Skip to main content
Therapeutic Advances in Gastroenterology logoLink to Therapeutic Advances in Gastroenterology
. 2019 May 6;12:1756284819847651. doi: 10.1177/1756284819847651

Management of Clostridioides difficile colitis: insights for the gastroenterologist

Srishti Saha 1, Sahil Khanna 2,
PMCID: PMC6505238  PMID: 31105766

Abstract

Clostridioides difficile infection (CDI) is a common cause of diarrhea in both inpatient and outpatient settings. The last few years have seen major changes in the treatment spectrum of CDI, most notably, recommendations against using metronidazole for initial CDI, the addition of fidaxomicin and bezlotoxumab, and emergence of microbial replacement therapies. Several other therapies are undergoing clinical trials. This narrative review focuses on the treatment of CDI with a summary of literature on the newer modalities and the treatment guidelines issued by Infectious Diseases Society of America and European Society of Clinical Microbiology and Infectious Diseases.

Keywords: Clostridioides difficile, Clostridium difficile, C. difficile diarrhea, fecal microbiota transplantation

Introduction

Clostridioides difficile (previously Clostridium difficile) is the most common pathogen implicated in healthcare-associated infections in the United States (US).1 Recent data suggest that ~40% of C. difficile infections (CDIs) in the US are community-associated, which often do not have risk factors such as antibiotic exposure or hospitalization commonly associated with healthcare-associated CDI.2 In Europe, the incidence of CDI is lower than in the US.3,4 It is the eighth most frequently reported microorganism in healthcare-associated infections, with increasing incidence in most countries; the majority of cases (76.4%) are healthcare-associated.5,6

CDI classically presents with watery diarrhea (Bristol stool scale 6–7, three or more times a day) and crampy abdominal pain, with or without fever. In rare cases with ileus or a megacolon, diarrhea may not be present. Diagnosis is made by a positive stool test in the presence of typical clinical features. A gamut of tests is available, and the choice of test depends on the institutional protocol and test availability.

Management of CDI has changed significantly in the last few years, with several new treatment options available. In this article, we describe the treatment of primary and recurrent CDI in adults as recommended by recent guidelines along with investigational therapies for CDI.

Classification of CDI episodes

Prior to starting treatment for CDI, it is essential to grade the severity of the episode, and also note whether there were prior episodes. CDI is classified as mild-to-moderate, severe or fulminant (earlier termed severe-complicated) based on laboratory parameters and clinical features. The commonly used criteria for classification are as follows7,8:

  • Severe disease is diagnosed in the presence of white blood cell (WBC) count > 15,000 × 106/l, or creatinine rise >1.5-times baseline or >1.5 mg/dl

  • Fulminant disease is diagnosed when there is hypotension, shock, sepsis, intensive care unit admission, megacolon, perforation or colectomy due to CDI

  • Mild-to-moderate disease is diagnosed when the criteria for severe or fulminant disease are not met.

Several other classification criteria have been used to determine severity [ATLAS, American College of Gastroenterology (ACG) 2013, European Society of Clinical Microbiology and Infectious Diseases (ESCMID) 2014 criteria and others], though none have been accepted universally.911 Most of these scores are based on expert opinion. Validation studies have yielded variable results, though acute kidney injury and leukocytosis have consistently predicted severe disease.1115 Large, well conducted prospective validation studies are needed to shed light on the best criteria for use in clinical practice.

The earlier guideline from the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA) from 2010 recommended different treatments based on severity of the episode, thus requiring the use of laboratory tests (e.g. WBC, creatinine) as a guide to treatment. The updated guideline recommends the same treatment for mild-to-moderate and severe episodes, no longer mandating these tests as a part of the initial evaluation of a patient. The ESCMID guidelines retain the need for severity criteria in guiding treatment options.

The first episode of CDI in a patient is termed primary CDI. Recurrent CDI is defined as recurrence of typical symptoms within 8 weeks of the previous episode, with documented symptom resolution in the interim. Physicians need to be careful when interpreting stool tests in the context of a possible recurrence. Nucleic acid amplification tests (NAATs) often remain positive following successful treatment of a prior episode. This is because they are highly sensitive and can detect small quantities of the toxin gene in the stool.

Updates on testing

The updated IDSA/SHEA 2017 guideline recommends a multistep algorithm with a stool toxin test when there are no institutional protocols for stool specimen submission.7 The algorithm uses a glutamate dehydrogenase (GDH) test plus toxin, toxin plus NAAT, or GDH plus toxin arbitrated by NAAT. Where there are predefined stool submission protocols, a single NAAT, or a toxin test as part of multistep algorithm can be used. The caveat is to test only on unformed stools in patients with watery diarrhea ⩾3 times in 24 h. There is no role of repeat testing in case of an initial negative test, unless there is a change in the clinical presentation or in epidemic settings; test of cure post-treatment is also not recommended.7

The ESCMID guidelines also support a two-step algorithmic approach to diagnosis.16 They suggest using either GDH or NAAT for screening, followed by reflex testing of positive samples with toxin A/B enzyme immunoassay (EIA). Samples with a negative second test may be tested with NAAT (if not used before) or toxigenic culture. An alternative approach is to use a combination of GDH and toxin A/B EIA for screening; samples with concordant results can be reported as such. Samples with a negative GDH and positive toxin test should be retested with NAAT or toxigenic culture. Repeat testing should be avoided for initial positive results, and no test of cure is recommended; initial negative results can be retested if there is ongoing clinical suspicion during an epidemic situation or when there is high suspicion during endemic situations.16

In situations where a patient tests positive for C. difficile but does not have the typical symptoms, a positive test represents an asymptomatic carrier who needs no further treatment. The clinical picture is often further complicated when patients have diarrhea after a prior CDI episode, with or without a positive stool test. Studies have shown that ~25% patients develop post-infectious irritable bowel syndrome following an episode of CDI.17,18

Management

All inpatients with CDI should be isolated; infection control measures should be initiated and continued till discharge. Concomitant acid suppressant medications and opioid medications increase the risk of severe disease and should not be used.19 Systemic antibiotics herald a poor outcome and increase recurrences, hence must be discontinued when possible. Continued proton pump inhibitor (PPI) use also increases the risk of recurrence, hence should be stopped.20,21

Antimicrobial treatment

The healthy human gut is host to a diverse microbiota which protects it from pathogenic bacteria by competitive exclusion and production of bacteriocins. When this microbial milieu is perturbed (‘gut dysbiosis’), the host becomes susceptible to infections. In order to develop CDI, both gut dysbiosis and colonization with C. difficile bacterium are predisposing factors. The ideal therapeutic agent for CDI is one that successfully kills the bacterium while restoring the disrupted gut microbiota.

Several drugs are used to treat CDI, each with its own spectrum of activity, efficacy, and toxicities. Metronidazole is the oldest drug used for CDI treatment, although it is not recommended by the United States (US) Food and Drug Administration (FDA) for this purpose and is not recommended by the 2017 IDSA/SHEA guideline in most situations.7,22 It is a broad-spectrum antibiotic used to treat anaerobic infections. Metronidazole kills the vegetative forms of the Clostridioides bacterium but not the spores.23 Thus, it terminates the infection, but leaves the patient susceptible to recurrences in the face of persistent or renewed risk factors for CDI. Metronidazole is almost completely absorbed in the small intestine with undetectable levels in the feces of healthy adults.24 In the presence of diarrhea however, the levels of the drug in feces increase; this is likely due to the decreased transit time and secretion of the drug across an inflamed colonic mucosa.24,25 Since it is absorbed from the gut, systemic side effects are of concern when repeated or prolonged courses are used.

Vancomycin is another commonly used drug for CDI. While its use in other infections is via the intravenous (i.v.) route, for CDI it is given as an oral suspension or liquid formulation. In this form, it achieves high concentrations in the stool, well above the minimum inhibitory concentration (MIC) required for its action (MIC90 for C. difficile is 1–2 mg/l).26 Although higher doses achieve higher fecal concentrations, even 125 mg dose achieves 500–1000 times the MIC90, which is adequate for clinical efficacy.26 However, the major drawbacks to its use are the propensity to select antibiotic-resistant forms of other bacteria present in the gut.27 Additionally, like metronidazole it is not sporicidal, leaving the patient vulnerable to recurrences.23

Fidaxomicin is a macrocyclic antibiotic with narrow spectrum of activity. It kills the vegetative form of C. difficile bacteria, and binds to its spores, preventing them from germinating and producing toxin23,28 It also causes less disruption of the gut microbiota compared with vancomycin.29 However, studies exploring its effect on the gut microbiota are on patients with CDI and not healthy humans. Since patients with CDI already have a disrupted microbiota, the clinical utility of this nondisruptive effect is questionable.

Rifaximin is a broad-spectrum, gut selective antibiotic used for the treatment of conditions such as hepatic encephalopathy and small bowel overgrowth. It has good in vitro activity against C. difficile and has been primarily used after a course of standard antibiotics.30 In large clinical trials, rifaximin did not meet the non-inferiority definition for cure of CDI.3032 It is recommended in the IDSA/SHEA guidelines to use as a chaser regimen after vancomycin for recurrent CDI.7 Its use in CDI is not recommended by the US FDA.

Treating the first episode

Prior to the year 2000, two randomized controlled trials (RCTs) found similar cure rates for metronidazole and vancomycin for treating CDI.33,34 However, these were small trials with <50 patients in each arm. More recently, three RCTs, with a total of 687 patients, found vancomycin to be superior to metronidazole in terms of cure rates.35,36 The two RCTs with a total of 1105 patients comparing fidaxomicin with vancomycin found similar cure rates for both drugs, but with lower recurrence rates in patients treated with fidaxomicin.37,38

On the basis of these studies, the IDSA/SHEA 2017 guideline recommends vancomycin [125 mg four times daily (QID) per oral (PO) for 10 days] or fidaxomicin [200 mg twice daily (BID) PO for 10 days] for the first mild-to-moderate/severe episode of CDI.7 Metronidazole [500 mg three times daily (TID) for 10 days] is recommended for the initial mild-to-moderate episode only if the first-line drugs are not available. This was a change from the previous guideline, wherein metronidazole (for mild-to-moderate) and vancomycin (for severe) were recommended for the first episode of CDI.8 The evidence supporting these recommendations is of high quality, leading to a strong recommendation for all regimens except metronidazole, which has a weak recommendation.7

The 2014 ESCMID guidelines recommend several options for the initial nonsevere episode.39 These include metronidazole (500 mg TID for 10 days; class IA recommendation), vancomycin (125 mg QID for 10 days; class IB recommendation), fidaxomicin (200 mg BID for 10 days; class IB recommendation), vancomycin (500 mg QID for 10 days; class IC recommendation), or observation for 48 h after stopping the inducing antibiotic (class IIC recommendation). If oral administration is not possible, i.v. metronidazole can be given (class IIA recommendation). The major difference is the recommendation for using metronidazole by ESCMID, which was changed in the IDSA/SHEA guideline following publication of complete results of the RCTs.35

Treating recurrences

For the treatment of the first recurrence, the IDSA/SHEA guideline recommends a different regimen than what was used in the first episode (Table 1). For the second or subsequent recurrence, there are several options (Table 1). The evidence supporting these recommendations is of low to moderate quality, leading to a weak recommendation for all regimens except fecal microbiota transplantation (FMT). The ESCMID guideline recommends oral vancomycin or fidaxomicin for the first recurrence, with a marginal strength of recommendation for using metronidazole (Table 2). For multiply recurrent CDI, FMT is strongly recommended; vancomycin (taper or pulse regimen) and fidaxomicin are given a moderate strength of recommendation, and metronidazole is not recommended (Table 2).

Table 1.

Treatment of recurrent Clostridioides difficile infection (IDSA/SHEA 2017 guideline).7

Indication Treatment recommended Treatment regimen Strength of recommendation/quality of evidence
First recurrence Vancomycin (if treated with metronidazole before) 125 mg QID PO for 10 days Weak/low
Fidaxomicin (if treated with vancomycin before) 200 mg BID PO for 10 days Weak/moderate
Tapered and pulsed vancomycin (if treated with vancomycin standard regimen before) 125 mg QID for 10–14 days,125 mg BID for a week, 125 mgOD for a week, and then 125 mgevery 2 or 3 days for 2–8 weeks Weak/low
Second or subsequent recurrence Tapered and pulsed vancomycin 125 mg QID for 10–14 days, 125 mg BID for a week, 125 mg OD for a week, and then 125 mg every 2 or 3 days for 2–8 weeks Weak/low
Fidaxomicin 200 mg BID for 10 days Weak/low
FMT FMT dosage, route etc. is not standardized Strong/moderate
Vancomycin followed by rifaximin chaser Vancomycin 125 mg QID PO for 10 days; rifaximin 400 mg TID PO for 20 days) Weak/low

BID, twice daily; FMT, fecal microbiota transplantation; IDSA, Infectious Diseases Society of America; OD, once daily; PO, per oral; QID, four times daily; SHEA, Society for Healthcare Epidemiology of America; TID, three times daily.

Table 2.

Treatment of recurrent Clostridioides difficile infection (ESCMID 2014 guideline).34

Indication Treatment recommended Treatment regimen Strength of recommendation/quality of evidence
First recurrence (or risk of recurrence) Vancomycin 125 mg QID PO for 10 days IB
Fidaxomicin 200 mg BID PO for 10 days IB
Metronidazole 500 mg TID for 10 days IC
Vancomycin 500 mg QID PO for 10 days IIIC
Multiply recurrent CDI Fecal or bacterial instillation Vancomycin, 500 mg QID for 4 days with bowel lavage and nasoduodenal infusion of donor feces IA
Vancomycin pulse 125 mg QID for 10 days, followed by 125–500 mg OD every 2–3 days for ⩾3 weeks. IIB
Vancomycin taper 125 mg QID for 10 days, gradually decreasing to 125 mg OD IIB
Fidaxomicin 200 mg BID for 10 days IIB
Vancomycin 500 mg QID PO for 10 days IIC
Metronidazole 500 mg TID for 10 days IID (recommendation against use)

BID, twice daily; CDI, Clostridioides difficile infection; ESCMID, European Society of Clinical Microbiology and Infectious Diseases; OD, once daily; PO, per oral; QID, four times daily; TID, three times daily.

Antibiotic regimens

Vancomycin taper/pulse regimen has not been extensively evaluated in recurrent CDI; in one study with 163 patients, a vancomycin taper/pulse lowered recurrence rates.36 However, a recent retrospective study in over 900 patients found no difference between vancomycin with and without taper regimen in terms of recurrence at 90 or 180 days.37 They also failed to find a mortality benefit of using a taper regimen. The patient population consisted predominantly of males, with a majority experiencing their first recurrence. There is a lack of consensus regarding the best dosing for the pulse/taper regimen; a recent study found lower recurrence rates when pulse dose was given to once every other followed by once every third day as compared with once every other day only.38 The duration of taper is also not standardized; a retrospective study suggested a longer duration was associated with a lower recurrence rate.39

The recommendation for using fidaxomicin for recurrent CDI comes from a subgroup analysis of patients included in the two phase III RCTs of fidaxomicin. Out of the total 1164 patients included in the trials, 128 patients who were enrolled for treatment of their first recurrence of CDI were included in this study. The study found comparable cure rates, but lower recurrence rates (19.7%) with fidaxomicin compared with a 10 day vancomycin regimen (35.5%) within 4 weeks of treatment.40 In an observational study, the recurrence rate after fidaxomicin treatment was 22% (within 4 weeks) or 19% (within 8 weeks) depending on the definition used.41 The patient population consisted primarily of those with two or more prior episodes, and the recurrence rate increased sequentially with the number of prior CDI episodes. A recent RCT studied the efficacy of fidaxomicin, FMT and vancomycin in 64 patients with multiply recurrent CDI; the primary outcome was a composite of clinical resolution and a negative C. difficile polymerase chain reaction test at the end of 8 weeks.42 They found FMT (by colonoscopy or nasojejunal tube after 4 days of vancomycin) to be superior to both fidaxomicin and vancomycin, and fidaxomicin and vancomycin to be comparable (the primary outcome achieved in 71%, 33% and 19% patients given FMT, fidaxomicin and vancomycin respectively). Recurrence within or after 8 weeks of treatment was seen in 8.3% of FMT, 45.8% of fidaxomicin, 68.7% of vancomycin-treated patients.42 This is a statistically significant difference between FMT and both antibiotics, and a comparable recurrence rate with fidaxomicin and vancomycin. The cure rates seen in this study are much lower, and the rate of recurrence is much higher than in the two previous studies; this could be due to the patient population which had a higher number of prior CDI episodes, and hence a higher baseline risk for recurrence.40,41 The small number of patients included could be another factor. This RCT supports findings from the observational study in which recurrence rates increased with the number of prior CDI episodes.41 More high-quality evidence is needed to provide better recommendations for this high-risk patient population.

Rifaximin is used as a chaser post-vancomycin for recurrent CDI. Efficacy data are limited; an RCT with 68 patients and found a lower rate of all-cause diarrhea, but not recurrent CDI-related diarrhea when compared with the placebo arm.26 The inclusion criteria were limited to patients with primary or one prior CDI episode, thus limiting applicability in multiply recurrent CDI. A recently published RCT with 151 patients found a statistically nonsignificant reduction in recurrence with rifaximin versus placebo following an episode of CDI.25 Other small open-label trials and retrospective studies found rifaximin to be comparable to standard therapies, but with highly heterogeneous study characteristics.27 Data from ongoing trials are awaited (ClinicalTrials.gov identifier: NCT01670149).

Other options such as a fidaxomicin taper course after standard treatment of recurrent CDI are being explored and have shown to be promising in small studies.43 The premise of such therapies is to control the C. difficile bacterial load in the colon, while giving time for colonization resistance to be re-established. The principle is similar to vancomycin taper regimens, with the added benefit of the narrow-spectrum activity of fidaxomicin minimizing additional gut dysbiosis. Details regarding microbial replacement for multiply recurrent CDI are given in the following section.

Overall, there is a lack of high-quality evidence for the management of the first recurrence of CDI; large, well conducted prospective studies are needed in the future to overcome this knowledge gap.

Microbial replacement therapies

FMT is a novel, highly effective therapy for treatment of CDI, with cure rates of over 80% in most clinical trials.44,45 It is based on the principle that restoration of a healthy gut microbiota will reduce susceptibility of a patient to CDI. At present, FMT is primarily used to treat recurrent CDI, though it is sometimes used for antibiotic refractory disease as well. In cases where there is the need for ongoing systemic antibiotics, FMT is delayed until after they can be stopped. Patients are kept on antibiotics (usually vancomycin or fidaxomicin) to control symptoms, until 24–72 h prior to the procedure; following FMT, antibiotics for CDI are not restarted. Mild, self-limited diarrhea is common after the procedure.46 Of note, the FDA has not approved FMT, but has allowed its use as an investigational drug.47

Since the first RCT established the efficacy of FMT in recurrent CDI, several others have followed and confirmed its efficacy.35,4850 Most trials have shown high cure rates (>80%), though meta-analyses show somewhat lower cure rates in controlled (67.7%) versus open-label trials (82.7%) after a single FMT.44,45 A concern with FMT is the lack of standardization of the procedure. There is a wide variability in all stages of the process: donor screening, donor type (related/unrelated, single/pooled, site-specific donors/stool banks), stool processing, volume of stool infused, specific stool product (whole stool/enriched), form (fresh/frozen/lyophilized), dose (single/multiple FMTs) and route of administration (oral/nasogastric/nasojejunal/enema/colonoscopy). The reporting of specific methods used is also inconsistent.51 Data are accumulating for many of these variables; studies have shown fresh and frozen stool to be equivalent and lyophilized stool to be inferior in efficacy.45,48,49 Overall, lower gastrointestinal (GI) routes are superior to upper GI routes in terms of efficacy (95% and 88% respectively), though enema has lower cure rates than colonoscopy (66% and 87% respectively).44,45,52,53 In terms of safety, minor adverse events (AEs) are more frequent in the upper GI route while serious AEs (SAEs) are more frequent in the lower GI route (minor AEs, 43.6% versus 17.7; SAEs, 2% versus 6% in the upper and lower GI routes respectively).46 In an RCT, oral capsules were as efficacious as colonoscopy, and with a more favorable safety profile.50

A small proof-of-principle RCT was done to assess the efficacy of FMT for treating primary CDI episode compared to metronidazole.54 In this trial, 20 patients were included, and primary full response (clinical cure without recurrence at day 70) was achieved in both groups equally, suggesting that FMT could be an effective treatment option for the first episode. However, the study evaluated metronidazole, which is now proven to be less effective than other first-line treatment options.

Though most studies have focused on the bacterial composition of the stool and its role in the efficacy of FMT, other components (nonbacterial organisms, bile acids and protein metabolites) may also have a role.55,56 Careful exploration of these components may in the future lead to the development of a refined stool product, theoretically with a lower risk of AEs. Several microbiome-based therapeutics are in the pipeline (RBX7455, RBX2660, SER-109), with the hope of developing a standardized stool-based product.57,58 RBX2660 is a microbiota-based suspension prepared from human stool (kept frozen, thawed before use and administered as an enema) currently undergoing a phase III trial (ClinicalTrials.gov identifier: NCT03244644). RBX7455 is an oral, room temperature stable, lyophilized microbial restoration therapy currently undergoing a phase I trial (ClinicalTrials.gov identifier: NCT02981316). SER-109 is an oral capsule containing different bacterial spores produced from purified, enriched human stool; it is currently undergoing phase III clinical trials (ClinicalTrials.gov identifiers: NCT03183128, NCT03183141). Development and approval of these microbiome-based products would reduce the heterogeneity of product administered while improving ease of administration and accessibility.

Despite the lack of standardization of stool processing methods and FMT procedure, multiple studies have proven the efficacy of FMT.44,45 The major issue at present is the scarcity of long-term safety data. Data from systematically followed up patients and registries, such as that set up by the American Gastroenterological Association (AGA), would go a long way in providing this much needed information.

Preventing recurrences

Antibody treatments

Several therapeutic modalities (e.g. an antibody against C. difficile toxins, nontoxigenic C. difficile spores, vaccines) are being evaluated for the prevention of recurrent CDI (secondary prevention). The two multicenter phase III RCTs (MODIFY I and MODIFY II) looked at the efficacy of monoclonal antibodies actoxumab (against toxin A) and bezlotoxumab (against toxin B) in preventing the recurrence of C. difficile.59 The study enrolled 2655 patients; the primary endpoint was recurrent CDI during 12 weeks of follow up. Actoxumab infusion alone was stopped after interim analysis due to higher rates of recurrence and more serious AEs. Initial cure rates were similar across groups. Bezlotoxumab and bezlotoxumab-actoxumab combination groups had a lower recurrence than placebo (decrease of 10.1% in MODIFY I and 9.9% in MODIFY II for bezlotoxumab alone; decrease of 11.6% and 10.7% in MODIFY I and II respectively for combination therapy). There was no difference between bezlotoxumab alone versus combination therapy in terms of the primary endpoint. These results held true in patients with a higher risk of recurrence. In 2016, the US FDA approved bezlotoxumab for preventing CDI recurrences in adults receiving antibiotics for the infection and with a high risk of recurrence. However, in the two trials conducted, only 4% patients were on fidaxomicin, and none received FMT as their standard of care. Considering the efficacy of these two therapies in preventing recurrences, the role of this drug in the real-world setting is yet undefined.

Nontoxigenic C. difficile

Nontoxigenic C. difficile spores protect against the colonization by toxigenic strains and halt the development of CDI; a phase II trial found very low (2%) recurrence rate within 6 weeks of treatment in patients colonized with nontoxigenic C. difficile spores.60 Phase III trials are warranted to elucidate the role of this treatment option in preventing CDI.

Are there data for vancomycin prophylaxis to prevent CDI?

Data on the efficacy of vancomycin as a CDI prophylaxis are emerging.6163 A study in 551 adult patients found vancomycin to be effective as a secondary but not primary prophylaxis, with a >50% decrease in risk of recurrent CDI.64 The protective effect was seen in patients receiving the prophylaxis for >50% duration of the antibiotic therapy; the most frequent dose used was 125 mg QID. Another study in 203 adults with prior CDI found CDI in 4.2% and 26.6% in those who did and did not receive secondary vancomycin prophylaxis.63 The follow-up period was 1 month, and vancomycin was given as 125 mg or 250 mg BID. These studies were limited by their retrospective design, limited follow up, heterogeneous duration and dose of therapy, and inability to study the effect of vancomycin on the bowel flora. Additionally, none of them reported on the occurrence of vancomycin-resistant Enterococci infections, which is a major concern with vancomycin prophylaxis. Considering the risks of using vancomycin (propagation of gut dysbiosis, selection of resistant organisms), the lack of high-quality evidence and the availability of other effective preventive strategies, at present, vancomycin prophylaxis should be used after a careful risk–benefit discussion.

Vaccines

Several vaccines against C. difficile are being developed and are currently undergoing clinical trials (ClinicalTrials.gov identifiers: NCT01887912, NCT02316470, NCT02117570, NCT02561195). This could be an effective approach in select high-risk populations, decreasing cost, morbidity and mortality for the patient. Other therapies such as ribaxamase (a beta lactamase inhibitor) are being studied.

Treating severe and severe-complicated (fulminant) disease

For the treatment of fulminant CDI, a high dose of vancomycin [500 mg QID both PO and per rectal (PR)] in addition to metronidazole (500 mg TID i.v.) is recommended by the IDSA/SHEA guideline.7 Evidence supporting this recommendation is limited to observational studies and case series.6567 These studies support the use of i.v. metronidazole in addition to vancomycin therapy. The addition of a vancomycin enema in cases with gut discontinuity or slowed transit (such as colectomy or ileus) is scientifically sound. For other cases however, data are limited and equivocal. Heterogeneity in defining severe disease also complicates the interpretation of these results. Hypothetically, use of a higher dose of vancomycin can be used to counteract the effect of slowed gut transit (in patients with ileus) which can affect fecal concentrations of the drug, although no pharmacokinetic studies have evaluated this particular subgroup of patients.

The ESCMID guideline recommends a 10 day course of oral vancomycin (125 mg QID, IA recommendation) or fidaxomicin (IB); vancomycin dose can be increased to 500 mg QID (IIIB).34 Use of metronidazole is strongly discouraged (I–D) for severe CDI. Intravenous metronidazole (500 mg TID for 10 days; IIA recommendation) with vancomycin retention enema (500 mg in 100 ml normal saline QID intracolonic), or with vancomycin PO/nasogastric tube (500 mg QID for 10 days; IIIB recommendation) can be given. For fulminant CDI, early colectomy is recommended.34

Other therapies have been evaluated for the treatment of severe/severe-complicated disease. A small study including 57 patients with severe (n = 19) and severe-complicated CDI (n = 38) found high cure rates with FMT in both groups (100% and 91% respectively).68 FMT was given when there was no response to 5 days of PO/PR vancomycin and i.v. metronidazole therapy; single or multiple FMTs were delivered through sigmoidoscopy/colonoscopy, with one patient undergoing an enema. Another case series with 17 patients found a cure rate of 88% with FMT.69

A small observational study found intravenous tigecycline (loading dose of 100 mg followed by 50 mg BID) as adjunct therapy to be marginally favorable to metronidazole-vancomycin therapy in terms of mortality.70 Tigecycline monotherapy was superior in terms of progression to complicated disease and clinical cure when compared with combination therapy in a retrospective study with 90 patients.71 A matched study did not find a difference in cure or recurrence when tigecycline was added to vancomycin therapy72; another study found no mortality benefit when it was added to standard of care in a patient cohort primarily comprising of severe-complicated CDI.73

A recent study with 287 patients found oral teicoplanin to be superior to oral vancomycin in severe or severe-complicated CDI, both in terms of cure as well as recurrence rates.74 Studies looking at alternative treatment options such as polyethylene glycol (PEG) instillation are underway, and would provide much needed insights.75 The use of i.v. immunoglobulin remains controversial with no large observational or randomized studies yet.76,77

For patients who do not respond to medical treatment, present with acute abdomen, have a rising lactate/WBC, or worsening clinical condition (organ failure, shock, sepsis), surgery is necessary. Overall, less than 1% of patients with CDI require a colectomy, but with a high associated mortality of ~30%.78 The procedure of choice is subtotal colectomy with end ileostomy. Leukocytosis and lactate levels have consistently been shown to predict worse outcomes post-surgery, while early surgery is protective.79,80

An alternative strategy using diverting loop ileostomy with colonic lavage decreased mortality while preserving the colon.81,82 However, recent data from over 400 patients in a surgical database found no difference in mortality between loop ileostomy and total colectomy, though there was benefit in terms of colon preservation.83 Despite the lack of high-quality evidence, it is prudent to say that early surgical consultation and careful monitoring of a patient’s clinical condition are vital. The choice of surgical procedure remains controversial, though loop ileostomy is clearly beneficial in colon preservation.

CDI in inflammatory bowel disease

Management of CDI in patients with inflammatory bowel disease (IBD) can be particularly challenging. This is due to lower response to the usual antibiotics, propensity to develop severe/severe-complicated disease and an increased risk of recurrences.84,85 CDI in these patients often lacks the typical risk factors such as antibiotic use, affects younger patients and is frequently community-acquired.86,87 Occurrence of CDI increases the risk of adverse outcomes such as IBD flares, intensification of therapy for IBD, colectomy and death.88,89 Due to the considerable overlap between clinical features of CDI and IBD, all patients with a suspected IBD flare should be tested for C. difficile.

Data regarding management of CDI in patients with IBD are limited. A study comparing vancomycin with metronidazole found a shorter length of stay and fewer readmissions with vancomycin in ulcerative colitis but not Crohn’s disease.90 In an open-label prospective study of 25 patients with CDI and IBD, fidaxomicin was found to be well tolerated, with cure rates of 80% and recurrence rate of 17%.91 A post-approval study of fidaxomicin evaluated its use in special populations, including IBD.92 They found similar resolution and recurrence rates in IBD and the general population with CDI at 2 and 6 months of follow up.92 In another study, resolution or improvement of diarrhea occurred in all patients with CDI and IBD treated with fidaxomicin, with a recurrence rate of 19%.41

FMT is another therapy for patients with IBD and recurrent CDI, with cure rates of 74–85.7%, and possibly higher efficacy with repeat FMT.9395 A recent study found recurrence rates after FMT to be similar in IBD and non-IBD groups with CDI; PPI use, severe CDI and hypertension predicted recurrence at 6 months.96 The risk of an IBD flare after FMT was 16%, which was lower than previous estimates, and there was no worsening of disease activity.96,97 Considering the emerging data on efficacy of FMT in treating IBD, it is an attractive option for treating patients with CDI and IBD, though treatment regimens are different.98,99

The issue of immunosuppression in patients with CDI and IBD is another major concern. Studies on the role of immunosuppression in causing adverse outcomes (colectomy, readmissions, hospital readmissions and death) have yielded conflicting results.90,100,101 Immunosuppression does not seem to affect the efficacy of FMT.93,95,96

To summarize, it seems prudent at this time to treat patients with CDI and IBD with vancomycin or fidaxomicin; FMT may be preferred in recurrent CDI. The decision regarding immunosuppression needs to be individualized. Above all, as patients with CDI and IBD are at an elevated risk of adverse outcomes, close monitoring and a low threshold for escalating therapy is essential. Ongoing clinical trials of FMT in this population will add much needed high-quality evidence (ClinicalTrials.gov identifiers: NCT03106844, NCT03829475).

Investigational therapies

Cadazolid and ridinilazole are new drugs with narrow-spectrum activity, high fecal concentrations and low systemic absorption. Both drugs were found to be well tolerated in phase II trials.102,103 The primary cure rate was comparable, and sustained cure rate (primary cure with no recurrences) was higher for all doses of cadazolid than for vancomycin, primarily due to lower recurrence rates with cadazolid.102 However, in one of the two phase III trials conducted (>600 patients in each), the primary endpoint was not achieved, and development of the drug was stopped (ClinicalTrials.gov identifiers: NCT01983683, NCT01987895).

In the published phase II trial of ridinilazole, primary cure was comparable and sustained cure (clinical cure with no recurrences within 30 days) was higher for ridinilazole than for vancomycin due to a lower recurrence rate.103 Two large multicenter phase III trials, Ri-CODIFY 1 and 2 are planned, and are expected to be completed by 2021 (ClinicalTrials.gov identifiers: NCT03595553, NCT03595566).

Surotomycin is a narrow-spectrum cyclic lipopeptide similar to vancomycin; two parallel multicenter phase III trials with over 570 patients each were conducted to evaluate surotomycin efficacy in CDI. While one trial demonstrated non-inferiority but not superiority over vancomycin, the other did not meet the non-inferiority criteria.104,105 Given these findings, the development of surotomycin was stopped.

Primary prevention strategies

The most important strategies for preventing CDI are judicious antibiotic use and infection control practices, particularly during an outbreak. However, these measures are often inadequate, implausible or ineffective. Probiotics are often used concurrently with antibiotics to prevent antibiotic-associated diarrhea. The role of probiotics in preventing CDI has been extensively studied, with most studies demonstrating benefit, especially with earlier administration.106,107 However, there is no consensus regarding the choice and dose of probiotic. Furthermore, despite a significant reduction (>50%) in CDI occurrence, the number needed to treat is high (>40), which may limit its value in clinical practice.

A recent retrospective study in 244 elderly (⩾65 years) patients looked at the efficacy of oral vancomycin prophylaxis (125 mg once daily) in primary prevention of CDI. Patients included were those who received systemic antibiotic therapy during hospital stay. CDI occurred during hospital stay in 0% and 10.4% patients in those who received prophylaxis versus those who did not62; another retrospective study found no benefit of vancomycin as a primary prevention strategy.64

Conclusion

The therapeutic landscape against CDI is rapidly evolving with accumulation of evidence from clinical trials and observational studies. At present, vancomycin and fidaxomicin are the drugs of choice in treating primary as well as recurrent CDI; the use of metronidazole is limited to severe-complicated disease. FMT should be offered to patients with multiply recurrent CDI. Further research into the treatment of recurrent and severe disease is warranted. Several drugs and microbiota-based products are undergoing clinical trials and may soon join the armamentarium available to physicians and patients to combat CDI.

Acknowledgments

S.S. and S.K. were responsible for drafting, revising and approving the manuscript.

Footnotes

Funding: Institutional funds were received from the Mayo Clinic, Rochester, MN, USA.

Conflict of interest statement: S.K. has served as a consultant for Rebiotix, Inc., Assembly Biosciences, Inc., and Summit Pharmaceuticals International. S.K. reports personal fees from Facile, ProBioTech, Premier Inc. and Shire Plc.

Contributor Information

Srishti Saha, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.

Sahil Khanna, Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA.

References

  • 1. Magill SS, O’Leary E, Janelle SJ, et al. Changes in prevalence of health care–associated infections in U.S. Hospitals. N Engl J Med 2018; 379: 1732–1744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Khanna S, Pardi DS, Aronson SL, et al. The epidemiology of community-acquired clostridium difficile infection: a population-based study. Am J Gastroenterol 2012; 107: 89–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Cassini A, Plachouras D, Eckmanns T, et al. Burden of six healthcare-associated infections on European population health: estimating incidence-based disability-adjusted life years through a population prevalence-based modelling study. PLOS Med 2016; 13: e1002150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Lessa FC, Mu Y, Bamberg WM, et al. Burden of clostridium difficile Infection in the United States. N Engl J Med 2015; 372: 825–834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. European Centre for Disease Prevention and Control. Point prevalence survey of healthcare-associated infections and antimicrobial use in European acute care hospitals 2011–2012. Stockholm, Sweden: ECDC, 2013. [Google Scholar]
  • 6. European Centre for Disease Prevention and Control. Healthcare-associated infections: clostridium difficile infections. Annual epidemiological report for 2016. Stockholm, Sweden: ECDC, 2018. [Google Scholar]
  • 7. McDonald LC, Gerding DN, Johnson S, et al. Clinical practice guidelines for clostridium difficile infection in adults and children: 2017 Update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA). Clin Infect Dis 2018; 66: 987–994. [DOI] [PubMed] [Google Scholar]
  • 8. 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: 431–455. [DOI] [PubMed] [Google Scholar]
  • 9. Surawicz CM, Brandt LJ, Binion DG, et al. Guidelines for diagnosis, treatment, and prevention of Clostridium difficile infections. Am J Gastroenterol 2013; 108: 478–98; quiz 99. [DOI] [PubMed] [Google Scholar]
  • 10. Miller MA, Louie T, Mullane K, et al. Derivation and validation of a simple clinical bedside score (ATLAS) for Clostridium difficile infection which predicts response to therapy. BMC Infect Dis 2013; 13: 148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Crobach MJT, T Planche, Eckert C, et al. European society of clinical microbiology and infectious diseases: update of the diagnostic guidance document for clostridium difficile infection. Clin Microbiol Infect 2016; 22: S63–S81. [DOI] [PubMed] [Google Scholar]
  • 12. Tariq R, Weatherly RM, Kammer PP, et al. Experience and outcomes at a specialized clostridium difficile clinical practice. Mayo Clin Proc Innov Qual Outcomes 2017; 1: 49–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Wadhwa A, Al Nahhas MF, Dierkhising RA, et al. High risk of post-infectious irritable bowel syndrome in patients with clostridium difficile infection. Aliment Pharmacol Ther 2016; 44: 576–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Shivashankar R, Khanna S, Kammer PP, et al. Clinical factors associated with development of severe-complicated clostridium difficile infection. Clin Gastroenterol Hepatol 2013; 11: 1466–1471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. 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: 440–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Garey KW, Sethi S, Yadav Y, et al. Meta-analysis to assess risk factors for recurrent clostridium difficile infection. J Hosp infect 2008; 70: 298–304. [DOI] [PubMed] [Google Scholar]
  • 17. FaDA (FDA). Drug label: Metronidazole (oral). Maryland, USA, 2018. [Google Scholar]
  • 18. Gomez A, Nguyen L, Sears P, et al. Fidaxomicin inhibits spore production in clostridium difficile. Clin Infect Dis 2012; 55: S162–S169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Bolton RP, Culshaw MA. Faecal metronidazole concentrations during oral and intravenous therapy for antibiotic associated colitis due to clostridium difficile. Gut 1986; 27: 1169–1172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Leclair MA, Allard C, Lesur O, et al. Clostridium difficile infection in the intensive care unit. J Intensive Care Med 2010; 25: 23–30. [DOI] [PubMed] [Google Scholar]
  • 21. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Nerandzic MM, Mullane K, Miller MA, et al. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Chilton CH, Crowther GS, Ashwin H, et al. Association of fidaxomicin with C. difficile spores: effects of persistence on subsequent spore recovery, outgrowth and toxin production. PLoS One 2016; 11: e0161200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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 (Reading, England) 2010; 156: 3354–3359. [DOI] [PubMed] [Google Scholar]
  • 25. Major G, Bradshaw L, Boota N, et al. Follow-on rifAximin for the prevention of recurrence following standard treatment of infection with Clostridium difficile (RAPID): a randomised placebo controlled trial. Gut. Epub 25 September 2018. doi: 10.1136/gutjnl-2018-316794 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. 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: 2850–2855. [DOI] [PubMed] [Google Scholar]
  • 27. Ng QX, Loke W, Foo NX, et al. A systematic review of the use of rifaximin for clostridium difficile infections. Anaerobe 2019; 55: 35–39. [DOI] [PubMed] [Google Scholar]
  • 28. Teasley DG, Gerding DN, Olson MM, et al. Prospective randomised trial of metronidazole versus vancomycin for clostridium-difficile-associated diarrhoea and colitis. Lancet (London, England) 1983; 2: 1043–1046. [DOI] [PubMed] [Google Scholar]
  • 29. Wenisch C, Parschalk B, Hasenhundl M, et al. Comparison of vancomycin, teicoplanin, metronidazole, and fusidic acid for the treatment of clostridium difficile-associated diarrhea. Clin Infect Dis 1996; 22: 813–818. [DOI] [PubMed] [Google Scholar]
  • 30. 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: 345–354. [DOI] [PubMed] [Google Scholar]
  • 31. Zar FA, Bakkanagari SR, Moorthi KM, et al. A comparison of vancomycin and metronidazole for the treatment of clostridium difficile-associated diarrhea, stratified by disease severity. Clin Infect Dis 2007; 45: 302–307. [DOI] [PubMed] [Google Scholar]
  • 32. Cornely OA, Crook DW, Esposito R, et al. Fidaxomicin versus vancomycin for infection with clostridium difficile in Europe, Canada, and the USA: a double-blind, non-inferiority, randomised controlled trial. Lancet Infect Dis 2012; 12: 281–289. [DOI] [PubMed] [Google Scholar]
  • 33. Louie TJ, Miller MA, Mullane KM, et al. Fidaxomicin versus vancomycin for clostridium difficile Infection. N Engl J Med 2011; 364: 422–431. [DOI] [PubMed] [Google Scholar]
  • 34. Debast SB, Bauer MP, Kuijper EJ, et al. 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. [DOI] [PubMed] [Google Scholar]
  • 35. van Nood E, Vrieze A, Nieuwdorp M, et al. Duodenal infusion of donor feces for recurrent clostridium difficile. N Engl J Med 2013; 368: 407–415. [DOI] [PubMed] [Google Scholar]
  • 36. McFarland LV, Elmer GW, Surawicz CM. Breaking the cycle: treatment strategies for 163 cases of recurrent clostridium difficile disease. Am J Gastroenterol 2002; 97: 1769–1775. [DOI] [PubMed] [Google Scholar]
  • 37. Gentry CA, Giancola SE, Thind S, et al. A propensity-matched analysis between standard versus tapered oral vancomycin courses for the management of recurrent clostridium difficile infection. Open Forum Infect Dis 2017; 4: ofx235-ofx. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Sirbu BD, Soriano MM, Manzo C, et al. Vancomycin taper and pulse regimen with careful follow-up for patients with recurrent clostridium difficile infection. Clin Infect Dis 2017; 65: 1396–1399. [DOI] [PubMed] [Google Scholar]
  • 39. Meehan AM, Khanna S, Pardi DS. Outcomes in patients with recurrent clostridium difficile infection treated with vancomycin taper. Gastroenterology 2017; 152: S348. [Google Scholar]
  • 40. Cornely OA, Miller MA, Louie TJ, et al. Treatment of first recurrence of clostridium difficile infection: fidaxomicin versus vancomycin. Clin Infect Dis 2012; 55: S154–S161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Spiceland CM, Khanna S, Pardi DS. Outcomes with fidaxomicin therapy in clostridium difficile infection. J Clin Gastroenterol 2018; 52: 151–154. [DOI] [PubMed] [Google Scholar]
  • 42. Hvas CL, Dahl Jørgensen SM, Jørgensen SP, et al. Fecal microbiota transplantation is superior to fidaxomicin for treatment of recurrent clostridium difficile infection. Gastroenterology 2018; 156: 1324–1332. [DOI] [PubMed] [Google Scholar]
  • 43. Soriano MM, Danziger LH, Gerding DN, et al. Novel fidaxomicin treatment regimens for patients with multiple clostridium difficile infection recurrences that are refractory to standard therapies. Open Forum Infect Dis 2014; 1: ofu069-ofu. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Quraishi MN, Widlak M, Bhala N, et al. Systematic review with meta-analysis: the efficacy of faecal microbiota transplantation for the treatment of recurrent and refractory clostridium difficile infection. Alimentary Pharmacol Ther 2017; 46: 479–493. [DOI] [PubMed] [Google Scholar]
  • 45. Tariq R, Pardi DS, Bartlett MG, et al. Low cure rates in controlled trials of fecal microbiota transplantation for recurrent clostridium difficile infection: a systematic review and meta-analysis. Clin Infect Dis 2018; 68: 1351–1358. ciy721-ciy. [DOI] [PubMed] [Google Scholar]
  • 46. Wang S, Xu M, Wang W, et al. Systematic review: adverse events of fecal microbiota transplantation. PLoS One 2016; 11: e0161174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. United States Food and Drug Administration. Enforcement policy regarding investigational new drug requirements for use of fecal microbiota for transplantation to treat clostridium difficile infection not responsive to standard therapies: draft guidance for industry Maryland, USA, 2016. [Google Scholar]
  • 48. Jiang ZD, Ajami NJ, Petrosino JF, et al. Randomised clinical trial: faecal microbiota transplantation for recurrent clostridum difficile infection - fresh, or frozen, or lyophilised microbiota from a small pool of healthy donors delivered by colonoscopy. Alimentary Pharmacol Ther 2017; 45: 899–908. [DOI] [PubMed] [Google Scholar]
  • 49. 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: 142–149. [DOI] [PubMed] [Google Scholar]
  • 50. Kao D, Roach B, Silva M, et al. Effect of oral capsule- vs colonoscopy-delivered fecal microbiota transplantation on recurrent clostridium difficile infection: a randomized clinical trial. JAMA 2017; 318: 1985–1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Bafeta A, Yavchitz A, Riveros C, et al. Methods and reporting studies assessing fecal microbiota transplantation: a systematic review. Ann Intern Med 2017; 167: 34–39. [DOI] [PubMed] [Google Scholar]
  • 52. Furuya-Kanamori L, Doi SA, Paterson DL, et al. Upper versus lower gastrointestinal delivery for transplantation of fecal microbiota in recurrent or refractory clostridium difficile infection: a collaborative analysis of individual patient data from 14 studies. J Clin Gastroenterol 2017; 51: 145–150. [DOI] [PubMed] [Google Scholar]
  • 53. Saha S, Khanna S. Microbiota replacement for clostridium difficile by capsule is as effective as via colonoscopy. J Thorac Dis 2018; 10(Suppl. 9): S1081–S1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Juul FE, Garborg K, Bretthauer M, et al. Fecal microbiota transplantation for primary clostridium difficile infection. N Eng J Med 2018; 378: 2535–2536. [DOI] [PubMed] [Google Scholar]
  • 55. Russell L, Monaghan T, Kao D. The need to move away from fecal transplant towards targeted, refined microbiome therapy. J Thorac Dis 2018; 10: E755–E757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Zuo T, Wong SH, Lam K, et al. Bacteriophage transfer during faecal microbiota transplantation in clostridium difficile infection is associated with treatment outcome. Gut 2018; 67: 634–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Dubberke ER, Lee CH, Orenstein R, et al. Results from a randomized, placebo-controlled clinical trial of a RBX2660-A microbiota-based drug for the prevention of recurrent clostridium difficile infection. Clin Infect Dis 2018; 67: 1198–1204. [DOI] [PubMed] [Google Scholar]
  • 58. Khanna S, Pardi DS, Kelly CR, et al. A novel microbiome therapeutic increases gut microbial diversity and prevents recurrent Clostridium difficile infection. J Infect Dis 2016; 214: 173–181. [DOI] [PubMed] [Google Scholar]
  • 59. Wilcox MH, Gerding DN, Poxton IR, et al. Bezlotoxumab for prevention of recurrent clostridium difficile infection. N Engl J Med 2017; 376: 305–317. [DOI] [PubMed] [Google Scholar]
  • 60. Gerding DN, Meyer T, Lee C, et al. Administration of spores of nontoxigenic clostridium difficile strain m3 for prevention of recurrent c difficile infection: a randomized clinical trial. JAMA 2015; 313: 1719–1727. [DOI] [PubMed] [Google Scholar]
  • 61. Ganetsky A, Han JH, Hughes ME, et al. Oral vancomycin prophylaxis is highly effective in preventing clostridium difficile infection in allogeneic hematopoietic cell transplant recipients. Clin Infect Dis 2018: ciy822-ciy, 10.1093/cid/ciy822. [DOI] [PMC free article] [PubMed]
  • 62. Papic N, Maric LS, Vince A. Efficacy of oral vancomycin in primary prevention of clostridium difficile infection in elderly patients treated with systemic antibiotic therapy. Infect Dis (Lond) 2018; 50: 483–486. [DOI] [PubMed] [Google Scholar]
  • 63. Van Hise NW, Bryant AM, Hennessey EK, et al. Efficacy of oral vancomycin in preventing recurrent clostridium difficile infection in patients treated with systemic antimicrobial agents. Clin Infect Dis 2016; 63: 651–653. [DOI] [PubMed] [Google Scholar]
  • 64. Carignan A, Poulin S, Martin P, et al. Efficacy of secondary prophylaxis with vancomycin for preventing recurrent clostridium difficile infections. Am J Gastroenterol 2016; 111: 1834–1840. [DOI] [PubMed] [Google Scholar]
  • 65. Rokas KE, Johnson JW, Beardsley JR, et al. The addition of intravenous metronidazole to oral vancomycin is associated with improved mortality in critically ill patients with clostridium difficile infection. Clin Infect Dis 2015; 61: 934–941. [DOI] [PubMed] [Google Scholar]
  • 66. Malamood M, Nellis E, Ehrlich AC, et al. Vancomycin enemas as adjunctive therapy for clostridium difficile infection. J Clin Med Res 2015; 7: 422–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Kim PK, Huh HC, Cohen HW, et al. Intracolonic vancomycin for severe clostridium difficile colitis. Surg Infect 2013; 14: 532–539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Fischer M, Sipe B, Cheng YW, et al. Fecal microbiota transplant in severe and severe-complicated clostridium difficile: a promising treatment approach. Gut Microbes 2017; 8: 289–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Aroniadis OC, Brandt LJ, Greenberg A, et al. Long-term follow-up study of fecal microbiota transplantation for severe and/or complicated clostridium difficile infection: a multicenter experience. J Clin Gastroenterol 2016; 50: 398–402. [DOI] [PubMed] [Google Scholar]
  • 70. Bishop EJ, Tiruvoipati R, Metcalfe J, et al. The outcome of patients with severe and severe-complicated clostridium difficile infection treated with tigecycline combination therapy: a retrospective observational study. Intern Med J 2018; 48: 651–660. [DOI] [PubMed] [Google Scholar]
  • 71. Gergely Szabo B, Kadar B, Szidonia Lenart K, et al. Use of intravenous tigecycline in patients with severe clostridium difficile infection: a retrospective observational cohort study. Clin Microbiol Infect 2016; 22: 990–995. [DOI] [PubMed] [Google Scholar]
  • 72. Manea E, Sojo-Dorado J, Jipa RE, et al. The role of tigecycline in the management of clostridium difficile infection: a retrospective cohort study. Clin Microbiol Infect 2018; 24: 180–184. [DOI] [PubMed] [Google Scholar]
  • 73. LaSalvia MT, Branch-Elliman W, Snyder GM, et al. Does adjunctive tigecycline improve outcomes in severe-complicated, nonoperative clostridium difficile infection? Open Forum Infect Dis 2017; 4: ofw264-ofw. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Popovic N, Korac M, Nesic Z, et al. Oral teicoplanin versus oral vancomycin for the treatment of severe clostridium difficile infection: a prospective observational study. Eur J Clin Microbiol Infect Dis 2018; 37: 745–754. [DOI] [PubMed] [Google Scholar]
  • 75. McCreery G, Jones PM, Kidane B, et al. Polyethylene glycol intestinal lavage in addition to usual antibiotic treatment for severe clostridium difficile colitis: a randomised controlled pilot study. BMJ Open 2017; 7: e016803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. 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: 131–137. [DOI] [PubMed] [Google Scholar]
  • 77. McPherson S, Rees CJ, Ellis R, et al. Intravenous immunoglobulin for the treatment of severe, refractory, and recurrent clostridium difficile diarrhea. Dis Colon Rectum 2006; 49: 640–645. [DOI] [PubMed] [Google Scholar]
  • 78. Halabi WJ, Nguyen VQ, Carmichael JC, et al. 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: 802–812. [DOI] [PubMed] [Google Scholar]
  • 79. Markelov A, Livert D, Kohli H. Predictors of fatal outcome after colectomy for fulminant Clostridium difficile colitis: a 10-year experience. Am Surg 2011; 77: 977–980. [DOI] [PubMed] [Google Scholar]
  • 80. 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: 400–405. [DOI] [PubMed] [Google Scholar]
  • 81. Neal MD, Alverdy JC, Hall DE, et al. 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: 423–427, discussion 7–9. [DOI] [PubMed] [Google Scholar]
  • 82. Ferrada P, Callcut R, Zielinski MD, et al. Loop ileostomy versus total colectomy as surgical treatment for clostridium difficile-associated disease: an eastern association for the surgery of trauma multicenter trial. J Trauma Acute Care Surg 2017; 83: 36–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Hall BR, Leinicke JA, Armijo PR, et al. No survival advantage exists for patients undergoing loop ileostomy for clostridium difficile colitis. Am J Surg 2019; 217: 34–39. [DOI] [PubMed] [Google Scholar]
  • 84. Razik R, Rumman A, Bahreini Z, et al. Recurrence of clostridium difficile infection in patients with inflammatory bowel disease: the RECIDIVISM study. Am J Gastroenterol 2016; 111: 1141–1146. [DOI] [PubMed] [Google Scholar]
  • 85. Khanna S, Shin A, Kelly CP. Management of clostridium difficile infection in inflammatory bowel disease: expert review from the clinical practice updates committee of the AGA Institute. Clin Gastroenterol Hepatol 2017; 15: 166–174. [DOI] [PubMed] [Google Scholar]
  • 86. Rodemann JF, Dubberke ER, Reske KA, et al. Incidence of clostridium difficile infection in inflammatory bowel disease. Clin Gastroenterol Hepatol 2007; 5: 339–344. [DOI] [PubMed] [Google Scholar]
  • 87. Bossuyt P, Verhaegen J, Van Assche G, et al. Increasing incidence of clostridium difficile-associated diarrhea in inflammatory bowel disease. J Crohns Colitis 2009; 3: 4–7. [DOI] [PubMed] [Google Scholar]
  • 88. Ananthakrishnan AN, Saeian K, McGinley EL, et al. Temporal trends in disease outcomes related to clostridium difficile infection in patients with inflammatory bowel disease. Inflamm Bowel Dis 2010; 17: 976–983. [DOI] [PubMed] [Google Scholar]
  • 89. Law CC, Tariq R, Khanna S, et al. Systematic review with meta-analysis: the impact of clostridium difficile infection on the short- and long-term risks of colectomy in inflammatory bowel disease. Aliment Pharmacol Ther 2017; 45: 1011–1020. [DOI] [PubMed] [Google Scholar]
  • 90. 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: 5054–5059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Högenauer C, Karas A, Reinisch W, et al. Pharmacokinetics and safety of fidaxomicin in patients with inflammatory bowel disease and clostridium difficile infection: an open-label phase IIIb/IV study (PROFILE). J Antimicrob Chemother 2018; 73: 3430–3441. [DOI] [PubMed] [Google Scholar]
  • 92. Vehreschild MJGT, S Taori, Goldenberg SD, et al. Fidaxomicin for the treatment of clostridium difficile infection (CDI) in at-risk patients with inflammatory bowel disease, fulminant CDI, renal impairment or hepatic impairment: a retrospective study of routine clinical use (ANEMONE). Eur J Clin Microbiol Infect Dis 2018; 37: 2097–2106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Khoruts A, Rank KM, Newman KM, et al. Inflammatory bowel disease affects the outcome of fecal microbiota transplantation for recurrent clostridium difficile infection. Clin Gastroenterol Hepatol 2016; 14: 1433–1438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Newman KM, Rank KM, Vaughn BP, et al. Treatment of recurrent clostridium difficile infection using fecal microbiota transplantation in patients with inflammatory bowel disease. Gut Microbes 2017; 8: 303–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Fischer M, Kao D, Kelly C, et al. Fecal microbiota transplantation is safe and efficacious for recurrent or refractory clostridium difficile infection in patients with inflammatory bowel disease. Inflamm Bowel Dis 2016; 22: 2402–2409. [DOI] [PubMed] [Google Scholar]
  • 96. Hirten RP, Grinspan A, Fu SC, et al. Microbial engraftment and efficacy of fecal microbiota transplant for clostridium difficile in patients with and without inflammatory bowel disease. Inflamm Bowel Dis 2019, 10.1093/ibd/izy398. [DOI] [PMC free article] [PubMed]
  • 97. Qazi T, Amaratunga T, Barnes EL, et al. The risk of inflammatory bowel disease flares after fecal microbiota transplantation: systematic review and meta-analysis. Gut Microbes 2017; 8: 574–588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Moayyedi P, Surette MG, Kim PT, et al. Fecal microbiota transplantation induces remission in patients with active ulcerative colitis in a randomized controlled trial. Gastroenterology 2015; 149: 102–109.e6. [DOI] [PubMed] [Google Scholar]
  • 99. Paramsothy S, Kamm MA, Kaakoush NO, et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial. Lancet (London, England) 2017; 389: 1218–28. [DOI] [PubMed] [Google Scholar]
  • 100. Ananthakrishnan AN, Guzman-Perez R, Gainer V, et al. Predictors of severe outcomes associated with clostridium difficile infection in patients with inflammatory bowel disease. Alimentary Pharmacol Ther 2012; 35: 789–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Ben-Horin S, Margalit M, Bossuyt P, et al. Combination immunomodulator and antibiotic treatment in patients with inflammatory bowel disease and clostridium difficile infection. Clin Gastroenterol Hepatol 2009; 7: 981–987. [DOI] [PubMed] [Google Scholar]
  • 102. Louie T, Nord CE, Talbot GH, et al. Multicenter, double-blind, randomized, phase 2 study evaluating the novel antibiotic cadazolid in patients with clostridium difficile infection. Antimicrob Agents Chemother 2015; 59: 6266–6273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Vickers RJ, Tillotson GS, Nathan R, et al. Efficacy and safety of ridinilazole compared with vancomycin for the treatment of clostridium difficile infection: a phase 2, randomised, double-blind, active-controlled, non-inferiority study. Lancet Infect Dis 2017; 17: 735–744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Daley P, Louie T, Lutz JE, et al. Surotomycin versus vancomycin in adults with clostridium difficile infection: primary clinical outcomes from the second pivotal, randomized, double-blind, Phase 3 trial. J Antimicrob Chemother 2017; 72: 3462–3470. [DOI] [PubMed] [Google Scholar]
  • 105. Boix V, Fedorak RN, Mullane KM, et al. Primary outcomes from a phase 3, randomized, double-blind, active-controlled trial of surotomycin in subjects with clostridium difficile infection. Open Forum Infect Dis 2017; 4: ofw275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Shen NT, Maw A, Tmanova LL, et al. Timely use of probiotics in hospitalized adults prevents clostridium difficile infection: a systematic review with meta-regression analysis. Gastroenterology 2017; 152: 1889–1900.e9. [DOI] [PubMed] [Google Scholar]
  • 107. Goldenberg JZ, Yap C, Lytvyn L, et al. Probiotics for the prevention of clostridium difficile-associated diarrhea in adults and children. Cochrane Database Syst Rev 2017; 12: Cd006095. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Therapeutic Advances in Gastroenterology are provided here courtesy of SAGE Publications

RESOURCES