International Journal of Food and Allied Sciences

Similar documents
Clinical Infectious Diseases Advance Access published December 7, 2012

Probiotics for Primary Prevention of Clostridium difficile Infection

Updates to pharmacological management in the prevention of recurrent Clostridium difficile

ENGLISH FOR PROFESSIONAL PURPOSES UNIT 3 HOW TO DEAL WITH CLOSTRIDIUM DIFFICILE

Safety of Saccharomyces boulardii (Florastor) in Solid Organ Transplant Patients

Fecal Microbiota Transplantation

ABSTRACT PURPOSE METHODS

Stony Brook Adult Clostridium difficile Management Guidelines. Discontinue all unnecessary antibiotics

Long-Term Care Updates

C. difficile: When to Do Fecal Microbiota Transplant (FMT)

Division of GIM Lecture Series Case Presentation David A. Erickson, M.D October 9th, 2013

The Epidemiology of Clostridium difficile DANIEL SAMAN, DRPH, MPH RESEARCH SCIENTIST ESSENTIA INSTITUTE OF RURAL HEALTH

EDUCATIONAL COMMENTARY CLOSTRIDIUM DIFFICILE UPDATE

Corporate Medical Policy Fecal Microbiota Transplantation

Clinical Policy Bulletin: Fecal Bacteriotherapy

Fecal Microbiota Transplantation in C. diff. colitis Benefits and Limitations

Update on Clostridium difficile infection.

Long-Term Care Updates

Patient Safety Summit 2014

Updated Clostridium difficile Treatment Guidelines

Terapia dell infezione da Clostridium difficile. Massimo Coen I Div Mal Inf AO L Sacco

When To Do Fecal Microbiota Transplant (FMT) For C. difficile

All POOPed out: fecal microbiota transplant in C. difficile

Clostridium difficile Infection: Diagnosis and Management

Star Articles in Review

Probiotics: Their Role in Medicine Today. Objectives. Probiotics: What Are They? 11/3/2017

CLOSTRIDIUM DIFICILE. Negin N Blattman Infectious Diseases Phoenix VA Healthcare System

Clostridium difficile CRISTINA BAKER, MD, MPH INFECTIOUS DISEASE PARK NICOLLET/HEALTH PARTNERS 11/9/2018

Title: Fecal microbiota transplantation in recurrent Clostridium difficile infection in a patient with concomitant inflammatory bowel disease

Clinical Review Criteria Fecal Microbial Transplant for Treatment of C. Difficile Infection Fecal GI Infusion Fecal Capsule (G3 OpenBiome)

Fecal microbiota transplantation: Breaking the chain of recurrent C. difficile infection

! Macrolide antibacterial. Fidaxomicin (Dificid ) package labeling. Optimer Pharmaceuticals, Inc. May 2011.

Literature Scan: Antibiotics for Clostridium difficile Infection. Month/Year of Review: May 2015 Date of Last Review: April 2012

Fecal Microbiota Transplantation. Description

Clostridium Difficile Infection in Adults Treatment and Prevention

Probiotics- basic definition

The good, the bad and the ugly. Our microbiome and it s effect on health and disease.

ACG Clinical Guideline: Diagnosis, Treatment, and Prevention of Clostridium difficile Infections

CLINICAL MEDICAL POLICY

Journey to Decreasing Clostridium Difficile and the Unexpected Twist. Jackie Morton, Infection Prevention Cortney Swiggart, Medication Safety Officer

Case 1. Which of the following would be next appropriate investigation/s regarding the pts diarrhoea?

Managing Clostridium Difficile: An Old Bug With

HEALTHCARE- ASSOCIATED INFECTIONS: A FOCUS ON Clostridium difficile

Homeopathic Products. Evidence??

CLINICAL MEDICAL POLICY

Treatment Update on Fecal Microbiota Transplantation. Arnab Ray, MD Ochsner Clinic Foundation Gastroenterology Department

What GI Physicians Need to Know About Probiotics

THE USE OF LACTOBACILLUS IN THE TREATMENT OF CLOSTRIDIUM DIFFICILE INFECTION IN HOSPITALIZED ADULT PATIENTS

Le infezioni da Clostridium difficile, gravi, ricorrenti e complicate Nicola Petrosillo

SMT19969: A Selective Therapy for C. difficile Infection

Gut Microbiota Transplant Pro Position. Christina Surawicz, MD, MACG Professor of Medicine University of Washington Seattle WA

Clinical Policy Title: Fecal transplantation for clostridium difficile infection

(No Image Selected) Video Submission Confirmation: No Video Upload: Abstract Author: Investigator Commercial Products or Services: No Designed Study:

Clostridium difficile Infection (CDI) Management Guideline

OCTOBER 7-10 PHILADELPHIA, PENNSYLVANIA

Probiotics for children receiving antibiotics

The Potential For Microbiome Modification In Critical Illness. Deborah Cook

Antibiotic treatment comparison in patients with diarrhea

March 3, To: Hospitals, Long Term Care Facilities, and Local Health Departments

DISCLOSURE Relevant relationships with commercial entities Wyeth (received advisory board & speaker honoraria) Potential for conflicts of interest wit

9/18/2018. Clostridium Difficile: Updates on Diagnosis and Treatment. Clostridium difficile Infection (CDI) Clostridium difficile Infection (CDI)

Understanding probiotics and health

Treating and Preventing. C. difficile Infections A Review of the Research for Adults

Atypical Presentation of Clostridium Difficille Infection (CDI).

Clostridium Difficile colitismore

Clostridium Difficile Infection: Applying New Treatment Guidelines and Strategies to Reduce Recurrence Rate

Responders as percent of overall members in each category: Region: New England 50 (57% of 87 members) 46 (57% of 81 members) 21 (55% of 38 members)

Antibiotic Associated Diarrhea (AAD) and Clostridium Difficile Infection (CDI)

Efficacy of a synbiotic chewable tablet in the prevention of antibiotic-associated diarrhea

Treatment of Clostridium Difficile Infection in Community Teaching Hospital: A Retrospective Study

Is FMT the answer? Challenging Cases in CDI. Ted Steiner, M.D. April 1, 2016

C. difficile Infection: How it all comes out

Microbiome GI Disorders

DETECTION OF TOXIGENIC CLOSTRIDIUM DIFFICILE

Fecal transplantation as a treatment option for recurrent Clostridium difficile infection

Labeled Uses: Treatment of Clostiridum Difficile associated diarrhea (CDAD)

Predictors of Mortality and Morbidity in Clostridium Difficile Infection

CDI The Impact. Disclosures. Acknowledgments. Objectives and Agenda. What s in the Name? 11/14/2012. Lets Talk Numbers

Fecal microbiota transplantation: The When,the How and the Don t. By Dr Rola Hussein

PROBIONA. PROBIOTICS with 5 bacterial strains. Suitable during and after the use of antibiotics to restore intestinal microflora.

Clostridium difficile infections: Drug treatment re-evaluated

Long-Term Clinical Outcome of Clostridium difficile Infection in Hospitalized Patients: A Single Center Study

Clostridium difficile (C difficile)

FECAL TRANSPLANTATION

A Pharmacist Perspective

Manipulating the gut microbiome

Fecal Microbiota Transplantation for Severe sepsis and Diarrhea : a Case Report

CLOSTRIDIUM DIFFICILE: IMPROVING DIAGNOSIS AND TREATMENT. Joshua T. Watson, M.D. Lowcountry Gastroenterology Associates

Duodenal infusion of donor feces for recurrent Clostridium difficile infection A French experience

Fecal Microbiota Transplantation

more intense treatments are needed to get rid of the infection.

Diagnosis, Management, and Prevention of Clostridium difficile infection in Long-Term Care Facilities: A Review

Disclosure. Objectives. Assessment Questions. History. Clinical Case 2/27/2015. Clostridium difficile update and new therapies

Pthaigastro.org. Drugs Used in Acute Diarrhea: Cons. Nipat Simakachorn, M.D. Department of Pediatrics Maharat Nakhon Ratchasima Hospital.

!Microbiology Profile, stool

Nicola Petrosillo Istituto Nazionale per le Malattie Infettive Lazzaro Spallanzani, IRCCS Roma. L infezione da C difficile grave o complicata

Azienda Ospedaliera S. Camillo Forlanini. Unità Operativa di Gastroenterologia. Moscow June Cosimo Prantera

12/1/2017. Disclosures. Objectives. The Microbiome Defined

MEDICAL POLICY SUBJECT: FECAL BACTERIOTHERAPY EFFECTIVE DATE: 08/16/12 REVISED DATE: 08/15/13, 07/17/14, 07/16/15, 06/16/16, 06/15/17

What Are Probiotics? PROBIOTICS

Transcription:

International Journal of Food and Allied Sciences ISSN: 2415-0290 (Print) ISSN: 2413-2543 (Online) DOI:10.21620/ijfaas.2017120-26 Research Article History The Role of Saccharomyces boulardii in the Treatment of Refractory Recurrent Clostridium difficile Infection Rachel M Warila a, *Rebecca M Hoover b a College of Pharmacy, Idaho State University, Pocatello, ID, USA b Director Idaho Drug Information, Department of Pharmacy Practice and Administrative Sciences, College of Pharmacy, Idaho State University, Pocatello, ID, USA Abstract Submitted: Nov 11, 2016 1 st Revision: Dec 12, 2017 2 nd Revision: Aug 29, 2017 Accepted: Sep 03, 2017 Online: Sep 12, 2017 Keywords Clostridium difficile, probiotics, recurrent, Saccharomyces boulardii *Correspondence hoovrebe@isu.edu 208-282-2587 To review the literature surrounding the efficacy of S. boulardii for the treatment of recurrent C. difficile infections. Eligible articles included S. boulardii in patients with recurrent C. difficile infection. The primary endpoint examined was clinical resolution of infection with no further recurrences during followup. Results: Six studies met inclusion criteria. A case report showed resolution of recurrences in one patient, and an experimental trial showed a trend towards decreased recurrences in patients receiving S. boulardii (85% no further recurrences). Two randomized controlled trials found a significant decrease in recurrences for S. boulardii versus placebo (34.6% vs 64.7%, P=0.04; 16.7% vs 50%, P=0.05). One metaanalysis determined significant efficacy for S. boulardii in reducing relapses (RR 0.59, 95% CI 0.35-0.98), while another concluded there was insufficient evidence to recommend probiotics for C. difficile infection. S. boulardii may be considered adjunctively for patients with recurrent C. difficile infection who are refractory to antibiotic regimens alone). 1. Introduction Clostridium difficile is a gram positive, spore-forming, anaerobic bacilli and the most frequent perpetrator of nosocomial infections affecting the gastrointestinal tract (Salliet, 2010; Segarra-Newnham, 2007). Clostridium difficile infections (CDI) are a major burden to the U.S. health system, accruing costs of about 3.2 billion dollars every year and is associated with a high rate of morbidity and mortality, resulting in possible bowel perforation, sepsis, or death (Surawicz, Brandt & Binion, 2013; Leong & Zelenitsky, 2013). Risk factors for the disease include recent antibiotic use, proton-pump inhibitors or other acid-suppressive therapies, advanced age, and immunosuppression. Altered normal flora as a result of antibiotic treatment greatly increases risk of CDI, but paradoxically, antibiotics are also first-line therapy for treatment. Antibiotics are 80% effective for the first CDI episode; however as the gut microbiota becomes further disrupted, risk of CDI relapse rises. CDIs reoccur in approximately 20% of patients and multiple recurrences lead to an even greater risk of relapse at about 50-60% (Segarra-Newnham, 2007). Recurrences are thought to arise when spores surviving an antibiotic treatment course germinate and reinitiate disease in the absence of protective gut flora (Surawicz, Brandt & Binion, 2013). A novel theory to combat recurrences uses colonization resistance by introducing healthy bacteria into the gut to restore the normal flora and prevent C. difficile from obtaining essential nutrients and surviving (Leong & Zelenitsky, 2013). This theory has led us to experimental treatments such as probiotics and fecal microbial transplant (FMT), in which feces of a healthy donor are implanted into the gastrointestinal tract of the diseased patient via oral, nasal, or rectal routes. Even patients who have had only a single CDI episode have markedly different bacterial flora and less bacterial diversity compared healthy individuals which demonstrates the great need to identify an effective therapy for restoring normal gut flora in these patients (Na & Kelly, 2011). Probiotics are readily accessible to patients in stores and online, making them a mainstay of preventative therapy. Common products include single or multi-strain probiotic capsules, live culture probiotic beverages such as kombucha (fermented tea) or kefir, (fermented milk) as well as many food products like yogurts. Probiotics International Journal of Food and Allied Sciences, 2017, 1(1): 20-26 20

provide benefits such as reinforcing the intestinal lining that acts as a barrier to invading organisms, enhancing the immune system, and increasing the growth and proliferation of the normal flora. (Surawicz, Brandt & Binion, 2013; Na & Kelly, 2011). Saccharomyces boulardii is a nonpathogenic yeast probiotic that has been studied in the treatment of recurrent CDI. S. boulardii produces a protease that cleaves C. difficile s toxins as well as their corresponding membrane receptors on the host intestinal cells (Castagliuolo, Riegler, Valenick, LaMont & Pothoulakis, 1999). By this mechanism, even after re-growth, C. difficile cannot reinitiate disease because its toxins can no longer bind their receptors (Surawicz, et al. 2000). S. boulardii grows at body temperature of 37 C and reaches a high steadystate concentration of 10 7 to 10 8 colony forming units in the colon in just a few days. The yeast is resistant to antibiotics, has a half-life of 2-5 days, and has been shown to clear the gastrointestinal tract after discontinuing use (McFarland, 2010). S. boulardii is readily available inexpensively for 0.21 USD/capsule as a generic product, or under the trade name Florastor (Biocodex, France) in the United States as lyophilized (freeze-dried) capsules, which are stable at room temperature for over a year. S. boulardii-containing probiotic capsules with their low cost, ease of use, and desirable administration may have a role for patients with recurrent CDI, defined as at least one prior episode previously treated with antibiotics. In this literature review, relevant sources were identified through a literature search of PubMed (1962 to February 2016), Cochrane Library (1990 to 2015), International Pharmaceutical Abstracts (2000 to 2015), and CINAHL (2001 to 2015) using the following keywords: Clostridium difficile, probiotic, recurrent, and Saccharomyces boulardii. Selection requirements included case reports, clinical studies, and meta-analyses utilizing S. boulardiicontaining probiotics in the treatment of recurrent CDI as the primary outcome. Inclusion criteria were human data and studies published in English. Excluded studies comprised antibiotic-associated diarrhea as the primary outcome or treatment of recurrent CDI as a secondary outcome. One study was excluded because probiotic efficacy was determined by measuring the concentration of probiotic in stool samples, a practice determined to be clinically irrelevant (Elmer, McFarland, Surawicz, Danko & Greenberg, 1999; Pillai & Nelson, 2008). However, the complete outcome data were included in with a later study (Surawicz, 2000). Studies reviewed comprised one case report, one experimental trial, two randomized controlled trials, and two meta-analyses. Literature Review: 1.1 Case Reports A 1990 case report described a 67-year-old woman with multiple recurrences of CDI (Kimmey, Elmer, Surawicz, McFarland, 1990). Over the course of 38 weeks the woman experienced 8 episodes of recurrent CDI treated with multiple vancomycin tapers. Shortly after her eighth CDI episode, S. boulardii was initiated, 250 mg capsules two by mouth twice daily, concurrently with a vancomycin taper. One week later, vancomycin was stopped and S. boulardii was continued for 2 months and then tapered to one capsule twice daily for 10 days. Follow-ups at 10 and 18 months after discontinuing S. boulardii determined that she experienced no further recurrences. While case report results have limited applicability to larger populations, this report demonstrates S. boulardii as a potential adjunctive therapy for recurrent CDI. 1.2 Experimental Trials Surawicz et al. performed an open trial in 1989 using S. boulardii adjunctively to vancomycin in 13 patients with recurrent CDI (average of 3.6 recurrences). Patients were given a combination of vancomycin 250 mg orally four times daily for 10 days, plus S. boulardii 2, 250 mg capsules twice daily started on day 5 of vancomycin treatment and continued for at least 30 days. The rationale for starting S. boulardii after 5 days of vancomycin and continuing beyond antibiotic therapy was explained that S. boulardii efficacy may be dependent on the ability of vancomycin to first clear C. difficile from the GI tract, then allowing S. boulardii to protectively colonize. Success was defined as no recurrence of diarrhea (3 or more loose stools per day for at least 2 consecutive days) for the duration of S. boulardii treatment. Eleven of the thirteen patients (85%) reported no further recurrences within the 30 days of receiving S. boulardii, while two (15%) failed treatment. Both patients remained C. difficile stool 21

culture positive following treatment, so it is possible that the vancomycin never cleared C. difficile, compared to the 11 successfully treated patients of which 82% were cytotoxin negative (P<0.001) following treatment. Seven patients were contacted (range 2-150 days, median 6 days) after cessation of S. boulardii and reported no further recurrences. This study s limitations included small patient population, lack of a control group or randomization to antibiotic groups, and the follow-up may not have been long enough to detect delayed recurrences. 1.3 Randomized Controlled Trials A double-blind randomized placebo-controlled trial utilized S. boulardii in combination with antibiotics for 64 patients with an initial episode of CDI and 60 with a recurrence (McFarland et al., 1999a). Patients received metronidazole or vancomycin (chosen by the physician) in combination with either placebo or S. boulardii (two 250 mg capsules twice daily) started within 4 days of antibiotics and continued for 4 weeks with an additional 4-week follow-up. Study drug failures were defined as presence of diarrhea, pseudomembranes, or a positive C. difficile assay. Of the 124 patients, 30 of 67 (44.8%) failed treatment on placebo whereas 15 of 57 (26.3%) failed treatment on S. boulardii (P=0.05). Multivariate analysis was performed and S. boulardii was shown to be significant in reducing recurrences in patients with a history of recurrences, (34.6% vs 64.7%, P=0.04) but not in patients presenting with their first CDI episode (19.3% vs 24.2%, P=0.86). Adverse reactions reported with S. boulardii were increased thirst (9%) and constipation (14%). This study suggests that S. boulardii may be useful following multiple CDI recurrences when the gut flora has been markedly altered. Limitations include that patients were not randomized to antibiotic groups and this study was underpowered to detect a significant difference in patients presenting with their first episode. An add-on study to the previous attempted to control for the dose and duration of antibiotics in combination with S. boulardii in hopes of defining a specific regimen for the treatment of recurrent CDI (McFarland et al., 1999b). The study extracted results from a different trial, which randomized patients to three antibiotic groups: metronidazole (1 g/day), low-dose vancomycin (500 mg/day), and high-dose vancomycin (2 g/day), plus S. boulardii. Both the metronidazole and low-dose vancomycin groups were found to be nonsignificant in reducing recurrences compared to placebo, and therefore this sub-study focused only on patients prescribed high-dose vancomycin. Patients were prescribed a 10-day course of high-dose vancomycin (2 g/day) plus placebo or S. boulardii two 250 mg capsules twice daily started on day 7 of vancomycin treatment and continued for 28 days. Study duration was 8 weeks with an additional 5-month follow-up. Of the 168 patients, only 32 were prescribed high-dose vancomycin due to more severe infection and all had a history of at least one or more prior episode of CDI within the past year. Drug failures were defined as presentation of diarrhea after discontinuation of antibiotics and before the end of the 8-week study duration. Of the patients on high-dose vancomycin, 3 of 18 (16.7%) taking S. boulardii experienced a relapse compared with 7 of 14 (50%) taking placebo (P=0.05). Four patients receiving placebo also experienced two relapses during the trial. During the 5-month follow-up, 29 patients were successfully contacted and 3 of 13 (23%) receiving placebo had recurred compared to zero patients taking S. boulardii. No adverse reactions were reported. Metronidazole and low-dose vancomycin were ineffective in the parent study possibly because C. difficile was not completely cleared. In these groups, 41% of metronidazole patients and 11% of low-dose vancomycin patients remained C. difficile positive following antibiotic treatment, versus 100% clearance in the high-dose vancomycin group. It is also important to note that the 1000 mg/day dose of metronidazole may have been subtherapeutic. The Infectious Diseases Society of America recommends 1500 mg daily (Cohen, Gerding & Johnson, 2010). A limitation of this study was the small number of patients receiving highdose vancomycin due to randomization based on disease severity. As these patients presented with more severe disease, they may be inherently different from the other two antibiotic groups. The results may only be applicable to this specific patient population. 1.4 Meta-analyses In a meta-analysis in 2006 of probiotics for prevention of antibiotic associated diarrhea and treatment of CDI, McFarland et al. evaluated 6 randomized controlled trials (354 patients) for CDI and 25 (2,810 patients) for 22

antibiotic associated diarrhea. Of the 6 CDI studies, 2 using S. boulardii reported a significant effect of Table 1: Summary of Studies Evaluating S. boulardii for the Treatment of Recurrent CDI Reference Study Type Patients Treatment/Duration Follow-up Results Kimmey (1990) Case Report n=1 S. boulardii: 2x250 mg caps twice daily x 2 months then tapered to 1 capsule twice daily x 10 days Antibiotics: vancomycin taper (dose not specified) x 1 week 10, 18 months No recurrences Surawicz (1989) Experimental Trial n=13 S. boulardii: 2x250 mg caps twice daily started on day 5 of vancomycin, continued for 30 days 30 days 11/13 (85%) reported no recurrences Antibiotics: vancomycin 250 mg four times daily x 10 days McFarland (1994) Randomized Controlled Trial n=124 (64 pts with an initial episode, 60 with a recurrence) S. boulardii: 2x250 mg caps twice daily started within 4 days of antibiotics x 4 weeks or placebo Antibiotics: metronidazole or vancomycin according to prescriber discretion (dose, duration not specified) 4 weeks S. boulardii in initial episode: (19.3% vs 24.2%, P=0.86) S. boulardii in history of recurrences: (34.6% vs 64.7%, P=0.04) Surawicz (2000) Randomized Controlled Trial n=168 (32 in highdose vancomycin group) S. boulardii: 2x250 mg caps twice daily started on day 7 of vancomycin x 28 days or placebo Antibiotics: metronidazole (1 g/day), low-dose vancomycin (500 mg/day), or high-dose vancomycin (2 g/day) x 10 days 8 weeks with additional 5 month follow-up S. boulardii plus high-dose vancomycin: 3/18 (16.7%) relapsed vs 7/14 (50%) taking placebo (P=0.05) 23

McFarland (2006) Meta-analysis n=354 (initial or recurrent CDI; one primary prevention study n=138) S. boulardii or placebo: 44% of patients. Other studies included different strains Antibiotics: varied among trials Varied Probiotics in frequency of CDI: (RR 0.59, 95% CI 0.41-0.85, P=0.005) S. boulardii only strain that significantly reduced CDI recurrences Pillai (2008) Meta-analysis n=328 (initial or recurrent CDI) S. boulardii or placebo: 89% of patients. Other studies included different strains Antibiotics: varied among trials Varied S. boulardii in initial or recurrent disease: (RR 0.59, 95% CI 0.35-0.98) probiotics versus placebo in reducing the incidence of recurrent CDI, while 4 studies using non-s. boulardii probiotics determined there was no difference. Both studies showing significance are discussed above and of note, these two studies and this meta-analysis were written by the same author. Combined efficacy for the 6 studies showed that probiotics are significantly effective for decreasing risk of CDI recurrences (RR for CDI 0.59 [95% CI 0.41-0.85] P=0.005). S. boulardii was the only probiotic strain that exhibited significant efficacy for reducing recurrences. Adverse effects reported were thirst and constipation with S. boulardii and no cases of fungemia, bacteremia, or other serious events were reported in the 31 trials and over 3,000 patients included. The authors concluded that probiotics are effective for the treatment of CDI. Conversely, a Cochrane review determined that there is insufficient evidence to recommend probiotics as an adjunct to antibiotics for the treatment of CDI (Pillai & Nelson, 2008). Four randomized controlled trials of probiotics in combination with antibiotics were evaluated, two of which are discussed above, while the other two used probiotics other than S. boulardii (Surawicz, 2000; McFarland, 1994; Wullt, Haglslatt, Odenholt, 2003; Lawrence, Korzenik, Mudy, 2005). The authors determined clinical heterogeneity among studies in terms of antibiotic use and defining recurrent CDI were such that no current recommendation can be made. The authors also stated that the trials analyzed were likely too small and lacked the power to detect a significant difference. Discussion: The 2013 American College of Gastroenterology guidelines for CDI conclude there is only weak evidence for S. boulardii in recurrent CDI, but fecal microbiota transplant (FMT) may be considered after a third recurrence and failure of a pulsed vancomycin regimen. The 2010 SHEA/IDSA guidelines for CDI reference the McFarland et al. clinical trial stating that S. boulardii in combination with vancomycin appears to decrease recurrences. However, they expressed about possible fungemia in immunocompromised patients (Cohen, Gerding, Johnson, 2010). The authors conclude that there is no compelling evidence for probiotics in the treatment of CDI recurrences. Although S. boulardii is traditionally safe and nonpathogenic, probiotics are not completely benign and the possibility of fungemia exists. Some major risk factors for bloodstream infections include severe immunosuppression and prematurity in infants, while minor risk factors include presence of a central venous catheter, an impaired intestinal barrier (diarrhea or inflammation), concomitant administration of a broadspectrum antibiotic that does not cover the probiotic, and administration of the probiotic via a J-tube. 2 Despite this, overall risk of bacteremia or fungemia from probiotic organisms is extremely low (< 1/1,000,000), 24

and there is no evidence that risk of infection from a probiotic is any greater than risk from a commensal organism (Hedge, Strain, Heins, Farver, 2008). Additionally, it is believed that bloodstream infection from one of these organisms is a marker of extremely bad health and poor prognosis. In general, the above studies in patients cited to be at increased risk of fungemia demonstrate only minor adverse reactions associated with S. boulardii and no serious adverse events were reported, although it is important to note that most excluded severely immunocompromised patients. Conclusion: Efficacy data for S. boulardii in recurrent CDI were limited and the studies analyzed used small patient populations with varying follow-up, which may have concealed further recurrences. Durations of antibiotics and probiotics among studies also varied making a precise recommendation on when and how to use S. boulardii in recurrent CDI difficult. Nonetheless, the studies examined generally showed benefit in patients taking the probiotic with minimal risk, demonstrating that it might be a reasonable option to consider in some patients. S. boulardii may be of benefit in patients who have suffered from multiple recurrences, having exhausted antibiotic avenues such as tapers, and are aesthetically or financially hesitant to undergo invasive FMT, as this is currently the only guideline-recommended option. If a patient is willing to attempt a trial of S. boulardii and is unsuccessful resulting in subsequent recurrences, FMT should be strongly considered. In those with severe immunosuppression or a central venous catheter, probiotics should be used cautiously or alternative treatment should be selected. S. boulardii is readily available, inexpensive, desirable to consumers, and poses little risk in individuals without severe immunosuppression. Due to the wide array of probiotic products available on the market, products with regulatory quality control programs should be selected if possible. As an adjunct treatment to antibiotics, S. boulardii may be an affordable and accessible option for trial in patients with recurrent CDI, refractory to multiple antibiotic regimens, before resorting to undergo invasive FMT. 7. References Cohen, S.H., Gerding, D.N., Johnson, S. et al. (2010). Clinical practice guidelines for Clostridium difficile infection in adults: 2010 update by the society for healthcare and epidemiology of America (SHEA) and the infectious diseases society of America (IDSA). Infect Control Hosp Epidemiol, 31(5): 431-455. doi:10.1086/651706 Elmer, G.W., McFarland, L.V., Surawicz, C.M., Danko, L., & Greenberg, R.N. (1999). Behaviour of Saccharomyces boulardii in recurrent Clostridium difficile disease patients. Aliment Pharmacol Ther, 13(12):1663-8. doi:10.1046/j.1365-2036.1999.00666.x Hedge, D.D., Strain, J.D., Heins, J.R., & Farver, D.K. (2008). New advances in the treatment of Clostridium difficile infection (CDI). Ther Clin Risk Manag 2008;4(5):949-964. Kimmey, M.B., Elmer, G.W., Surawicz, C.M., & McFarland, L.V. (1990). Prevention of further recurrences of Clostridium difficile colitis with Saccharomyces boulardii. Dig Dis Sci, 35(7):897-901. Lawrence, S.J., Korzenik, J.R., & Mundy, L.M. (2005). Probiotics for recurrent Clostridium difficile disease. J Med Microbiol 2005;54:905 6. 0 Leong, C. & Zelenitsky, S. (2013). Treatment strategies for recurrent Clostridium difficile infection. Can J of Hosp Pharm, 66(6):361-8. McFarland, L.V., Surawicz, C.M., Greenberg, R.N. et al. (1994). A randomized placebo-controlled trial of of Saccharomyces boulardii in combination with standard antibiotics for Clostridium difficile disease. JAMA, 271(24):1913-8. McFarland, L.V., Surawicz, C.M., Rubin, M., Fekety, R., Elmer, G.W., & Greenberg, R.N. (1992). Recurrent Clostridium difficile disease: epidemiology and clinical characteristics. Infect Control Hosp Epidemiol, 20(1): 43-50. McFarland, L.V. (2006). Meta-analysis of probiotics for prevention of antibiotic associated diarrhea and treatment of Clostridium difficile disease. Am J Gastroenterol, 101(4): 812 22. McFarland, L.V. (2010). Systematic review and meta-analysis of Saccharomyces boulardii in adult patients. World J Gastroenterol, 16(18): 2202 22. Na, X. & Kelly, C. (2011). Probiotics in Clostridium difficile infection. J Clin Gastroenterol, 45:(Suppl)154-8. Pillai, A. & Nelson, R.L. (2008). Probiotics for treatment of Clostridium difficile-associated colitis in adults. Cochrane Database Syst Rev, 1:CD004611. Sallit, J. (2010). C.diff prevention and adjunctive therapy with prebiotics & probiotics. Connections newsletter, Dietetics in Healthcare Communities, Academy of Nutrition and Dietetics. 35,2:1-8.http://www.amerinetgpo.com/Portals/4/Documents/C-diff-Prevention-whitepaper.pdf Date Accessed: February 10, 2016 Segarra-Newnham, M. (2007). Probiotics for Clostridium difficileassociated diarrhea: focus on Lactobacillus rhamnosus GG and Saccharomyces boulardii. Ann Pharmacother; 41(7):1212 21. 25

Surawicz, C.M., Brandt, L.J, Binion, D.G. et al. (2013). Guidelines for diagnosis, treatment, and prevention of Clostridium difficile infections. Am J Gastroenterol, 108(4): 478-98. Surawicz, C.M., McFarland, L.V., Elmer, G., & Chinn, J. (1989). Treatment of recurrent Clostridium difficile colitis with vancomycin and Saccharomyces boulardii. Am J Gastroenterol 84(10): 1285-7. Surawicz, C.M., McFarland, L.V., Greenberg, R.N. et al. (2000). The search for a better treatment for recurrent Clostridium difficile disease: the use of high dose vancomycin combined with Saccharomyces boulardii. Clin Infect Dis, 31(4): 1012 7. 26