Management of Clostridium difficile Infection

Gastroenterology & Hepatology
October 2016, Volume 12, Issue 10

Layth S. Al-Jashaami, MD, and Herbert L. DuPont, MD

Dr Al-Jashaami is a clinical assistant professor at the University of Arizona College of Medicine in Phoenix, Arizona. Dr DuPont is a professor and director of the Center for Infectious Diseases at the University of Texas Houston School of Public Health and the McGovern Medical School in Houston, Texas; president of the Kelsey Research Foundation in Houston, Texas; and a clinical professor at Baylor College of Medicine in Houston, Texas.

Address correspondence to:
Dr Herbert L. DuPont
1200 Herman Pressler Street, Suite 733
Houston, TX 77030
Tel: 713-500-9366
Fax: 713-500-9364
E-mail: Herbert.l.Dupont@uth.tmc.edu

Abstract:Since the discovery of Clostridium difficile infection (CDI) in the 1970s, there has been an increase in the incidence, severity, and recurrence rate of the disease. We reviewed the recent CDI literature in PubMed published before February 28, 2016 that focused on advances in therapy. Despite a large number of studies describing methods for diagnosing the disease, there is currently no definitive test that identifies this infection with certainty, which complicates therapy. Recommended therapy for CDI includes oral metronidazole for mild cases and oral vancomycin or fidaxomicin for moderate to severe cases, each given for 10 to 14 days. For infection with spore-forming C difficile, this length of treatment may be insufficient to lead to cure; however, continuing antibiotics for longer periods of time may unfavorably alter the microbiome, preventing recovery. Treatment with metronidazole has been associated with an increasing failure rate, and the only clear recommended form of metronidazole for treatment of CDI is the intravenous formulation for patients unable to take oral medications. For vancomycin or fidaxomicin treatment of first CDI recurrences, the drug used in the initial bout can be repeated. For second or future recurrences, vancomycin can be given in pulsed or tapered doses. New modalities of treatment, such as bacteriotherapy and immunotherapy, show promise for the treatment of recurrent CDI.

Introduction

Clostridium difficile is a Gram-positive, anaerobic, spore-forming, toxin-producing bacteria first identified as a cause of antibiotic-associated colitis by 3 investigators working with animal models in the 1970s.1 The organism is now the most commonly identified infectious cause of antibiotic- and health care–associated diarrhea.  The Centers for Disease Control and Prevention estimated that almost half a million infections of this disease occurred in the United States in 2011 and that the infection was associated with death in 29,000 people that year.2 A recent cost estimate for hospitalized patients with primary Clostridium difficile infection (CDI) was $20,693, and for recurrent CDI, the estimate was $45,148.3

Despite advances in the treatment of CDI, there has been a steady increase in incidence, severity, mortality, and disease recurrence.4-6 Prior antibiotic exposure is the most important risk factor for CDI, leading to disruption of the normal colonic flora, which results in reduced intestinal colonization resistance. Additional risk factors for CDI are inflammatory bowel disease, immuno-deficiency, hypoalbuminemia, malignancy, organ transplant, and chemotherapy.7-9 The high recurrence rate of CDI questions the current recommendations for therapy for first episodes of CDI.

This article discusses treatment for initial and recurrent CDI. Two medical societies have provided overviews of this topic.10,11 The current article focuses on recent data obtained after these reports were published and includes controversial areas and recommendations for treatment.

Overview of Initial Treatment for Clostridium difficile Infection            

The diagnosis of CDI is still challenging despite the many laboratory tests for the infection and its growing importance. There are 2 factors complicating laboratory diagnosis of CDI: colonization by C difficile, which is common in hospitalized patients, causes a positive test result for fecal toxin; and CDI explains less than one-fourth of antibiotic-associated diarrhea cases in the hospital setting. Both of these factors explain why many of the patients treated for CDI do not actually have the infection.

An important part of therapy is to discontinue the antibiotics that predisposed the patient to CDI. In very mild cases, this may be sufficient to reverse the disease process, requiring no CDI-directed therapy. Specific CDI treatment recommendations are influenced by 2 factors: the severity of the disease (Table 1) and the number of previous discrete bouts of CDI experienced.

Pharmacologic Treatment for the First Episode of  Clostridium difficile Infection             

Guidelines for the treatment of CDI provided by the Infectious Diseases Society of America in 2010 recommended that oral metronidazole be used for all but the more severe cases of CDI, where oral vancomycin would be preferred.10 Based upon 2 studies showing that metronidazole was inferior to oral vancomycin for CDI,12,13 metronidazole should be considered for treatment of only the mildest cases. Vancomycin or fidaxomicin (Dificid, Merck) is a better choice for all clinically important cases of CDI because of metronidazole’s flawed pharmacokinetics for intestinal infections. Nearly all of the drug is absorbed from the small bowel, and low to absent colonic levels of the drug are seen during therapy,14 producing lower cure rates than oral vancomycin.12 In contrast, oral administration of vancomycin leads to high fecal drug concentrations and higher rates of recovery.15

Our recommended approach to treatment of the first bout of CDI experienced is provided in Table 2. We feel that the main use of metronidazole is for patients who cannot take oral anti-CDI drugs because of ileus, shock, or toxic megacolon, situations in which the intravenous route is employed. In these cases, it should be possible to also administer vancomycin as an enema.10 Once oral drugs can be used, oral vancomycin or fidaxomicin should be initiated.

The recommended oral dose of vancomycin is 125 mg 4 times daily for 10 to 14 days. The capsule form of vancomycin is expensive (>$1000 for 10 days), but the cost can be reduced to less than $200 through the use of compounded liquid vancomycin, which is given in the same dose and has equivalent expected efficacy.16 However, insurance companies may not pay for this form of the drug, information that should be sought before prescribing it. In patients with severe complicated CDI (Table 1), the recommended treatment is intravenous metronidazole with high-dose vancomycin 250 to 500 mg 4 times daily orally or, if oral administration is not possible, via a nasogastric tube or via an enema.

In 2011, fidaxomicin was approved by the US Food and Drug Administration for the treatment of CDI. Fidaxomicin is a macrocyclic antibiotic with little systemic absorption after oral administration,17 which leads to high colonic concentrations of the drug.18 CDI cure rates are comparable between oral vancomycin and fidaxomicin.19 Fidaxomicin given in a dose of 200 mg twice daily for 10 days is associated with a lower rate of recurrence compared with a 10-day course of oral vancomycin (125 mg 4 times daily) for CDI caused by non-NAP1/ribotype 027 strains.19,20 Possible explanations for reduced recurrence rates with fidaxomicin include effective inhibition of C difficile toxin production,21 inhibition of spore production,22 and improved preservation of the intestinal bacterial microbiome during and after treatment of CDI.23,24 Fidaxomicin was shown to be less likely than other treatments to lead to new-onset colonization by vancomycin-resistant enterococci and Candida species.25

Fidaxomicin is up to 3 times the cost of other anti-CDI therapy, which has prevented the drug’s widespread use. Nevertheless, fidaxomicin is an appropriate first-line treatment in CDI considering the reduced rate of disease recurrence and prevention of subsequent costs of therapy and hospitalization.26,27 The drug may actually be a less-expensive option considering all of the management costs of treating CDI with vancomycin, and fidaxomicin was shown in one study to be associated with an overall improvement in quality of life.28

A new anti-CDI antibiotic is tigecycline, which has been used in limited studies in critically ill patients infected with C difficile who have failed standard anti-CDI therapy.29,30 Randomized, controlled trials are needed to assess the safety and efficacy of this agent and to better define the drug’s role in the treatment of CDI.

Another antimicrobial agent evaluated in patients with CDI is the antiparasitic drug nitazoxanide (Alinia, Romark), which was found in a study to be as effective as metronidazole31 or oral vancomycin as treatment for CDI.32

The orally administered, poorly absorbed rifamycin, rifaximin (Xifaxan, Salix), has been evaluated for treatment of CDI in preliminary studies. This agent has been shown to successfully treat patients with mild to moderate CDI who failed metronidazole treatment.33 Rifaximin, like fidaxomicin, is less damaging to intestinal flora than other drugs34 despite achieving very high fecal levels of the drug,35 which is important in the prevention of CDI recurrence. Rifaximin, together with tigecycline with or without vancomycin, has been used successfully to treat refractory or fulminant CDI.36,37

Ramoplanin shows in vitro activity against strains of C difficile, including strains that have reduced susceptibilities to vancomycin and metronidazole.38 More study is needed on this agent.

Teicoplanin, a nonabsorbed glycopeptide antibiotic,39 requires further study to determine its usefulness in managing CDI cases. In one study, teicoplanin, at a dose of 400 mg administered orally and twice daily, compared favorably with vancomycin in terms of cure and side-effect profile.40

Indications for Surgery

Surgical management is indicated in patients with CDI who are not responding to medical treatment. Fulminant colitis with colonic perforation and rapidly progressive disease are also indications for surgical treatment.41 The traditional surgical approach to CDI, subtotal or total colectomy, is associated with poor outcomes and mortality as high as 50%.42 An alternative to colectomy in severe CDI is the creation of a diverting loop ileostomy with colonic lavage and treatment with vancomycin.43 Another colon-sparing approach includes a loop ileostomy with intraoperative colonic lavage using warmed polyethylene glycol solution via the ileostomy with instillation of vancomycin flushes postoperatively via the ileostomy.44

Recurrent Clostridium difficile Infection

Recurrent CDI is the most common complication of the infection. In recurrent cases of CDI, the same clinical findings are seen, including diarrhea and abdominal pain together with positive C difficile fecal toxin testing. Recurrences are often seen within days of stopping anti-CDI antibiotics in patients with apparent clinical response to treatment. Recurrences may be seen up to 2 months after apparent recovery from a bout of CDI.10 In such a situation, the recurrence more likely represents a new infection or reinfection. The rate of recurrence after a first bout of CDI treated with metronidazole or oral vancomycin is approximately 25%.45 After a first recurrence, the risk of additional recurrences is at least 40%.46 Patients with 3 or more recurrences often have one recurrence after another, leading to near-total disability and poor quality of life with limited options for therapy.

Risk factors for recurrence are older age, comorbidity, use of proton pump inhibitors,45 continuation of the antibiotic that led to the first CDI bout, reduced diversity of the intestinal microbiota from continued exposure to antibiotics,47 and failure to mount a serum antibody to the toxins of C difficile.48,49

Antibiotic Treatment for Recurrent Clostridium difficile Infection               

Treatment approaches for recurrent CDI are listed in Table 3. Recommendations for the first recurrence of CDI are to stop any non-CDI antibiotics, if possible, and to take anti-CDI antibiotics. If oral vancomycin or fidaxomicin was used in the first episode of CDI, the same drug can be used again because the reason for recurrence is not the development of antimicrobial resistance by the infecting strain of C difficile. Metronidazole is not recommended for recurrent disease for reasons previously discussed.

In patients with a first recurrence of CDI, fidaxomicin was similar to vancomycin in achieving an initial clinical response, but the rate of subsequent recurrence was lower (19% vs 35%, respectively).50 For the treatment of a second recurrence of CDI, a repeat 10-day course of vancomycin followed by 6 to 7 weeks of tapering or a pulse strategy51 is recommended to inhibit the vegetative cells of C difficile while allowing restoration of the intestinal microbiota.52

Some investigators have followed standard treatment in patients with recurrent CDI with a second anti-CDI antimicrobial agent as a chaser treatment. Drugs used this way include fidaxomicin53 and rifaximin.54-58 A second approach to chaser therapy is to administer a probiotic after the full course of anti-CDI antibiotics. Some probiotics have immunoprotective effects on the gastrointestinal tract by increasing intestinal secretory immunoglobulin A and inhibiting production of
proinflammatory cytokines (eg, interleukin-8).59,60 Saccharomyces boulardii has been successfully used after anti-CDI antibiotic therapy in 2 randomized, double-blind, placebo-controlled trials.61,62 However, the concern with the use of a probiotic such as S boulardii is the risk of bloodstream invasion in immunocompromised patients,63 hosts often seen in CDI. The fermented milk drink Kefir, with its multiple probiotics, has been used successfully as a chaser treatment after a course of vancomycin in a preliminary study of recurrent CDI.64

Fecal Microbiota Transplantation

Because patients with recurrent CDI have decreased diversity in their fecal microbiome, restoration of the intestinal microbiota, which can be achieved by fecal microbiota transplantation (FMT), is considered the most effective therapeutic approach to treat patients with at least 3 recurrences of CDI.65-67 FMT has been shown to lead to normalization of the diversity of intestinal microbiota.68 FMT was first described hundreds of years ago in China for the treatment of diarrhea and has been used for many years in treating animal disorders (in a process called rumen transfaunation).69 FMT was successfully used in the 1950s in a small number of patients with pseudomembranous colitis.70 In 1970, one of the authors of this article (HLD) performed a successful FMT by enema in a patient with progressive postantibiotic colitis complicated by renal failure (unpublished data). This was 8 years before the discovery of C difficile as the cause of the disorder.

Cure rates exceed 90% following FMT for multiple-recurrent CDI.71-74 FMT is a cost-effective strategy for the treatment of multiple-recurrent CDI75 if available, and it is less expensive and more effective than a prolonged course of vancomycin.76 FMT can be delivered to the intestine by colonoscopy,65 nasogastric route,77 enema,78 or capsules with frozen product.79 Frozen product from a healthy donor is as effective as using freshly prepared product,78 allowing more convenient product development. The increased cost and the occurrence of adverse events are limitations of colonoscopic delivery, compared with administration by enema. Two possible advantages of colonoscopic delivery of FMT product are that insurance companies may pay for colonoscopy, making FMT less expensive in these patients, and the colonoscopist will be able to evaluate the integrity of the gut mucosa in the face of coexistent inflammatory bowel disease. The major limitation of FMT is that the procedure is not being performed in all medical centers. Comprehensive donor screening is needed to reduce the possibility of transmitting infectious agents to the recipients.80

Clinical trials are underway to produce a product that can have widespread availability to restore intestinal microbiota in patients with refractory bouts of CDI. Two product approaches in development include culturable gut organisms81 and purified nonpathogenic Clostridium spp spores isolated from donor stools that can be administered by oral or colonic routes.

Immunologic Approaches

Another approach to the treatment of recurrent CDI is to facilitate immune responsiveness to the toxins of C difficile. A randomized, double-blind, placebo-
controlled study using monoclonal antibodies against C difficile toxins significantly reduced CDI recurrence.82 This approach is under development and is not yet available. However, a high cost of the monoclonal antibody will limit its broad use in preventing CDI recurrence. The logical indication for this preparation once licensed is the treatment of patients with multiple bouts of CDI when FMT is not available.

Intravenous immunoglobulin (IVIG) is another option that can be considered in the treatment of recurrent CDI. In case reports, IVIG was found to be associated with improvement of intestinal vascular permeability and mucosal damage in mice with experimentally induced CDI.83 There are little data in humans with recurrent CDI to show benefit of IVIG.84,85 Further randomized, placebo-controlled studies are needed to evaluate the cost and benefit of IVIG in recurrent CDI.

Toxoid vaccines made from C difficile toxins are in development to prevent CDI in high-risk populations and may reduce rates of CDI in high-risk patients.

Toxin-Binding Agents

Cholestyramine and colestipol have been shown to bind C difficile toxins A and B in vitro,86 and in mild cases of CDI, an extended course of cholestyramine may have value.87

Conclusion

Further studies are needed to establish an accurate test to diagnose CDI to prevent treating patients colonized by C difficile who have diarrhea due to another cause. Treatment of CDI continues to be challenging due to the high rate of infection recurrence.

The currently recommended duration of anti-microbial therapy for primary CDI is 10 to 14 days. For anthrax, another infection by a spore-forming organism, 2 months of therapy are indicated to prevent recurrence. Thus, longer-term treatment may be needed for many patients with CDI. Clinicians face a therapeutic dilemma in that CDI cure requires the reestablishment of gut microbiota diversity but prolonged treatment with full doses of antibiotics reduces intestinal microbiota diversity and gut colonization resistance. Administering standard initial therapy for 10 days to patients with primary CDI and then also giving them low-dose antibiotics intermittently for 3 to 4 weeks may reduce rates of recurrence while allowing the microbiota to become reestablished. Alternatively, 2 or more weeks of probiotic therapy may be used after standard antibiotics in primary infection to prevent recurrence.

Regarding patients with multiple recurrences, if tapering or intermittent doses of anti-CDI antibiotics are not helpful, FMT is the best approach. This treatment, which is designed to restore intestinal microbiota, should become available in all medical centers, and commercialization of the microbiota restoration methods under development is needed.

Because of the importance of anti–C difficile antibody development to disease prevention and recovery, immunologic approaches are being developed to prevent CDI in high-risk patients and to treat cases of CDI to prevent recurrences. Toxoid preparations to prevent CDI are in clinical trials, and monoclonal antibodies against toxins A and B of C difficile are being commercialized to prevent recurrences in patients with CDI.

Dr DuPont has received grants issued through the University of Texas Health Science Center from Seres Health, Rebiotix, Takeda Pharmaceuticals, and Texas Department of State Health Services. He has also received personal honorarium from and/or been on one-time advisory boards in the last 12 months for Bio-K Plus International, Merck Pharmaceuticals, Romark, Salix Pharmaceuticals, and Seres Health. Dr Al-Jashaami has no relevant conflicts of interest to disclose.

References

1. Bartlett JG. Historical perspectives on studies of Clostridium difficile and C. difficile infection. Clin Infect Dis. 2008;46(suppl 1):S4-S11.

2. Lessa FC, Mu Y, Bamberg WM, et al. Burden of Clostridium difficile infection in the United States. N Engl J Med. 2015;372(9):825-834.

3. Shah DN, Aitken SL, Barragan LF, et al. Economic burden of primary compared with recurrent Clostridium difficile infection in hospitalized patients: a prospective cohort study. J Hosp Infect. 2016;93(3):286-289.

4. Loo VG, Poirier L, Miller MA, et al. A predominantly clonal multi-institutional outbreak of Clostridium difficile-associated diarrhea with high morbidity and mortality. N Engl J Med. 2005;353(23):2442-2449.

5. Muto CA, Pokrywka M, Shutt K, et al. A large outbreak of Clostridium difficile-associated disease with an unexpected proportion of deaths and colectomies at a teaching hospital following increased fluoroquinolone use. Infect Control Hosp Epidemiol. 2005;26(3):273-280.

6. 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.

7. Ananthakrishnan AN, McGinley EL, Binion DG. Excess hospitalisation burden associated with Clostridium difficile in patients with inflammatory bowel disease. Gut. 2008;57(2):205-210.

8. Kyne L, Sougioultzis S, McFarland LV, Kelly CP. Underlying disease severity as a major risk factor for nosocomial Clostridium difficile diarrhea. Infect Control Hosp Epidemiol. 2002;23(11):653-659.

9. Patriarchi F, Rolla M, Maccioni F, et al. Clostridium difficile-related pancolitis in lung-transplanted patients with cystic fibrosis. Clin Transplant. 2011;25(1):
E46-E51.

10. Cohen SH, Gerding DN, Johnson S, et al; Society for Healthcare Epidemiology of America; Infectious Diseases Society of America. 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.

11. Surawicz CM, Brandt LJ, Binion DG, et al. Guidelines for diagnosis, treatment, and prevention of Clostridium difficile infections. Am J Gastroenterol. 2013;108(4):478-498.

12. Johnson S, Louie TJ, Gerding DN, et al; Polymer Alternative for CDI Treatment (PACT) investigators. Vancomycin, metronidazole, or tolevamer for Clostridium difficile infection: results from two multinational, randomized, controlled trials. Clin Infect Dis. 2014;59(3):345-354.

13. 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.

14. 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.

15. Gonzales M, Pepin J, Frost EH, et al. Faecal pharmacokinetics of orally administered vancomycin in patients with suspected Clostridium difficile infection. BMC Infect Dis. 2010;10:363.

16. Bass SN, Lam SW, Bauer SR, Neuner EA. Comparison of oral vancomycin capsule and solution for treatment of initial episode of severe Clostridium difficile infection. J Pharm Pract. 2015;28(2):183-188.

17. Gerber M, Ackermann G. OPT-80, a macrocyclic antimicrobial agent for the treatment of Clostridium difficile infections: a review. Expert Opin Investig Drugs. 2008;17(4):547-553.

18. Sears P, Crook DW, Louie TJ, Miller MA, Weiss K. Fidaxomicin attains high fecal concentrations with minimal plasma concentrations following oral administration in patients with Clostridium difficile infection. Clin Infect Dis. 2012;55(suppl 2):S116-S120.

19. Louie TJ, Miller MA, Mullane KM, et al; OPT-80-003 Clinical Study Group. Fidaxomicin versus vancomycin for Clostridium difficile infection. N Engl J Med. 2011;364(5):422-431.

20. Cornely OA, Nathwani D, Ivanescu C, Odufowora-Sita O, Retsa P, Odeyemi IA. Clinical efficacy of fidaxomicin compared with vancomycin and metronidazole in Clostridium difficile infections: a meta-analysis and indirect treatment comparison. J Antimicrob Chemother. 2014;69(11):2892-2900.

21. Babakhani F, Bouillaut L, Sears P, Sims C, Gomez A, Sonenshein AL. Fidaxomicin inhibits toxin production in Clostridium difficile. J Antimicrob Chemother. 2013;68(3):515-522.

22. Babakhani F, Bouillaut L, Gomez A, Sears P, Nguyen L, Sonenshein AL. Fidaxomicin inhibits spore production in Clostridium difficile. Clin Infect Dis. 2012;55(suppl 2):S162-S169.

23. Louie TJ, Cannon K, Byrne B, et al. Fidaxomicin preserves the intestinal microbiome during and after treatment of Clostridium difficile infection (CDI) and reduces both toxin reexpression and recurrence of CDI. Clin Infect Dis. 2012;55(suppl 2):S132-S142.

24. Tannock GW, Munro K, Taylor C, et al. A new macrocyclic antibiotic, fidaxomicin (OPT-80), causes less alteration to the bowel microbiota of Clostridium difficile-infected patients than does vancomycin. Microbiology. 2010;156(pt 11):
3354-3359.

25. Nerandzic MM, Mullane K, Miller MA, Babakhani F, Donskey CJ. Reduced acquisition and overgrowth of vancomycin-resistant enterococci and Candida species in patients treated with fidaxomicin versus vancomycin for Clostridium difficile infection. Clin Infect Dis. 2012;55(suppl 2):S121-S126.

26. 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.

27. 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.

28. Rubio-Terrés C, Cobo Reinoso J, Grau Cerrato S, et al. Economic assessment of fidaxomicin for the treatment of Clostridium difficile infection (CDI) in
special populations (patients with cancer, concomitant antibiotic treatment
or renal impairment) in Spain. Eur J Clin Microbiol Infect Dis. 2015;34(11):
2213-2223.

29. 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.

30. Larson KC, Belliveau PP, Spooner LM. Tigecycline for the treatment of severe Clostridium difficile infection. Ann Pharmacother. 2011;45(7-8):1005-1010.

31. Musher DM, Logan N, Hamill RJ, et al. Nitazoxanide for the treatment of Clostridium difficile colitis. Clin Infect Dis. 2006;43(4):421-427.

32. Musher DM, Logan N, Bressler AM, Johnson DP, Rossignol JF. Nitazoxanide versus vancomycin in Clostridium difficile infection: a randomized, double-blind study. Clin Infect Dis. 2009;48(4):e41-e46.

33. Patrick Basu P, Dinani A, Rayapudi K, et al. Rifaximin therapy for metronidazole-unresponsive Clostridium difficile infection: a prospective pilot trial. Therap Adv Gastroenterol. 2010;3(4):221-225.

34. DuPont HL, Jiang ZD, Okhuysen PC, et al. A randomized, double-blind, placebo-controlled trial of rifaximin to prevent travelers’ diarrhea. Ann Intern Med. 2005;142(10):805-812.

35. Jiang ZD, Ke S, Palazzini E, Riopel L, DuPont H. In vitro activity and fecal concentration of rifaximin after oral administration. Antimicrob Agents Chemother. 2000;44(8):2205-2206.

36. El-Herte RI, Baban TA, Kanj SS. Recurrent refractory Clostridium difficile colitis treated successfully with rifaximin and tigecycline: a case report and review of the literature. Scand J Infect Dis. 2012;44(3):228-230.

37. Lao D 2nd, Chiang T, Gomez E. Refractory Clostridium difficile infection successfully treated with tigecycline, rifaximin, and vancomycin. Case Rep Med. 2012;2012:702910.

38. Peláez T, Alcalá L, Alonso R, et al. In vitro activity of ramoplanin against Clostridium difficile, including strains with reduced susceptibility to vancomycin or with resistance to metronidazole. Antimicrob Agents Chemother. 2005;49(3):
1157-1159.

39. The Swedish CDAD Study Group. Treatment of Clostridium difficile associated diarrhea and colitis with an oral preparation of teicoplanin; a dose finding study. Scand J Infect Dis. 1994;26(3):309-316.

40. Nelson R. Antibiotic treatment for Clostridium difficile-associated diarrhea in adults. Cochrane Database Syst Rev. 2007;(3):CD004610.

41. Sailhamer EA, Carson K, Chang Y, et al. Fulminant Clostridium difficile colitis: patterns of care and predictors of mortality. Arch Surg. 2009;144(5):433-439.

42. Longo WE, Mazuski JE, Virgo KS, Lee P, Bahadursingh AN, Johnson FE. Outcome after colectomy for Clostridium difficile colitis. Dis Colon Rectum. 2004;47(10):1620-1626.

43. 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):423-427.

44. Synnott K, Mealy K, Merry C, Kyne L, Keane C, Quill R. Timing of surgery for fulminating pseudomembranous colitis. Br J Surg. 1998;85(2):229-231.

45. 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.

46. 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.

47. Chang JY, Antonopoulos DA, Kalra A, et al. Decreased diversity of the fecal microbiome in recurrent Clostridium difficile-associated diarrhea. J Infect Dis. 2008;197(3):435-438.

48. Bauer MP, Nibbering PH, Poxton IR, Kuijper EJ, van Dissel JT. Humoral immune response as predictor of recurrence in Clostridium difficile infection. Clin Microbiol Infect. 2014;20(12):1323-1328.

49. Kyne L, Warny M, Qamar A, Kelly CP. Association between antibody response to toxin A and protection against recurrent Clostridium difficile diarrhoea. Lancet. 2001;357(9251):189-193.

50. Cornely OA, Miller MA, Louie TJ, Crook DW, Gorbach SL. Treatment of first recurrence of Clostridium difficile infection: fidaxomicin versus vancomycin. Clin Infect Dis. 2012;55(suppl 2):S154-S161.

51. Gerding DN, Muto CA, Owens RC Jr. Treatment of Clostridium difficile infection. Clin Infect Dis. 2008;46(suppl 1):S32-S42.

52. 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.

53. Johnson S, Gerding DN. Fidaxomicin “chaser” regimen following vancomycin for patients with multiple Clostridium difficile recurrences. Clin Infect Dis. 2013;56(2):309-310.

54. Boero M, Berti E, Morgando A, Verme G. Treatment for colitis caused by Clostridium difficile: results of a randomized, open-label study of rifaximin vs. vancomycin. Microbiologia Medica. 1990;5:74-77.

55. Garey KW, Ghantoji SS, Shah DN, et al. A randomized, double-blind, placebo-controlled pilot study to assess the ability of rifaximin to prevent recurrent diarrhoea in patients with Clostridium difficile infection. J Antimicrob Chemother. 2011;66(12):2850-2855.

56. Garey KW, Jiang ZD, Bellard A, DuPont HL. Rifaximin in treatment of recurrent Clostridium difficile-associated diarrhea: an uncontrolled pilot study. J Clin Gastroenterol. 2009;43(1):91-93.

57. 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.

58. Mattila E, Arkkila P, Mattila PS, Tarkka E, Tissari P, Anttila VJ. Rifaximin in the treatment of recurrent Clostridium difficile infection. Aliment Pharmacol Ther. 2013;37(1):122-128.

59. Buts JP, Bernasconi P, Vaerman JP, Dive C. Stimulation of secretory IgA and secretory component of immunoglobulins in small intestine of rats treated with Saccharomyces boulardii. Dig Dis Sci. 1990;35(2):251-256.

60. Dahan S, Dalmasso G, Imbert V, Peyron JF, Rampal P, Czerucka D. Saccharomyces boulardii interferes with enterohemorrhagic Escherichia coli-induced signaling pathways in T84 cells. Infect Immun. 2003;71(2):766-773.

61. 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.

62. 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.

63. Muñoz P, Bouza E, Cuenca-Estrella M, et al. Saccharomyces cerevisiae fungemia: an emerging infectious disease. Clin Infect Dis. 2005;40(11):1625-1634.

64. Bakken JS. Staggered and tapered antibiotic withdrawal with administration of kefir for recurrent Clostridium difficile infection. Clin Infect Dis. 2014;59(6):
858-861.

65. 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.

66. Khoruts A, Dicksved J, Jansson JK, Sadowsky MJ. Changes in the composition of the human fecal microbiome after bacteriotherapy for recurrent Clostridium difficile-associated diarrhea. J Clin Gastroenterol. 2010;44(5):354-360.

67. Petrof EO, Khoruts A. From stool transplants to next-generation microbiota therapeutics. Gastroenterology. 2014;146(6):1573-1582.

68. Song Y, Garg S, Girotra M, et al. Microbiota dynamics in patients treated with fecal microbiota transplantation for recurrent Clostridium difficile infection. PLoS One. 2013;8(11):e81330.

69. DePeters EJ, George LW. Rumen transfaunation. Immunol Lett. 2014;
162(2 pt A):69-76.

70. Eiseman B, Silen W, Bascom GS, Kauvar AJ. Fecal enema as an adjunct in the treatment of pseudomembranous enterocolitis. Surgery. 1958;44(5):854-859.

71. 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.

72. Guo B, Harstall C, Louie T, Veldhuyzen van Zanten S, Dieleman LA. Systematic review: faecal transplantation for the treatment of Clostridium difficile-associated disease. Aliment Pharmacol Ther. 2012;35(8):865-875.

73. 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.

74. Sofi AA, Silverman AL, Khuder S, Garborg K, Westerink JM, Nawras A. Relationship of symptom duration and fecal bacteriotherapy in Clostridium difficile infection-pooled data analysis and a systematic review. Scand J Gastroenterol. 2013;48(3):266-273.

75. Konijeti GG, Sauk J, Shrime MG, Gupta M, Ananthakrishnan AN. Cost-effectiveness of competing strategies for management of recurrent Clostridium difficile infection: a decision analysis. Clin Infect Dis. 2014;58(11):1507-1514.

76. Lapointe-Shaw L, Tran KL, Coyte PC, et al. Cost-effectiveness analysis of six strategies to treat recurrent Clostridium difficile infection. PLoS One. 2016;11(2):e0149521.

77. 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.

78. 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.

79. Hirsch BE, Saraiya N, Poeth K, Schwartz RM, Epstein ME, Honig G. Effectiveness of fecal-derived microbiota transfer using orally administered capsules for recurrent Clostridium difficile infection. BMC Infect Dis. 2015;15:191.

80. Bakken JS, Borody T, Brandt LJ, et al; Fecal Microbiota Transplantation Workgroup. Treating Clostridium difficile infection with fecal microbiota transplantation. Clin Gastroenterol Hepatol. 2011;9(12):1044-1049.

81. Petrof EO, Gloor GB, Vanner SJ, et al. Stool substitute transplant therapy for the eradication of Clostridium difficile infection: ‘rePOOPulating’ the gut. Microbiome. 2013;1(1):3.

82. 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.

83. Saito T, Kimura S, Tateda K, et al. Evidence of intravenous immunoglobulin as a critical supportive therapy against Clostridium difficile toxin-mediated lethality in mice. J Antimicrob Chemother. 2011;66(5):1096-1099.

84. Abougergi MS, Broor A, Cui W, Jaar BG. Intravenous immunoglobulin for the treatment of severe Clostridium difficile colitis: an observational study and review of the literature. J Hosp Med. 2010;5(1):E1-E9.

85. Abougergi MS, Kwon JH. Intravenous immunoglobulin for the treatment of Clostridium difficile infection: a review. Dig Dis Sci. 2011;56(1):19-26.

86. Hedge DD, Strain JD, Heins JR, Farver DK. New advances in the treatment of Clostridium difficile infection (CDI). Ther Clin Risk Manag. 2008;4(5):949-964.

87. Moncino MD, Falletta JM. Multiple relapses of Clostridium difficile-associated diarrhea in a cancer patient. Successful control with long-term cholestyramine therapy. Am J Pediatr Hematol Oncol. 1992;14(4):361-364.

88. Henrich TJ, Krakower D, Bitton A, Yokoe DS. Clinical risk factors for severe Clostridium difficile-associated disease. Emerg Infect Dis. 2009;15(3):415-422.

89. Clutter DS, Dubrovskaya Y, Merl MY, Teperman L, Press R, Safdar A. Fidaxomicin versus conventional antimicrobial therapy in 59 recipients of solid organ and hematopoietic stem cell transplantation with Clostridium difficile-associated diarrhea. Antimicrob Agents Chemother. 2013;57(9):4501-4505.

Millennium Medical Publishing, Inc