SER-109, an Investigational Microbiome Drug to Reduce Recurrence After Clostridioides difficile Infection: Lessons Learned From a Phase 2 Trial

Barbara H McGovern, Christopher B Ford, Matthew R Henn, Darrell S Pardi, Sahil Khanna, Elizabeth L Hohmann, Edward J O'Brien, Christopher A Desjardins, Patricia Bernardo, Jennifer R Wortman, Mary-Jane Lombardo, Kevin D Litcofsky, Jonathan A Winkler, Christopher W J McChalicher, Sunny S Li, Amelia D Tomlinson, Madhumitha Nandakumar, David N Cook, Roger J Pomerantz, John G Auninš, Michele Trucksis, Barbara H McGovern, Christopher B Ford, Matthew R Henn, Darrell S Pardi, Sahil Khanna, Elizabeth L Hohmann, Edward J O'Brien, Christopher A Desjardins, Patricia Bernardo, Jennifer R Wortman, Mary-Jane Lombardo, Kevin D Litcofsky, Jonathan A Winkler, Christopher W J McChalicher, Sunny S Li, Amelia D Tomlinson, Madhumitha Nandakumar, David N Cook, Roger J Pomerantz, John G Auninš, Michele Trucksis

Abstract

Background: Recurrent Clostridioides difficile infection (rCDI) is associated with loss of microbial diversity and microbe-derived secondary bile acids, which inhibit C. difficile germination and growth. SER-109, an investigational microbiome drug of donor-derived, purified spores, reduced recurrence in a dose-ranging, phase (P) 1 study in subjects with multiple rCDIs.

Methods: In a P2 double-blind trial, subjects with clinical resolution on standard-of-care antibiotics were stratified by age (< or ≥65 years) and randomized 2:1 to single-dose SER-109 or placebo. Subjects were diagnosed at study entry by PCR or toxin testing. Safety, C. difficile-positive diarrhea through week 8, SER-109 engraftment, and bile acid changes were assessed.

Results: 89 subjects enrolled (67% female; 80.9% diagnosed by PCR). rCDI rates were lower in the SER-109 arm than placebo (44.1% vs 53.3%) but did not meet statistical significance. In a preplanned analysis, rates were reduced among subjects ≥65 years (45.2% vs 80%, respectively; RR, 1.77; 95% CI, 1.11-2.81), while the <65 group showed no benefit. Early engraftment of SER-109 was associated with nonrecurrence (P < .05) and increased secondary bile acid concentrations (P < .0001). Whole-metagenomic sequencing from this study and the P1 study revealed previously unappreciated dose-dependent engraftment kinetics and confirmed an association between early engraftment and nonrecurrence. Engraftment kinetics suggest that P2 dosing was suboptimal. Adverse events were generally mild to moderate in severity.

Conclusions: Early SER-109 engraftment was associated with reduced CDI recurrence and favorable safety was observed. A higher dose of SER-109 and requirements for toxin testing were implemented in the current P3 trial.

Clinical trials registration: NCT02437487, https://ichgcp.net/clinical-trials-registry/NCT02437487?term=SER-109&draw= 2&rank=4.

Keywords: Clostridioides difficile infection; Clostridium difficile diagnostics; dysbiosis; fecal microbiota transplantation; microbiome.

© The Author(s) 2020. Published by Oxford University Press for the Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
CONSORT diagram. One subject randomized to placebo was dosed with SER-109; this subject was analyzed with the placebo intention-to-treat population in all efficacy analyses* and with the SER-109 safety population in all safety analyses* (see Supplementary Materials for further details). Abbreviation: CONSORT, Consolidated Standards of Reporting Trials.
Figure 2.
Figure 2.
Rates of recurrence of CDI within 8 weeks of study drug treatment in the ITT population of the phase 2 population. Among all study subjects, SER-109 was not associated with a statistically significant reduction in risk of recurrence. In an age-based subgroup analysis, SER-109 was associated with a significant reduction in recurrence among subjects aged ≥65 years (Mantel-Haenszel test: RR, 1.77; *95% CI, 1.1–2.8) but not in subjects Clostridioides difficile infection; CI, confidence interval; ns, nonsignificant; RR, relative risk.
Figure 3.
Figure 3.
Number of SER-109 species stratified by time point and treatment in the phase 2 study. In the phase 2 study, subjects receiving treatment had significantly more SER-109 species than those receiving placebo at weeks 1, 4, and 8 posttreatment (P < .001, all comparisons; Mann-Whitney U test). At pretreatment baseline, SER-109 diversity was not significantly different between subjects receiving placebo or SER-109 (Mann-Whitney U test). Boxplots display the median (horizontal line), 25th and 75th percentiles of distribution (box edges), range of nonoutlier observations (whiskers), and outlier observations (dots; >1.5 times the interquartile range). Sample sizes are shown below the x axis. ***P < .001. Abbreviations: BL, baseline; ns, nonsignificant; wk, week.
Figure 4.
Figure 4.
Number of SER-109 species stratified by outcome in the phase 2 study. Nonrecurrent subjects had significantly more SER-109 species than recurrent subjects within the treatment arm 1 week postdosing (wk1; P < .05, Mann-Whitney U test) but not at baseline (Mann-Whitney U test). SER-109 species diversity was not significantly different for subjects receiving placebo at either baseline or 1 week posttreatment (Mann-Whitney U test). Boxplots display the median (horizontal line), 25th and 75th percentiles of distribution (box edges), range of nonoutlier observations (whiskers), and outlier observations (dots; >1.5 times the interquartile range). Sample sizes are shown below the x axis, *P < .05. Abbreviations: BL, baseline; ns, nonsignificant; wk1, week 1.
Figure 5.
Figure 5.
Relationship of the engraftment of SER-109 species and concentration of secondary bile acids in the phase 2 study. In the phase 2 study, the number of SER-109 species, assessed in both placebo and SER-109 subjects at week 1, was significantly correlated with the concentration (ng/mg DW of lyophilized samples) of secondary bile acids LCA and DCA. Spearman correlation significance and ρ are shown for each comparison. In the boxplots shown, samples were binned by number of dose-species detected. The concentration of LCA and DCA is shown on the y axis. Boxplots display the median (horizontal line), 25th and 75th percentiles of distribution (box edges), range of nonoutlier observations (whiskers), and outlier observations (dots; >1.5 times the interquartile range). Abbreviations: DCA, deoxycholic acid; DW, dry weight; LCA, lithocholic acid.
Figure 6.
Figure 6.
Relationship of engraftment of SER-109 species to dose administered in the phase 1 and phase 2 studies. Subjects receiving the high dose, as defined by SporQ (see Methods), in the open-label phase 1 dose-ranging study (SERES- 001) had significantly more SER-109 species than subjects in the treatment arm of the phase 2 study (SERES-004), who received a fixed dose (wk1; P < .0001, Mann-Whitney U test). The number of SER-109 species was not significantly different between subjects receiving the low dose in the phase 1 trial and subjects receiving the fixed dose in the phase 2 trial (Mann-Whitney U test). The number of SER-109 species stratified by treatment and time point is shown; dots represent outliers. Sample sizes are shown below the x axis. ***P < .0001. Abbreviations: BL, baseline; ns, nonsignificant; wk1, week 1.

References

    1. 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; 313:1719–27.
    1. Lowy I, Molrine DC, Leav BA, et al. . Treatment with monoclonal antibodies against Clostridium difficile toxins. N Engl J Med 2010; 362:197–205.
    1. Seekatz AM, Theriot CM, Molloy CT, Wozniak KL, Bergin IL, Young VB. Fecal microbiota transplantation eliminates clostridium difficile in a murine model of relapsing disease. Infect Immun 2015; 83:3838–46.
    1. Chang JY, Antonopoulos DA, Kalra A, et al. . Decreased diversity of the fecal microbiome in recurrent Clostridium difficile-associated diarrhea. J Infect Dis 2008; 197:435–8.
    1. Eckburg PB, Bik EM, Bernstein CN, et al. . Diversity of the human intestinal microbial flora. Science 2005; 308:1635–8.
    1. Weingarden AR, Dosa PI, DeWinter E, et al. . Changes in colonic bile acid composition following fecal microbiota transplantation are sufficient to control clostridium difficile germination and growth. PLoS One 2016; 11:e0147210.
    1. Sorg JA, Sonenshein AL. Chenodeoxycholate is an inhibitor of Clostridium difficile spore germination. J Bacteriol 2009; 191:1115–7.
    1. Theriot CM, Young VB. Interactions between the gastrointestinal microbiome and Clostridium difficile. Annu Rev Microbiol 2015; 69:445–61.
    1. 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. Aliment Pharmacol Ther 2017; 45:899–908.
    1. Tariq R, Pardi DS, Bartlett MG, Khanna S. Low cure rates in controlled trials of fecal microbiota transplantation for recurrent Clostridium difficile infection: a systematic review and meta-analysis. Clin Infect Dis 2019; 68:1351–8.
    1. Bafeta A, Yavchitz A, Riveros C, Batista R, Ravaud P. Methods and reporting studies assessing fecal microbiota transplantation: a systematic review. Ann Intern Med 2017; 167:34–9.
    1. Dubberke ER, Lee CH, Orenstein R, Khanna S, Hecht G, Gerding DN. 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; 67:1198–204.
    1. Kelly CR, Khoruts A, Staley C, et al. . Effect of fecal microbiota transplantation on recurrence in multiply recurrent clostridium difficile infection: a randomized trial. Ann Intern Med 2016; 165:609–16.
    1. DeFilipp Z, Bloom PP, Torres Soto M, et al. . Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N Engl J Med 2019; 381:2043–50.
    1. Glover SC, Burstiner S, Jones D, Teymoorian A, et al. . E. coli sepsis following FMT in an IgA deficient IBD subject. Available at: . Accessed 9 March 2020.
    1. Holshue ML, DeBolt C, Lindquist S, et al. . First case of 2019 Novel coronavirus in the United States. N Engl J Med 2020; 382:929–36.
    1. Almomani SA, Button J, Schuster BM, Auniņš JA, Lombardo MJ, McKenzie GJ. Inactivation of vegetative bacteria during production of SER-109, a microbiome-based therapeutic for recurrent Clostridium difficile Infection. Poster 450. Presented at: American Society for Microbiology;Boston, MA; 16–20 June 2016.
    1. 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; 214:173–81.
    1. Carlson PE Jr. Regulatory considerations for fecal microbiota transplantation products. Cell Host Microbe 2020; 27:173–5.
    1. Mantel N, Haenszel W. Statistical aspects of the analysis of data from retrospective studies of disease. J Natl Cancer Inst 1959; 22:719–48.
    1. Robins J, Breslow N, Greenland S. Estimators of the Mantel-Haenszel variance consistent in both sparse data and large-strata limiting models. Biometrics 1986; 42:311–23.
    1. Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc 1958; 53:457–81.
    1. Segata N, Waldron L, Ballarini A, Narasimhan V, Jousson O, Huttenhower C. Metagenomic microbial community profiling using unique clade-specific marker genes. Nat Methods 2012; 9:811–4.
    1. Truong DT, Franzosa EA, Tickle TL, et al. . MetaPhlAn2 for enhanced metagenomic taxonomic profiling. Nat Methods 2015; 12:902–3.
    1. Mann HB, Whitney DR.. On a test of whether one of two random variables is stochastically larger than the other. Ann Math Statist 1947; 18:50–60.
    1. Spearman C. The proof and measurement of association between two things. Am J Psychology 1904; 15:72–101.
    1. 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; 1:1–16.
    1. Abujamel T, Cadnum JL, Jury LA, et al. . Defining the vulnerable period for re-establishment of Clostridium difficile colonization after treatment of C. difficile infection with oral vancomycin or metronidazole. PLoS One 2013; 8:e76269.
    1. Kelly CP. Can we identify patients at high risk of recurrent Clostridium difficile infection? Clin Microbiol Infect 2012; 18(Suppl 6):21–7.
    1. 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:e1–48.
    1. Planche TD, Davies KA, Coen PG, et al. . Differences in outcome according to Clostridium difficile testing method: a prospective multicentre diagnostic validation study of C difficile infection. Lancet Infect Dis 2013; 13:936–45.
    1. Gonzalez-Orta M, Saldana C, Cadnum J, Donskey CJ. Are patients with prior Clostridium difficile infection (CDI) a potential source of transmission during hospitalization ? Presented at IDWeek in San Diego, CA, 4–8 October 2017. Poster 494.
    1. Jackson M, Olefson S, Machan JT, Kelly CR. A high rate of alternative diagnoses in patients referred for presumed clostridium difficile infection. J Clin Gastroenterol 2016; 50:742–6.
    1. 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; 44:576–82.
    1. Beaulieu C, Dionne LL, Julien AS, Longtin Y. Clinical characteristics and outcome of patients with Clostridium difficile infection diagnosed by PCR versus a three-step algorithm. Clin Microbiol Infect 2014; 20:1067–73.
    1. Polage CR, Gyorke CE, Kennedy MA, et al. . Overdiagnosis of Clostridium difficile infection in the molecular test era. JAMA Intern Med 2015; 175:1792–801.
    1. Kong LY, Davies K, Wilcox MH. The perils of PCR-based diagnosis of Clostridioides difficile infections: painful lessons from clinical trials. Anaerobe 2019; 60:102048.
    1. Eastwood K, Else P, Charlett A, Wilcox M. Comparison of nine commercially available Clostridium difficile toxin detection assays, a real-time PCR assay for C. difficile tcdB, and a glutamate dehydrogenase detection assay to cytotoxin testing and cytotoxigenic culture methods. J Clin Microbiol 2009; 47:3211–7.
    1. Lessa FC, Mu Y, Bamberg WM, et al. . Burden of Clostridium difficile infection in the United States. N Engl J Med 2015; 372:825–34.
    1. Ma GK, Brensinger CM, Wu Q, Lewis JD. Increasing incidence of multiply recurrent Clostridium difficile infection in the United States: a cohort study. Ann Intern Med 2017; 167:152–8.
    1. Canavan C, West J, Card T. The epidemiology of irritable bowel syndrome. Clin Epidemiol 2014; 6:71–80.
    1. Hohmann EL. Are microbial politics local? Ann Intern Med 2016; 165:667–8.
    1. Ford C, Henn M, Bryant J, et al. . Treatment of recurrent Clostridium difficile infection with SER-109 reduces gastrointestinal carriage of antimicrobial resistance genes. Available at: . Accessed 9 March 2020.

Source: PubMed

3
Sottoscrivi