2,233
Views
11
CrossRef citations to date
0
Altmetric
Commentaries and Views

Advancing therapeutics for recurrent clostridioides difficile infections: an overview of vowst’s FDA approval and implications

ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Article: 2232137 | Received 24 May 2023, Accepted 28 Jun 2023, Published online: 11 Jul 2023

ABSTRACT

Clostridioides difficile infections (CDI) are a leading cause of healthcare-associated infections with a high relapse rate. Current treatment guidelines recommend fidaxomicin as the primary therapy for initial CDI episodes and suggest alternative approaches for recurrent episodes, including fecal microbiota transplantation (FMT). This paper explores the recent approval of Vowst, a novel oral FMT drug, by the United States Food and Drug Administration (FDA) as a prophylactic therapy to prevent recurrent CDIs. Vowst comprises a formulation of live fecal microbiota spores and works by reestablishing the disrupted gut microbiota, limiting C. difficile spore germination, and promoting microbiome repair. Furthermore, this paper will discuss the product’s approval journey and the uncertainties regarding its efficacy in CDI patients beyond the ones who participated in the clinical trials, pharmacovigilance, cost estimates, and the need for a more stringent donor screening process. Overall, Vowst’s approval marks a significant step forward in the prevention of recurrent CDI infections with various beneficial implications for future gastroenterology.

Introduction

As a recognized public health threat, Clostridioides difficile infections (CDIs) are one of the most challenging to treat and are a leading cause of healthcare-associated infections (HAIs). The overall rate of healthcare-associated CDIs worldwide, estimated at 2.24 infections per 1,000 admissions annually, is known to be substantially higher in the intensive care units and internal medicine wards (11.08 and 10.80 infections per 1,000 admissions annually, respectively) when compared with community associated CDIs.Citation1 In addition, CDIs have a very high relapse rate of 20–30%, thereby prolonging hospital stays and contributing significantly to health-care costs ($5.4–$6.3 billion annually in the United States).Citation1

Transmission of CDIs occurs primarily horizontally via the fecal-oral route. As such there are numerous known sources of CDIs, including contact with infected roommates, shared equipment, the hands of medical personnel, and environmental surfaces.Citation2 The development of highly decontamination-resistant spores by C. difficile makes it particularly challenging to eradicate and may play arole in facilitating its nosocomial transmission.Citation2 Furthermore, in cases of gut dysbiosis, some individuals may acquire the infection endogenously due to the presence of the bacteria in the normal intestinal microflora (up to 5% of normal adults harbor the bacteria).Citation3

C. difficile outbreaks have been associated with the emergence of hypervirulent drug-resistant strains due to gut dysbiosis and worsening innate colonization resistance caused by irrational antibiotic prescribing and increased antibiotic resistance worldwide.Citation4,Citation5 Several antibiotics, including clindamycin, extended spectrum cephalosporins, and fluoroquinolones, have been associated with CDIs.Citation6 According to the 2021 Clinical Practice Guideline by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA), fidaxomicin (macrocyclic antibiotic) is recommended for the treatment of initial CDI episode in adults, with vancomycin (glycopeptide antibiotic) being an acceptable alternative.Citation7 In cases of recurrent CDI episodes, it is recommended to use either vancomycin (tapered and pulsed regimen), vancomycin followed by rifaximin, or fecal microbiota transplantation (FMT) in addition to fidaxomicin.Citation7

Despite its superior safety profile and lack of contraindications for use, the adoption of fidaxomicin has been slow due to its high cost and the lack of a readmission penalty.Citation8 Emerging evidence suggests that the efficacy of fidaxomicin may be reduced when used in combination therapy with CDI-directed or systemic antibiotics.Citation9 Furthermore, research and development of new antibiotics has been slow due to lengthy approval process with low success rates.Citation10 According to the World Health Organization (WHO), for existing classes of antibiotics, only 1 in 15 candidate antibiotics reached patients, a figure that declines to 1 in 30 for new classes of antibiotics.Citation10 Therefore, alternative secondary treatment modalities are being explored, particularly for treating fulminant, severe, recurrent, and antibiotic-resistant CDIs.

One such avenue is FMT, in which fecal solution from a healthy donor is administered into the recipient’s intestine to directly alter the local microflora.Citation11 It has been demonstrated that FMT after short-course vancomycin therapy is superior to fidaxomicin and standard-dose vancomycin monotherapies for the clinical and microbiological resolution of recurrent CDIs.Citation12 Additionally, FMT, by replacing the multi-drug resistance bacteria with healthy bacteria, has been shown to downregulate the expression of antibiotic resistance genes in CDI patients.Citation13 This article hence, aims to elaborate on the recent approval of Vowst, a novel oral FMT capsule, as a prophylactic therapeutic to prevent recurrent CDIs.

Vowst: route of FMT delivery

To date, majority of patients with recurrent CDIs are directed to gastrointestinal or infectious disease specialists, where the technique and route of FMT administration may depend on the specialty.Citation14 For example, in the US, FMT is commonly delivered via colonoscopy, whereas in Europe, administration through the nasogastric or nasoduodenal tube is preferred.Citation15,Citation16 However, recent analysis has shown a shifting trend in Europe with colonoscopy and rectal enema being performed more often than upper gastrointestinal tube insertion.Citation17 The efficacy of these routes of administration varies slightly, with uncontrolled studies showing a 90% efficacy in preventing recurrent CDIs upon administration via colonoscopy compared to an 80% efficacy in patients administered FMT via the upper gastrointestinal route.Citation18,Citation19

Compared to the more traditional approaches such as esophagogastroduodenoscopy, nasogastric tube, naso-jejunal tube, colonoscopy, or retention enema, newer oral FMTs like Vowst, are noninvasive, more comfortable for the patient, minimizes the risk of iatrogenic complications, and reduces the waiting time for these procedures, thereby, allowing better triage. Capsular formulations, therefore, combine an antibiotic’s convenience of delivery with the effectiveness of FMT for treating recurrent CDIs.Citation20 Vowst is delivered as a daily dose of four capsules each on an empty stomach for three consecutive days. The regimen must be initiated within the first 4 days of antibiotic discontinuation and requires the patients to take an initial dose of approximately 300 mL (10 ounces) of magnesium citrate, or 250 mL of polyethylene glycol electrolyte solution in case of renal impairment, for bowel cleansing and neutralization of the residual effects of antibiotics.Citation21

Vowst: mechanism of action

On April 26, 2023, the US Food and Drug Administration (FDA) approved the first oral microbiota-based product to prevent recurrent CDI in individuals 18 years and older who have previously had antimicrobial therapy for recurrent CDI.Citation22 The product is a formulation of live fecal microbiota spores from Seres Therapeutics, marketed as Vowst (formerly SER-109) and consists of a highly purified collection of about 50 species of Firmicutes spores that act by reestablishing the gut microbiota, limiting spore germination, bacterial replication, and toxin production by C. difficile.

Although its biological action of microbiome repair remains poorly understoodCitation23, Vowst is believed to influence the bile acid metabolism in the intestines by restoring the balance between primary and secondary bile acids. Primary bile acids, like cholic acid and taurocholic acid (TCA), facilitate C. difficile spore formation in the small intestines, while secondary bile acids, like deoxycholic acid (DCA) and lithocholic acid (LCA), inhibit its spore germination and vegetative growth.Citation24,Citation25 Although some degree of C. difficile spore formation occurs in healthy small intestines, the abundance of secondary bile acids in the large intestines prevents CDI infection in healthy individuals.Citation24,Citation25 Antibiotics-induced dysbiosis often results in imbalances in these bile acid pools, thereby increasing CDI risk.Citation25 Vowst corrects this imbalance by engrafting 7α-dehydroxylase activity possessing Firmicute species, which converts excess primary bile acids into secondary bile acids.Citation26 This mechanism was clinically observed as a moderate positive correlation at the end of week 1 between the number of SER-109 species and DCA (ϱ = .508, P < .001) and LCA concentrations (ϱ = .544, P < .001).Citation27

Furthermore, Vowst-mediated early microbiome repair was associated with a rapid and sustained pharmacodynamic response in patients, up to 24 weeks post-therapy, thereby, resulting in fewer CDI recurrences.Citation28 This was supported by a decline in proinflammatory Enterobacteriaceae species and an increase in Firmicute species in patients receiving Vowst.Citation26 Thus, the primary attribute of Vowst was identified as the restoration of colonization resistance in the gut and the metabolic competition between the microbiome therapy and C. difficile.

Vowst: clinical trials and approval process

The road to the approval of Vowst has been associated with many challenges. One of the main barriers was in 2016, when the Phase II ECOSPOR trial failed to meet its primary efficacy endpoint, with investigators finding that the relapse rate was similar between the experimental drugs and placebo after 8 weeks of therapy (44% vs. 53%). A subgroup analysis of subjects aged <65 years (43% on experimental drug vs. 27% on placebo) demonstrated even more unfavorable results.Citation29 Furthermore, in comparison with the Phase I study, the ECOSPOR-II trial demonstrated a marked increase in the rate of recurrences (13% vs 44%, respectively). The authors attributed these findings to the primary differences in the number of sites (40 vs 4), study design (open-label vs. randomized-controlled) and dose regimen (dose-ranging vs. fixed-dose).Citation27

Nevertheless, the more recent ECOSPOR-III and ECOSPOR-IV trials were able to re-establish the clinical efficacy of Vowst ().Citation26–28,Citation30,Citation31 These results were achieved by amending the dosing frequency from one dose in Phase II trial to one dose daily for three consecutive days in Phase III trial.Citation26 Furthermore, in comparison with Phase II trial, a three times higher daily dose (3 × 107 spore colony forming units) was chosen in Phase III.Citation26 These decisions were guided by the findings of dose-dependent engraftment kinetics in Phase II trial which further revealed that early and rapid engraftment during the first week was critical in preventing recurrent CDIs.Citation26,Citation27

Table 1. Overview of Phase 1 study and ECOSPOR clinical trials.

The positive results of the open-label ECOSPOR-IV clinical trial were the basis for the FDA approval of Vowst ().Citation31 An impressive clinical response was observed in 91.3% of patients receiving Vowst at week 8, regardless of the number of prior CDI infections, age, or type of antibiotic (vancomycin or fidaxomicin), and was maintained in 94.6% of participants through week 24 of treatment. Furthermore, it was demonstrated that Vowst reduced CDI recurrence, with 87.6% of Vowst participants remaining recurrence-free at the end of 8 weeks compared to 60.2% of participants in the placebo group. Vowst also demonstrated a superior safety profile, with relatively mild to moderate and transient adverse events.Citation26,Citation31

Table 2. Designation of Vowst FDA approval application.

Despite the milestone approval, uncertainties remain regarding the specific efficacy of Vowst in CDI patients beyond the ones who participated in the clinical trials, given the strict inclusion criteria adopted during the clinical trials. For example, female participants who were pregnant, breastfeeding, or lactating, oncological patients, patients with indications for other antibacterial therapy (urinary tract infections, surgical prophylaxis), and patients with a history of inflammatory bowel disease were all excluded.Citation27,Citation28 Besides, Vowst’s interaction with probiotics also remains to be elucidated since patients were advised to stop the use of probiotics during the trials. Moreover, its effectiveness in patients with previous FMT procedures also remains to be established.

In addition to these clinico-demographic characteristics, the efficacy of such clinical trials can also be strongly influenced by variations in experimental factors such as dosing regimens, patient characteristics, and/or primary endpoints. This was illustrated by a recent meta-analysis that evaluated the efficacy and safety of oral-fecal microbiota transplant capsules for recurrent CDI. Although the meta-analysis, which examined a mix of 12 case studies and 3 randomized control trials, reported an overall treatment efficacy of 0.821, significant variability in treatment efficacy was noted between studies due to the practicalities of treatment administration.Citation32

Finally, because the purification process removes several components from the feces that may be important to the success of FMT, questions remain regarding the efficacy of Vowst compared to complete FMT. As such, the generalized clinical use of Vowst will require obtaining further information on its efficacy, dosing strategies, inter-person differences, potential adverse effects, and contraindications. Its success will, in part, depend on the flexibility of stakeholders to modify current protocols; for example, the currently recommended four-capsule dosing schedule for three consecutive days may be subject to change over time.

Vowst: safety and pharmacovigilance

Several minimal side effects, such as diarrhea or abdominal discomfort, have been associated with Vowst (), most of which had occurred due to the very nature of FMT.Citation33 Since FMT capsules contain metabolically active bacteria, the use of such complex consortia as therapeutics raises questions about its off-target effects compared to standard pharmacological drugs. Particularly in the gut where the bacteria are exposed to the local microbiota, dietary by-products, other drug metabolites, and probiotics.Citation34 Therefore, further understanding of the complex dynamics associated with engraftment and invasion will be necessary to optimize adjunctive therapies to improve therapeutic outcomes.

As with any other FMT modalities, the adoption of good manufacturing purification steps is needed to minimize the risk of transfer of contaminants like infectious organisms (such as ethanol-inactivated viruses) and food allergens from the product source, i.e., processed fecal samples.Citation22,Citation26 Such potential safety concerns could be addressed via thorough donor screening and registration within an international biobank to ensure the safety of Vowst and other potential oral FMT-related therapeutics. Furthermore, international biobanking would make donor fecal material more readily available and enable the sharing of samples among different regions, thereby, minimizing logistical barriers related to the procurement of fresh fecal samples.Citation35,Citation36 If successful, these measures have the potential to spur personalized FMTs in the future.

For example, the OpenBiome Stool Bank (Massachusetts, US; https://openbiome.org/), an independent nonprofit organization, had delivered its 60,000th FMT product in 2021. The bank has been operational since 2013 and has recently launched a global initiative called Global Microbiome Conservancy to collect samples from populations outside the US and Western Europe, which collectively account for 70% of all biobanked samples. In fact, the Stool Bank estimates that more than 120 countries lack representative samples in public biobanks. Furthermore, in the absence of a clear resolution regarding the features of optimal donors and international consensus regarding the use of banked frozen donor material, the logical next step would be the formulation of relevant international guidelines for FMT screening before donation.Citation36,Citation37

Vowst: economical comparison

Naturally, as with the experiences of fidaxomicin, extensive economic analyses will also be needed to comprehend the benefits and effectiveness of FMT by oral capsule (). It should be, however, noted that most studies have shown that despite the higher costs of individual products (like fidaxomicin, FMTs), the overall health-care costs are significantly reduced due to reduced hospitalizations and shorter stays.Citation38,Citation39 A recent review of 12 studies has revealed that FMT (different routes of administration) is the most economical overall therapeutic modality for patients with recurrent CDIs.Citation40 Furthermore, in a simulated model study, the authors reported that the use of fidaxomicin for the first recurrence followed by FMT for subsequent recurrences is the most economical strategy.Citation41 However, it is pertinent to note that these comparisons were analyzed based on cheaper FMT products and not oral FMT products like Vowst. Future comparisons are needed to provide accurate results.

Table 3. Estimated list price for approved treatment and therapeutic products in recurrent CDIs.

Future prospects

The approval of Vowst as the first oral-fecal microbiota product is a significant step forward in the prevention of the recurrent CDIs, with various beneficial implications for future gastroenterology. Another FMT based biotherapeutic (Rebyota; Ferrings Pharmaceuticals Inc.) is already commercially available. Similar to Vowst, Rebyota is also intended for use as a preventative biotherapeutic in patients aged 18 or over following antibiotic treatment for recurrent CDI. However, unlike Vowst, it is administered rectally and requires a single administration. We believe that these approvals will further personalized therapies by analyzing the microbiome of individuals to determine which specific microbial species they need. Finally, FMT may become a more affordable treatment option in the long-term for patients as logistical aspects of the treatment become more affordable with future research.

We hope that the collective benefits of oral FMTs also reach developing countries and provide treatment availability for those with limited medical resources. Manufacturing and distribution of Vowst are anticipated to begin in June 2023 with Nestle Health Science partnering with Seres Therapeutics for the commercialization of Vowst in the US and Canada. However, besides these two countries, its further distribution globally remains unknown and would be clear over the course of time. In the first-year post-launch, the company anticipates Vowst to be paid under the New to Market Block (NTMB; out of pocket) until it becomes covered by Medicare and Medicaid. It would, hence, be essential to make the formulation economical and involve international collaborations between the developed and developing countries (North–South collaborations) for manufacturing, production, testing, and future research and development. This would ensure fair and equitable distribution of the collective benefits of oral FMTs. Even among the developed European countries, FMT remains niche with limited adoption, despite the publication of the European Consensus Statement on FMT in clinical practice.Citation42 It is estimated that only 10% of the patients with indication receive FMT in Europe.Citation17 Furthermore, the procedure was reportedly performed only at around 30 to 40 centers located across 17 European countries.Citation17

Finally, with other similar products in development and testing pipelines (RBX7455 by Rebiotix Inc., administered as a single enemaCitation43 and CP101 by Finch Research and Development LLC., administered orally as one-dose capsulesCitation44), it remains to be seen how the FDA and other regulators will revisit their views and guidelines. In fact, the expansion of FMT modalities and enforcement of stricter screening regulations have led to the OpenBiome stool bank announcing their plans for phasing out services as and when a FDA approved treatment becomes available.Citation45 According to the OpenBiome’s executive director, isolation of specific bacterial strains for future products should be the future direction of research and development after the widespread availability of fecal products.Citation46

In this regard, Vedanta Biosciences’ VE303 is pushing the boundaries of FMT further by eliminating the need for direct sourcing of fecal donor material. VE303, an oral bacterial consortium candidate, consists of eight types of nontoxic, nonpathogenic, commensal strains of Clostridia manufactured from clonal cell banks.Citation47,Citation48 Both Phase I and II trials of VE303 were able to meet the primary endpoints with promising outlook for future large-scale trials. Overall, whilst we remain optimistic on the future of FMT products for the treatment of recurrent, treatment resistant CDIs, it would be essential to address the issues surrounding ease-of-access, biomedical safety guidelines, and the cost-effectiveness of such modalities.

Author’s contribution

NJ conceptualized the present paper whilst NJ, TPU, and AFF were involved in data collection, data validation, and preparation of the initial draft of the manuscript. NJ, TPU, AFF, and VG were responsible for revising the manuscript. Supervision and project administration was done by VG. All authors have read and agreed to the final version of the paper for publication.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data availability statement

Not applicable. No new data was generated for this paper.

Additional information

Funding

The present paper did not receive any external funding and was self-supported by the authors.

References

  • Balsells E, Shi T, Leese C, Lyell I, Burrows J, Wiuff C, Campbell H, Kyaw MH, Nair H. Global burden of Clostridium difficile infections: a systematic review and meta-analysis. J Glob Health. 2019 Jun;9(1):010407. doi:10.7189/jogh.09.010407. PMID: 30603078; PMCID: PMC6304170.
  • Dilnessa T, Getaneh A, Hailu W, Moges F, Gelaw B, Chang Y-F. Prevalence and antimicrobial resistance pattern of Clostridium difficile among hospitalized diarrheal patients: a systematic review and meta-analysis. PLoS One. 2022 Jan 13;17(1):e0262597. doi:10.1371/journal.pone.0262597. PMID: 35025959; PMCID: PMC8758073.
  • Vaishnavi C. Clinical spectrum & pathogenesis of Clostridium difficile associated diseases. Indian J Med Res. 2010 Apr;131:487–10. PMID: 20424299.
  • Sarker MR, Paredes-Sabja D. Molecular basis of early stages of Clostridium difficile infection: germination and colonization. Future Microbiol. 2012 Aug;7(8):933–943. doi:10.2217/fmb.12.64. PMID: 22913353.
  • Britton RA, Young VB. Role of the intestinal microbiota in resistance to colonization by Clostridium difficile. Gastroenterology. 2014 May;146(6):1547–1553. doi:10.1053/j.gastro.2014.01.059. Epub 2014 Feb 4. PMID: 24503131; PMCID: PMC3995857.
  • Spigaglia P. Recent advances in the understanding of antibiotic resistance in Clostridium difficile infection. Ther Adv Infect Dis. 2016 Feb;3(1):23–42. doi:10.1177/2049936115622891. PMID: 26862400; PMCID: PMC4735502.
  • Johnson S, Lavergne V, Skinner AM, Gonzales-Luna AJ, Garey KW, Kelly CP, Wilcox MH. Clinical practice guideline by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA): 2021 focused update guidelines on management of clostridioides difficile infection in adults. Clin Infect Dis. 2021 Sep 7;73(5):755–757. doi:10.1093/cid/ciab718. PMID: 34492699.
  • Bainum TB, Reveles KR, Hall RG 2nd, Cornell K, Alvarez CA. Controversies in the prevention and treatment of clostridioides difficile infection in adults: a narrative review. Microorganisms. 2023 Feb 3;11(2):387. doi:10.3390/microorganisms11020387. PMID: 36838352; PMCID: PMC9963748.
  • Shah DN, Chan FS, Kachru N, Garcia KP, Balcer HE, Dyer AP, Emanuel JE, Jordan MD, Lusardi KT, Naymick G, et al. A multi-center study of fidaxomicin use for Clostridium difficile infection. Springerplus. 2016 Aug 2;5(1):1224. doi:10.1186/s40064-016-2825-x. PMID: 27536508; PMCID: PMC4970984.
  • World Health Organization. Lack of innovation set to undermine antibiotic performance and health gains. WHO. June 2022 [online]. [accessed 2023 June 24]. https://www.who.int/news/item/22-06-2022-22-06-2022-lack-of-innovation-set-to-undermine-antibiotic-performance-and-health-gains.
  • Gupta S, Allen-Vercoe E, Petrof EO. Fecal microbiota transplantation: in perspective. Therap Adv Gastroenterol. 2016 Mar;9(2):229–239. doi:10.1177/1756283X15607414. PMID: 26929784; PMCID: PMC4749851.
  • Hvas CL, Dahl Jørgensen SM, Jørgensen SP, Storgaard M, Lemming L, Hansen MM, Erikstrup C, Dahlerup JF. Fecal microbiota transplantation is superior to fidaxomicin for treatment of recurrent clostridium difficile infection. Gastroenterology. 2019 Apr;156(5):1324–1332.e3. doi:10.1053/j.gastro.2018.12.019. Epub 2019 Jan 2. PMID: 30610862.
  • Hyun J, Lee SK, Cheon JH, Yong DE, Koh H, Kang YK, Kim MH, Sohn Y, Cho Y, Baek YJ. et al. Faecal microbiota transplantation reduces amounts of antibiotic resistance genes in patients with multidrug-resistant organisms. Antimicrob Resist Infect Control. 2022 Jan 29;11(1):20. doi:10.1186/s13756-022-01064-4. PMID: 35093183; PMCID: PMC8800327.
  • Kao D, Roach B, Silva M, Beck P, Rioux K, Kaplan GG, Chang HJ, Coward S, Goodman KJ, Xu H, et al. Effect of oral capsule– vs colonoscopy-delivered fecal microbiota transplantation on recurrent clostridium difficile infection. JAMA. 2017 Nov 28;318(20):1985–1993. doi:10.1001/jama.2017.17077. PMID: 29183074; PMCID: PMC5820695.
  • Patel NC, Griesbach CL, DiBaise JK, Orenstein R. Fecal microbiota transplant for recurrent Clostridium difficile infection: mayo clinic in Arizona experience. Mayo Clin Proc. 2013 Aug;88(8):799–805. doi:10.1016/j.mayocp.2013.04.022. PMID: 23910407.
  • van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal EG, de Vos WM, Visser CE, van Nood E, de Vos WM, Kuijper EJ, et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med. 2013 Jan 31;368(5):407–415. doi:10.1056/NEJMoa1205037. Epub 2013 Jan 16. PMID: 23323867.
  • Baunwall SMD, Terveer EM, Dahlerup JF, Erikstrup C, Arkkila P, Vehreschild MJ, Ianiro G, Gasbarrini A, Sokol H, Kump PK, et al. The use of Faecal Microbiota Transplantation (FMT) in Europe: a Europe-wide survey. Lancet Reg Health Eur. 2021 Jul 19;9:100181. doi:10.1016/j.lanepe.2021.100181. PMID: 34693388; PMCID: PMC8513118.
  • Postigo R, Kim JH. Colonoscopic versus nasogastric fecal transplantation for the treatment of Clostridium difficile infection: a review and pooled analysis. Infection. 2012 Dec;40(6):643–648. doi:10.1007/s15010-012-0307-9. Epub 2012 Jul 31. PMID: 22847629.
  • Kelly CR, de Leon L, Jasutkar N. Fecal microbiota transplantation for relapsing Clostridium difficile infection in 26 patients: methodology and results. J Clin Gastroenterol. 2012 Feb;46(2):145–149. doi:10.1097/MCG.0b013e318234570b. PMID: 22157239.
  • Varga A, Kocsis B, Sipos D, Kása P, Vigvári S, Pál S, Dembrovszky F, Farkas K, Péterfi Z. How to apply FMT more effectively, conveniently and flexible - a comparison of FMT methods. Front Cell Infect Microbiol. 2021 Jun 4;11:657320. doi:10.3389/fcimb.2021.657320. PMID: 34150673; PMCID: PMC8213398.
  • Nagarakanti S, Orenstein R. Treating Clostridioides difficile: could microbiota-based live biotherapeutic products provide the answer? Infect Drug Resist. 2023 May 20;16:3137–3143. doi:10.2147/IDR.S400570. PMID: 37235073; PMCID: PMC10208241.
  • U.S. Food and Drug Administration. FDA approves first orally administered fecal microbiota product for the prevention of recurrence of clostridioides difficile infection [Internet]. 2023 [accessed 2023 May 15]. https://www.fda.gov/news-events/press-announcements/fda-approves-first-orally-administered-fecal-microbiota-product-prevention-recurrence-clostridioides.
  • U.S. Food and Drug Administration. VOWST (fecal microbiota spores, live-brpk) capsules, for oral administration [Internet]. 2023 [accessed 2023 May 14].: https://www.fda.gov/media/167579/download.
  • Łukawska A, Mulak A. Impact of primary and secondary bile acids on clostridioides difficile infection. Pol J Microbiol. 2022 Mar 14;71(1):11–18. doi:10.33073/pjm-2022-007. PMID: 35635171; PMCID: PMC9152914.
  • Theriot CM, Bowman AA, Young VB. Antibiotic-induced alterations of the gut microbiota alter secondary bile acid production and allow for clostridium difficile spore germination and outgrowth in the large intestine. mSphere. 2016 Jan 6;1(1):e00045–15. doi: 10.1128/mSphere.00045-15. PMID: 27239562; PMCID: PMC4863611.
  • Feuerstadt P, Louie TJ, Lashner B, Wang EEL, Diao L, Bryant JA, Sims M, Kraft CS, Cohen SH, Berenson CS, et al. SER-109, an oral microbiome therapy for recurrent clostridioides difficile infection. N Engl J Med. 2022 Jan 20;386(3):220–229. doi:10.1056/NEJMoa2106516. PMID: 35045228.
  • McGovern BH, Ford CB, Henn MR, Pardi DS, Khanna S, Hohmann EL, O’Brien EJ, Desjardins CA, Bernardo P, Wortman JR, et al. SER-109, an investigational microbiome drug to reduce recurrence after clostridioides difficile infection: lessons learned from a phase 2 trial. Clin Infect Dis. 2021 Jun 15;72(12):2132–2140. doi:10.1093/cid/ciaa387. PMID: 32255488; PMCID: PMC8204772.
  • Sims MD, Khanna S, Feuerstadt P, Louie TJ, Kelly CR, Huang ES, Hohmann EL, Wang EEL, Oneto C, Cohen SH, et al. Safety and Tolerability of SER-109 as an investigational microbiome therapeutic in adults with recurrent clostridioides difficile infection: a phase 3, open-label, single-arm trial. JAMA Netw Open. 2023 Feb 1;6(2):e2255758. doi:10.1001/jamanetworkopen.2022.55758. PMID: 36780159; PMCID: PMC9926325.
  • Seres Therapeutics. Seres Therapeutics announces interim results from SER-109 phase 2 ECOSPOR study in multiply recurrent Clostridium difficile infection [Internet]. 2016 [accessed 2023 May 14]. https://ir.serestherapeutics.com/news-releases/news-release-details/seres-therapeutics-announces-interim-results-ser-109-phase-2.
  • Khanna S, Pardi DS, Kelly CR, Kraft CS, Dhere T, Henn MR, Lombardo MJ, Vulic M, Ohsumi T, Winkler J, et al. A novel microbiome therapeutic increases gut microbial diversity and prevents recurrent clostridium difficile infection. J Infect Dis. 2016 Jul 15;214(2):173–181. doi:10.1093/infdis/jiv766. Epub 2016 Feb 8. PMID: 26908752.
  • Seres Therapeutics. ECOSPOR IV: an open-label study evaluating SER-109 in recurrent clostridioides difficile infection [Internet]. 2023 [accessed 2023 May 14]. https://clinicaltrials.gov/ct2/show/NCT03183141?term=ecospor+IV&draw=1&rank=1.
  • Du C, Luo Y, Walsh S, Grinspan A. Oral fecal microbiota transplant capsules are safe and effective for recurrent clostridioides difficile infection: a systematic review and meta-analysis. J Clin Gastroenterol. 2021 Apr 1;55(4):300–308. doi:10.1097/MCG.0000000000001495. PMID: 33471490.
  • Marcella C, Cui B, Kelly CR, Ianiro G, Cammarota G, Zhang F. Systematic review: the global incidence of faecal microbiota transplantation-related adverse events from 2000 to 2020. Aliment Pharmacol Ther. 2021 Jan;53(1):33–42. doi:10.1111/apt.16148. Epub 2020 Nov 7. PMID: 33159374.
  • Halaweish HF, Boatman S, Staley C. Encapsulated fecal microbiota transplantation: development, efficacy, and clinical application. Front Cell Infect Microbiol. 2022 Mar 17;12:826114. doi:10.3389/fcimb.2022.826114. PMID: 35372103; PMCID: PMC8968856.
  • Satokari R, Mattila E, Kainulainen V, Arkkila PE. Simple faecal preparation and efficacy of frozen inoculum in faecal microbiota transplantation for recurrent Clostridium difficile infection–an observational cohort study. Aliment Pharmacol Ther. 2015 Jan;41(1):46–53. doi:10.1111/apt.13009. Epub 2014 Oct 29. PMID: 25355279.
  • Cammarota G, Ianiro G, Kelly CR, Mullish BH, Allegretti JR, Kassam Z, Putignani L, Fischer M, Keller JJ, Costello SP, et al. International consensus conference on stool banking for faecal microbiota transplantation in clinical practice. Gut. 2019 Dec;68(12):2111–2121. doi:10.1136/gutjnl-2019-319548. Epub 2019 Sep 28. PMID: 31563878; PMCID: PMC6872442.
  • Wynn AB, Beyer G, Richards M, Procedure EL. Screening, and cost of fecal microbiota transplantation. Cureus. 2023 Feb 17;15(2):e35116. doi:10.7759/cureus.35116. PMID: 36938236; PMCID: PMC10023044.
  • Konijeti GG, Sauk J, Shrime MG, Gupta M, Ananthakrishnan AN. Cost-effectiveness of competing strategies for management of recurrent Clostridium difficile infection: a decision analysis. Clin Infect Dis. 2014 Jun;58(11):1507–1514. doi:10.1093/cid/ciu128. Epub 2014 Mar 31. PMID: 24692533; PMCID: PMC4017891.
  • Lapointe-Shaw L, Tran KL, Coyte PC, Hancock-Howard RL, Powis J, Poutanen SM, Hota S, Deshpande A. Cost-effectiveness analysis of six strategies to treat recurrent clostridium difficile infection. PLoS One. 2016 Feb 22;11(2):e0149521. doi:10.1371/journal.pone.0149521. PMID: 26901316; PMCID: PMC4769325.
  • Gupta A, Ananthakrishnan AN. Economic burden and cost-effectiveness of therapies for Clostridiodes difficile infection: a narrative review. Therap Adv Gastroenterol. 2021 May 30;14:17562848211018654. doi:10.1177/17562848211018654. PMID: 34104214; PMCID: PMC8170348.
  • Rajasingham R, Enns EA, Khoruts A, Vaughn BP. Cost-effectiveness of treatment regimens for clostridioides difficile infection: an evaluation of the 2018 infectious diseases society of America guidelines. Clin Infect Dis. 2020 Feb 14;70(5):754–762. doi:10.1093/cid/ciz318. PMID: 31001619; PMCID: PMC7319265.
  • Cammarota G, Ianiro G, Tilg H, Rajilić-Stojanović M, Kump P, Satokari R, Sokol H, Arkkila P, Pintus C, Hart A, et al. European consensus conference on faecal microbiota transplantation in clinical practice. Gut. 2017 Apr;66(4):569–580. doi:10.1136/gutjnl-2016-313017. Epub 2017 Jan 13. PMID: 28087657; PMCID: PMC5529972.
  • Khanna S, Pardi DS, Jones C, Shannon WD, Gonzalez C, Blount K. RBX7455, a non-frozen, orally administered investigational live biotherapeutic, is safe, effective, and shifts patients’ microbiomes in a phase 1 study for recurrent clostridioides difficile infections. Clin Infect Dis. 2021 Oct 5;73(7):e1613–e1620. doi:10.1093/cid/ciaa1430. PMID: 32966574; PMCID: PMC8492147.
  • Finch research and development LLC. Efficacy, safety, and tolerability study of oral full-spectrum microbiotaTM (CP101) in subjects with recurrent C. Diff (PRISM3). 2022 [online]. [accessed 25 June 2023]. https://classic.clinicaltrials.gov/ct2/show/study/NCT03110133.
  • OpenBiome. OpenBiome Announces New Direct Testing for SARS-CoV-2 in Fecal Microbiota Transplantation (FMT) preparations and release of new inventory. February 2021 [online]. [accessed 2023 June 25]. https://openbiome.org/feature/openbiome-announces-new-direct-testing-for-sars-cov-2-in-fecal-microbiota-transplantation-fmt-preparations-and-release-of-new-inventory/.
  • Williams S Fecal microbiota transplantation is poised for a makeover. The Scientist. June 2021 [online]. [accessed 25th June 2023]. https://www.the-scientist.com/bio-business/fecal-microbiota-transplantation-is-poised-for-a-makeover-68805.
  • Louie T, Golan Y, Khanna S, Bobilev D, Erpelding N, Fratazzi C, Carini M, Menon R, Ruisi M, Norman JM, et al. VE303, a defined bacterial consortium, for prevention of recurrent clostridioides difficile infection: a randomized clinical trial. JAMA. 2023 Apr 25;329(16):1356–1366. doi:10.1001/jama.2023.4314. PMID: 37060545; PMCID: PMC10105904.
  • Dsouza M, Menon R, Crossette E, Bhattarai SK, Schneider J, Kim YG, Reddy S, Caballero S, Felix C, Cornacchione L, et al. Colonization of the live biotherapeutic product VE303 and modulation of the microbiota and metabolites in healthy volunteers. Cell Host & Microbe. 2022 Apr 13;30(4):583–598.e8. doi:10.1016/j.chom.2022.03.016. PMID: 35421353.