridium difficile infection. Clin Infect

Dis. 2011;53(1):42-48.

72. Pop-Vicas A, Shaban E, Letourneau C, Pechie A.

Empirical antimicrobial prescriptions in patients with

clostridium difficile infection at hospital admission and

impact on clinical outcome. Infect Control Hosp Epidemiol.

2012;33(11):1101-1106.

73. Modena S, Gollamudi S, Friedenberg F. Continuation of

antibiotics is associated with failure of metronidazole for

clostridium difficile-associated diarrhea. J Clin

Gastroenterol. 2006;40(1):49-54.

74. Drekonja DM, Amundson WH, Decarolis DD,

Kuskowski MA, Lederle FA, Johnson JR. Antimicrobial

use and risk for recurrent clostridium difficile infection. Am

J Med. 2011;124(11):1081.e1-1081.e7.

75. Ehrhardt S, Guo N, Hinz R, et al. Saccharomyces

boulardii to prevent antibiotic-associated diarrhea: A

randomized, double-masked, placebo-controlled trial. Open

Forum Infect Dis. 2016;3(1):ofw011.

Page 25: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

25 Michigan Medicine C. difficile Guideline December 2016

76. Lau CS, Chamberlain RS. Probiotics are effective at

preventing clostridium difficile-associated diarrhea: A

systematic review and meta-analysis. Int J Gen Med.

2016;9:27-37.

77. Luong ML, Sareyyupoglu B, Nguyen MH, et al.

Lactobacillus probiotic use in cardiothoracic transplant

recipients: A link to invasive lactobacillus infection?

Transpl Infect Dis. 2010;12(6):561-564.

78. Besselink MG, van Santvoort HC, Buskens E, et al.

Probiotic prophylaxis in predicted severe acute pancreatitis:

A randomised, double-blind, placebo-controlled trial.

Lancet. 2008;371(9613):651-659.

79. Snydman DR. The safety of probiotics. Clin Infect Dis.

2008;46 Suppl 2:S104-11; discussion S144-51.

80. Deshpande A, Pant C, Pasupuleti V, et al. Association

between proton pump inhibitor therapy and clostridium

difficile infection in a meta-analysis. Clin Gastroenterol

Hepatol. 2012;10(3):225-233.

81. Howell MD, Novack V, Grgurich P, et al. Iatrogenic

gastric acid suppression and the risk of nosocomial

clostridium difficile infection. Arch Intern Med.

2010;170(9):784-790.

82. Mayfield JL, Leet T, Miller J, Mundy LM.

Environmental control to reduce transmission of

clostridium difficile. Clin Infect Dis. 2000;31(4):995-1000.

83. Hacek DM, Ogle AM, Fisher A, Robicsek A, Peterson

LR. Significant impact of terminal room cleaning with

bleach on reducing nosocomial clostridium difficile. Am J

Infect Control. 2010;38(5):350-353.

84. Wilcox MH, Fawley WN, Wigglesworth N, Parnell P,

Verity P, Freeman J. Comparison of the effect of detergent

versus hypochlorite cleaning on environmental

contamination and incidence of clostridium difficile

infection. J Hosp Infect. 2003;54(2):109-114.

85. Weber DJ, Anderson DJ, Sexton DJ, Rutala WA. Role

of the environment in the transmission of clostridium

difficile in health care facilities. Am J Infect Control.

2013;41(5 Suppl):S105-10.

86. Pepin J, Saheb N, Coulombe MA, 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.

87. Johnson S, Homann SR, Bettin KM, et al. Treatment of

asymptomatic clostridium difficile carriers (fecal excretors)

with vancomycin or metronidazole. A randomized, placebo-

controlled trial. Ann Intern Med. 1992;117(4):297-302.

88. Curry SR, Muto CA, Schlackman JL, et al. Use of

multilocus variable number of tandem repeats analysis

genotyping to determine the role of asymptomatic carriers

in clostridium difficile transmission. Clin Infect Dis.

2013;57(8):1094-1102.

89. Johnson S, Louie TJ, Gerding DN, et al. Vancomycin,

metronidazole, or tolevamer for clostridium difficile

infection: Results from two multinational, randomized,

controlled trials. Clin Infect Dis. 2014;59(3):345-354.

90. Zar FA, Bakkanagari SR, Moorthi KM, Davis MB. A

comparison of vancomycin and metronidazole for the

treatment of clostridium difficile-associated diarrhea,

stratified by disease severity. Clin Infect Dis.

2007;45(3):302-307.

91. 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.

92. Olson MM, Shanholtzer CJ, Lee JT,Jr, Gerding DN.

Ten years of prospective clostridium difficile-associated

disease surveillance and treatment at the minneapolis VA

medical center, 1982-1991. Infect Control Hosp Epidemiol.

1994;15(6):371-381.

93. Apisarnthanarak A, Razavi B, Mundy LM. Adjunctive

intracolonic vancomycin for severe clostridium difficile

colitis: Case series and review of the literature. Clin Infect

Dis. 2002;35(6):690-696.

94. Seril DN, Shen B. Clostridium difficile infection in

patients with ileal pouches. Am J Gastroenterol.

2014;109(7):941-947.

95. McFarland LV, Elmer GW, Surawicz CM. Breaking the

cycle: Treatment strategies for 163 cases of recurrent

clostridium difficile disease. Am J Gastroenterol.

2002;97(7):1769-1775.

96. Crook DW, Walker AS, Kean Y, et al. Fidaxomicin

versus vancomycin for clostridium difficile infection: Meta-

analysis of pivotal randomized controlled trials. Clin Infect

Dis. 2012;55 Suppl 2:S93-103.

97. Bartsch SM, Umscheid CA, Fishman N, Lee BY. Is

fidaxomicin worth the cost? an economic analysis. Clin

Infect Dis. 2013;57(4):555-561.

98. Nathwani D, Cornely OA, Van Engen AK, Odufowora-

Sita O, Retsa P, Odeyemi IA. Cost-effectiveness analysis of

fidaxomicin versus vancomycin in clostridium difficile

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

2912.

Page 26: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

26 Michigan Medicine C. difficile Guideline December 2016

99. Johnson S, Gerding DN. Fidaxomicin "chaser" regimen

following vancomycin for patients with multiple

clostridium difficile recurrences. Clin Infect Dis.

2013;56(2):309-310.

100. Johnson S, Schriever C, Galang M, Kelly CP, Gerding

DN. Interruption of recurrent clostridium difficile-

associated diarrhea episodes by serial therapy with

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

848.

101. Herpers BL, Vlaminckx B, Burkhardt O, et al.

Intravenous tigecycline as adjunctive or alternative therapy

for severe refractory clostridium difficile infection. Clin

Infect Dis. 2009;48(12):1732-1735.

102. Musher DM, Logan N, Mehendiratta V, Melgarejo

NA, Garud S, Hamill RJ. Clostridium difficile colitis that

fails conventional metronidazole therapy: Response to

nitazoxanide. J Antimicrob Chemother. 2007;59(4):705-

710.

103. Rao K, Micic D, Natarajan M, 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.

104. Mullane KM, Miller MA, Weiss K, et al. Efficacy of

fidaxomicin versus vancomycin as therapy for clostridium

difficile infection in individuals taking concomitant

antibiotics for other concurrent infections. Clin Infect Dis.

2011;53(5):440-447.

105. Stranges PM, Hutton DW, Collins CD. Cost-

effectiveness analysis evaluating fidaxomicin versus oral

vancomycin for the treatment of clostridium difficile

infection in the united states. Value Health.

2013;16(2):297-304.

106. McFarland LV, Surawicz CM, Greenberg RN, et al. A

randomized placebo-controlled trial of saccharomyces

boulardii in combination with standard antibiotics for

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

1918.

107. Surawicz CM, McFarland LV, Greenberg RN, 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.

108. McFarland LV. Probiotics for the primary and

secondary prevention of C. difficile infections: A meta-

analysis and systematic review. Antibiotics (Basel).

2015;4(2):160-178.

109. Enache-Angoulvant A, Hennequin C. Invasive

saccharomyces infection: A comprehensive review. Clin

Infect Dis. 2005;41(11):1559-1568.

110. Bakken JS. Staggered and tapered antibiotic

withdrawal with administration of kefir for recurrent

clostridium difficile infection. Clin Infect Dis.

2014;59(6):858-861.

111. Juang P, Skledar SJ, Zgheib NK, et al. Clinical

outcomes of intravenous immune globulin in severe

clostridium difficile-associated diarrhea. Am J Infect

Control. 2007;35(2):131-137.

112. Lowy I, Molrine DC, Leav BA, et al. Treatment with

monoclonal antibodies against clostridium difficile toxins.

N Engl J Med. 2010;362(3):197-205.

113. Wilcox M, Gerding D, Poxton Iea. Bezlotoxumab

(BEZ) alone and with actoxumab (ACT) for prevention of

recurrent C. difficile infection (rCDI) in patients on

standard of care (SoC) antibiotics: Integrated results of 2

phase 3 studies (MODIFY I and MODIFY II). ID Week.

2015(October 7-11).

114. Mogg GA, George RH, Youngs D, et al. Randomized

controlled trial of colestipol in antibiotic-associated colitis.

Br J Surg. 1982;69(3):137-139.

115. Pantosti A, Luzzi I, Cardines R, Gianfrilli P.

Comparison of the in vitro activities of teicoplanin and

vancomycin against clostridium difficile and their

interactions with cholestyramine. Antimicrob Agents

Chemother. 1985;28(6):847-848.

116. Yatsunenko T, Rey FE, Manary MJ, et al. Human gut

microbiome viewed across age and geography. Nature.

2012;486(7402):222-227.

117. van der Waaij D, Berghuis-de Vries JM, Lekkerkerk

L. Colonization resistance of the digestive tract in

conventional and antibiotic-treated mice. J Hyg (Lond).

1971;69(3):405-411.

118. Vollaard EJ, Clasener HA. Colonization resistance.

Antimicrob Agents Chemother. 1994;38(3):409-414.

119. Britton RA, Young VB. Role of the intestinal

microbiota in resistance to colonization by clostridium

difficile. Gastroenterology. 2014;146(6):1547-1553.

120. Theriot CM, Koenigsknecht MJ, Carlson PE,Jr, et al.

Antibiotic-induced shifts in the mouse gut microbiome and

metabolome increase susceptibility to clostridium difficile

infection. Nat Commun. 2014;5:3114.

121. Figueroa I, Johnson S, Sambol SP, Goldstein EJ,

Citron DM, Gerding DN. Relapse versus reinfection:

Recurrent clostridium difficile infection following

treatment with fidaxomicin or vancomycin. Clin Infect Dis.

2012;55 Suppl 2:S104-9.

Page 27: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

27 Michigan Medicine C. difficile Guideline December 2016

122. Eyre DW, Babakhani F, Griffiths D, et al. Whole-

genome sequencing demonstrates that fidaxomicin is

superior to vancomycin for preventing reinfection and

relapse of infection with clostridium difficile. J Infect Dis.

2014;209(9):1446-1451.

123. Borody TJ, Warren EF, Leis SM, Surace R, Ashman

O, Siarakas S. Bacteriotherapy using fecal flora: Toying

with human motions. J Clin Gastroenterol. 2004;38(6):475-

483.

124. Bakken JS. Fecal bacteriotherapy for recurrent

clostridium difficile infection. Anaerobe. 2009;15(6):285-

289.

125. Bakken JS, Polgreen PM, Beekmann SE, Riedo FX,

Streit JA. Treatment approaches including fecal microbiota

transplantation for recurrent clostridium difficile infection

(RCDI) among infectious disease physicians. Anaerobe.

2013;24:20-24.

126. Lofgren ET, Moehring RW, Anderson DJ, Weber DJ,

Fefferman NH. A mathematical model to evaluate the

routine use of fecal microbiota transplantation to prevent

incident and recurrent clostridium difficile infection.

Infection Control & Hospital Epidemiology. 2014;35(1):18-

27.

127. Rao K, Young VB, Aronoff DM. Fecal microbiota

therapy: Ready for prime time? Infect Control Hosp

Epidemiol. 2014;35(1):28-30.

128. You DM, Franzos MA, Holman RP. Successful

treatment of fulminant clostridium difficile infection with

fecal bacteriotherapy. Ann Intern Med. 2008;148(8):632-

633.

129. Gallegos-Orozco JF, Paskvan-Gawryletz CD, Gurudu

SR, Orenstein R. Successful colonoscopic fecal transplant

for severe acute clostridium difficile pseudomembranous

colitis. Rev Gastroenterol Mex. 2012;77(1):40-42.

130. Neemann K, Eichele DD, Smith PW, Bociek R,

Akhtari M, Freifeld A. Fecal microbiota transplantation for

fulminant clostridium difficile infection in an allogeneic

stem cell transplant patient. Transpl Infect Dis.

2012;14(6):E161-5.

131. Trubiano JA, Gardiner B, Kwong JC, Ward P, Testro

AG, Charles PG. Faecal microbiota transplantation for

severe clostridium difficile infection in the intensive care

unit. Eur J Gastroenterol Hepatol. 2013;25(2):255-257.

132. Gough E, Shaikh H, Manges AR. Systematic review of

intestinal microbiota transplantation (fecal bacteriotherapy)

for recurrent clostridium difficile infection. Clin Infect Dis.

2011;53(10):994-1002.

133. Hamilton MJ, Weingarden AR, Sadowsky MJ,

Khoruts A. Standardized frozen preparation for

transplantation of fecal microbiota for recurrent clostridium

difficile infection. Am J Gastroenterol. 2012;107(5):761-

767.

134. Kassam Z, Hundal R, Marshall JK, Lee CH. Fecal

transplant via retention enema for refractory or recurrent

clostridium difficile infection. Arch Intern Med.

2012;172(2):191-193.

135. Kassam Z, Lee CH, Yuan Y, Hunt RH. Fecal

microbiota transplantation for clostridium difficile

infection: Systematic review and meta-analysis. Am J

Gastroenterol. 2013;108(4):500-508.

136. van Nood E, Vrieze A, Nieuwdorp M, et al. Duodenal

infusion of donor feces for recurrent clostridium difficile. N

Engl J Med. 2013;368(5):407-415.

137. Dutta SK, Girotra M, Garg S, et al. Efficacy of

combined jejunal and colonic fecal microbiota

transplantation for recurrent clostridium difficile infection.

Clin Gastroenterol Hepatol. 2014;12(9):1572-1576.

138. Emanuelsson F, Claesson BE, Ljungstrom L, Tvede

M, Ung KA. Faecal microbiota transplantation and

bacteriotherapy for recurrent clostridium difficile infection:

A retrospective evaluation of 31 patients. Scand J Infect

Dis. 2014;46(2):89-97.

139. Friedman-Moraco RJ, Mehta AK, Lyon GM, Kraft

CS. Fecal microbiota transplantation for refractory

clostridium difficile colitis in solid organ transplant

recipients. Am J Transplant. 2014;14(2):477-480.

140. Kelly CR, Ihunnah C, Fischer M, et al. Fecal

microbiota transplant for treatment of clostridium difficile

infection in immunocompromised patients. Am J

Gastroenterol. 2014;109(7):1065-1071.

141. Sha S, Liang J, Chen M, et al. Systematic review:

Faecal microbiota transplantation therapy for digestive and

nondigestive disorders in adults and children. Aliment

Pharmacol Ther. 2014;39(10):1003-1032.

142. Youngster I, Sauk J, Pindar C, et al. Fecal microbiota

transplant for relapsing clostridium difficile infection using

a frozen inoculum from unrelated donors: A randomized,

open-label, controlled pilot study. Clinical Infectious

Diseases. 2014;58(11):1515-1522.

143. Cammarota G, Masucci L, Ianiro G, et al. Randomised

clinical trial: Faecal microbiota transplantation by

colonoscopy vs. vancomycin for the treatment of recurrent

clostridium difficile infection. Aliment Pharmacol Ther.

2015;41(9):835-843.

Page 28: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

28 Michigan Medicine C. difficile Guideline December 2016

144. Lagier JC, Delord M, Million M, et al. Dramatic

reduction in clostridium difficile ribotype 027-associated

mortality with early fecal transplantation by the nasogastric

route: A preliminary report. Eur J Clin Microbiol Infect

Dis. 2015.

145. Zanella Terrier MC, Simonet ML, Bichard P, Frossard

JL. Recurrent clostridium difficile infections: The

importance of the intestinal microbiota. World J

Gastroenterol. 2014;20(23):7416-7423.

146. Lee CH, Steiner T, Petrof EO, et al. Frozen vs fresh

fecal microbiota transplantation and clinical resolution of

diarrhea in patients with recurrent clostridium difficile

infection: A randomized clinical trial. JAMA.

2016;315(2):142-149.

147. Kassam Z, Lee CH, Yuan Y, Hunt RH. Fecal

microbiota transplantation for clostridium difficile

infection: Systematic review and meta-analysis. Am J

Gastroenterol. 2013;108(4):500-508.

148. Fischer M, Allegretti JR, Smith M, et al. A multi-

center, cluster randomized dose-finding study of fecal

microbiota transplantation capsules for recurrent

clostridium difficile infection. United European

Gastroenterology Week 2015 (oct 24-28, 2015);.

2015;OP054-LB703.

149. Angelberger S, Reinisch W, Makristathis A, et al.

Temporal bacterial community dynamics vary among

ulcerative colitis patients after fecal microbiota

transplantation. Am J Gastroenterol. 2013;108(10):1620-

1630.

150. De Leon LM, Watson JB, Kelly CR. Transient flare of

ulcerative colitis after fecal microbiota transplantation for

recurrent clostridium difficile infection. Clin Gastroenterol

Hepatol. 2013;11(8):1036-1038.

151. Kump PK, Grochenig HP, Lackner S, et al. Alteration

of intestinal dysbiosis by fecal microbiota transplantation

does not induce remission in patients with chronic active

ulcerative colitis. Inflamm Bowel Dis. 2013;19(10):2155-

2165.

152. Solari PR, Fairchild PG, Noa LJ, Wallace MR.

Tempered enthusiasm for fecal transplant. Clin Infect Dis.

2014;59(2):319.

153. Sekirov I, Russell SL, Antunes LC, Finlay BB. Gut

microbiota in health and disease. Physiol Rev.

2010;90(3):859-904.

154. Brandt LJ, Aroniadis OC, Mellow M, et al. Long-term

follow-up of colonoscopic fecal microbiota transplant for

recurrent clostridium difficile infection. Am J

Gastroenterol. 2012;107(7):1079-1087.

155. Koo HL, Koo DC, Musher DM, DuPont HL.

Antimotility agents for the treatment of clostridium difficile

diarrhea and colitis. Clin Infect Dis. 2009;48(5):598-605.

156. Pepin J, Vo TT, Boutros M, et al. Risk factors for

mortality following emergency colectomy for fulminant

clostridium difficile infection. Dis Colon Rectum.

2009;52(3):400-405.

157. Ali SO, Welch JP, Dring RJ. Early surgical

intervention for fulminant pseudomembranous colitis. Am

Surg. 2008;74(1):20-26.

158. Bhangu A, Nepogodiev D, Gupta A, Torrance A,

Singh P, West Midlands Research Collaborative.

Systematic review and meta-analysis of outcomes following

emergency surgery for clostridium difficile colitis. Br J

Surg. 2012;99(11):1501-1513.

159. Lipsett PA, Samantaray DK, Tam ML, Bartlett JG,

Lillemoe KD. Pseudomembranous colitis: A surgical

disease? Surgery. 1994;116(3):491-496.

160. Bass SN, Bauer SR, Neuner EA, Lam SW.

Comparison of treatment outcomes with vancomycin alone

versus combination therapy in severe clostridium difficile

infection. J Hosp Infect. 2013;85(1):22-27.

161. Kelly CR, Kahn S, Kashyap P, et al. Update on fecal

microbiota transplantation 2015: Indications,

methodologies, mechanisms, and outlook.

Gastroenterology. 2015;149(1):223-237.

162. Goldenberg JZ, Ma SS, Saxton JD, et al. Probiotics for

the prevention of clostridium difficile-associated diarrhea in

adults and children. Cochrane Database of Systematic

Reviews. 2013;5:006095.

163. Horton HA, Dezfoli S, Berel D, et al. Antibiotics for

treatment of clostridium difficile infection in hospitalized

patients with inflammatory bowel disease. Antimicrob

Agents Chemother. 2014;58(9):5054-5059.

Page 29: lostridium difficile Clostridium difficile Infection Team ... · PDF fileClostridium difficile Infection in Adults and Children Patient population: Adult and pediatric patients with

29 Michigan Medicine C. difficile Guideline December 2016

Appendix A. Available diagnostic tests for toxigenic C. difficilea

Methods Test Target(s) Notes on Testing

Reference Tests

Cytotoxigenic Culture Toxigenic C. difficile Expertise required

Takes 24-48 hours

Cultures C. difficile and determines if isolate

can produce toxin in vitro

Cell Cytotoxicity Assay Toxins A or Ba Highly sensitive for toxin production

Expertise required

Takes 24-48 hours

Rapid Tests

EIAc

GDH

Must be paired with a test for toxinb

Sensitivity and specificity vary considerably by

manufacturer

EIAc Toxins A or Ba Sufficient for diagnosis alone

Sensitivity and specificity vary considerably by

manufacturer

NAAT More expensive than EIA

Sensitive and specific for presence of toxigenic

C. difficile

Possible increase in false positive rate if used

alone without specimen screening: increases

detection of colonization

RT-PCRc tcdBc or tcdC genes tcdA and tcdB are the genes for toxins A and B,

respectively

tcdB presence is necessary for disease

tcdC is a regulatory gene for toxin production

LAMP tcdA or tcdB genes Strains that are tcdA-positive / tcdB-negative do

not cause disease; thus false negative results

can occur with tcdA testing alone by LAMP

CDI, Clostridium difficile infection; EIA, enzyme immunoassay; GDH, glutamate dehydrogenase; LAMP, loop-mediated

isothermal amplification; NAAT, nucleic acid amplification testing; RT-PCR, real-time polymerase chain reaction. a C. difficile can produce toxin A and/or toxin B. Although both play a role in clinical disease, it is not known if strains producing

only toxin A are associated with symptomatic infection in humans. b Non-toxigenic C. difficile strains also exist, do not cause disease, and will be GDH positive. c Test available at University of Michigan (EIA for GDH, EIA for toxin A, and RT-PCR)