Analysis of C. difficile infection-related outcomes in European participants in the bezlotoxumab MODIFY I and II trials

Emilio Bouza, Oliver A Cornely, Antonio Ramos-Martinez, Robert Plesniak, Misoo C Ellison, Mary E Hanson, Mary Beth Dorr, Emilio Bouza, Oliver A Cornely, Antonio Ramos-Martinez, Robert Plesniak, Misoo C Ellison, Mary E Hanson, Mary Beth Dorr

Abstract

The MODIFY I/II trials demonstrated that bezlotoxumab, a human monoclonal antibody against Clostridioides difficile toxin B, given during antibiotic treatment for Clostridioides difficile infection (CDI) significantly reduced C. difficile recurrence (rCDI) in adults at high risk for rCDI. Efficacy of CDI-directed intervention may depend on ribotype regional epidemiology, and patient characteristics. This post hoc analysis assessed the efficacy of bezlotoxumab in the subgroup of MODIFY I/II trial participants enrolled in Europe. Data from the bezlotoxumab (10 mg/kg single intravenous infusion) and placebo (0.9% saline) groups from MODIFY I/II were compared to assess initial clinical cure (ICC), rCDI, all-cause, and CDI-associated rehospitalizations within 30 days of discharge, and mortality through 12 weeks post-infusion. Of 1554 worldwide participants, 606 were from Europe (bezlotoxumab n = 313, 51%; placebo n = 292; 48%). Baseline characteristics were generally similar across groups, although there were more immunocompromised participants in the bezlotoxumab group (27.2%) compared with placebo (20.1%). Fifty-five percent of participants were female, and 86% were hospitalized at randomization. The rate of ICC was similar between treatment groups. The rate of rCDI in the bezlotoxumab group was lower compared with placebo among European participants overall, and among those with ≥ 1 risk factor for rCDI. Bezlotoxumab reduced 30-day CDI-associated rehospitalizations compared with placebo. These results are consistent with overall results from the MODIFY trials and demonstrate that bezlotoxumab reduces rCDI and CDI-associated rehospitalizations in European patients with CDI. MODIFY I/II (NCT01241552 and NCT01513239).

Keywords: Bezlotoxumab; CDI recurrence; Clostridioides difficile infection; Rehospitalization.

Conflict of interest statement

EB has participated in clinical trials and advisory boards sponsored by MSD, Pfizer, and Astellas. ARM has participated in clinical trials sponsored by MSD. RP has participated in clinical trials sponsored by MSD. OAC has received research grants from, is an advisor to, or received lecture honoraria from Actelion, Allecra Therapeutics, Amplyx, Arsanis, Astellas, AstraZeneca, Basilea, Bayer, Cidara, Da Volterra, F2G, Gilead, GSK, IQVIA, Janssen, Leeds University, Matinas, The Medicines Company, Medpace, Melinta, Menarini, MSD, Miltenyi, Paratek, Pfizer, PSI, Rempex, Roche, Sanofi Pasteur, Scynexis, Seres, Summit, Tetraphase, and Vical. MCE, MEH, and MBD are employees of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA, and may own stock and/or restricted stock units in Merck & Co., Inc., Kenilworth, NJ, USA.

Figures

Fig. 1
Fig. 1
Number and proportion of participants enrolled in the European region shown by country in MODIFY I and MODIFY II (mITT population; numbers represent n’s)
Fig. 2
Fig. 2
Proportion of participants with ICC and SCC (mITT population) and rCDI (clinical cure population). Numbers above bars indicate difference and 95% confidence interval. All, all European participants; ≥ 1 risk factor, European participants with at least 1 risk factor for CDI recurrence (≥ 65 years of age, ≥ 1 CDI episodes in past 6 months, severe CDI (Zar score ≥ 2), immunocompromised, ribotypes 027, 078, or 244 strain; BEZ, bezlotoxumab; CI, confidence interval; ICC, initial clinical cure; mITT, modified intent-to-treat; PBO, placebo; rCDI, recurrent Clostridium difficile infection; SCC, sustained clinical cure
Fig. 3
Fig. 3
C. difficile infection recurrence rates by risk factor subgroup in European participants (clinical cure population). Unless otherwise specified, each subgroup includes all patients with the risk factor(s) (i.e., those with only the specified risk factor[s] and those with the specified risk factor[s] and ≥ 1 additional risk factor). CDI Hx, Clostridium difficile infection history in the previous 6 months; CI, confidence interval. aBased on Miettinen and Nurminen method without stratification. bZar score ≥ 2 based on the following: (1) age > 60 years (1 point); (2) body temperature > 38.3 °C (> 100 °F) (1 point); (3) albumin level ˂ 2.5 mg/dL (1 point); (4) peripheral WBC count > 15,000 cells/mm3 within 48 h (1 point); (5) endoscopic evidence of pseudomembranous colitis (2 points); and (6) treatment in an intensive care unit (2 points). cDefined on the basis of a subject’s medical history or use of immunosuppressive therapy. dDenominator is subjects in the mITT population with a positive culture
Fig. 4
Fig. 4
Kaplan-Meier plot of time to CDI recurrence

References

    1. Suetens C, Hopkins S, Kolman J, et al. Point prevalence survey of healthcare-associated infections and antimicrobial use in European acute care hospitals. Stockholm: ECDC; 2013. pp. 1–216.
    1. Asensio A, Di Bella S, Lo Vecchio A, et al. The impact of Clostridium difficile infection on resource use and costs in hospitals in Spain and Italy: a matched cohort study. Int J Infect Dis. 2015;36:31–38. doi: 10.1016/j.ijid.2015.05.013.
    1. Tresman R, Goldenberg SD. Healthcare resource use and attributable cost of Clostridium difficile infection: a micro-costing analysis comparing first and recurrent episodes. J Antimicrob Chemother. 2018;73:2851–2855. doi: 10.1093/jac/dky250.
    1. Leblanc S, Blein C, Andremont A, et al. Burden of Clostridium difficile infections in French hospitals in 2014 from the national health insurance perspective. Infect Control Hosp Epidemiol. 2017;38:906–911. doi: 10.1017/ice.2017.114.
    1. Heimann SM, Vehreschild JJ, Cornely OA, et al. Economic burden of Clostridium difficile associated diarrhoea: a cost-of-illness study from a German tertiary care hospital. Infection. 2015;43:707–714. doi: 10.1007/s15010-015-0810-x.
    1. Le Monnier A, Duburcq A, Zahar JR, et al. Hospital cost of Clostridium difficile infection including the contribution of recurrences in French acute-care hospitals. J Hosp Infect. 2015;91:117–122. doi: 10.1016/j.jhin.2015.06.017.
    1. Poli A, Di Matteo S, Bruno GM, et al. Economic burden of Clostridium difficile in five hospitals of the Florence health care system in Italy. Risk Manag Healthc Policy. 2015;8:207–213.
    1. Cornely OA, Miller MA, Louie TJ, et al. Treatment of first recurrence of Clostridium difficile infection: fidaxomicin versus vancomycin. Clin Infect Dis. 2012;55(Suppl 2):S154–SS61. doi: 10.1093/cid/cis462.
    1. 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: 10.1093/cid/ciu313.
    1. Louie TJ, Miller MA, Mullane KM, et al. Fidaxomicin versus vancomycin for Clostridium difficile infection. N Engl J Med. 2011;364:422–431. doi: 10.1056/NEJMoa0910812.
    1. Sheitoyan-Pesant C, Abou Chakra CN, Pepin J, et al. Clinical and healthcare burden of multiple recurrences of Clostridium difficile infection. Clin Infect Dis. 2016;62:574–580. doi: 10.1093/cid/civ958.
    1. Surawicz CM. Treatment of recurrent Clostridium difficile-associated disease. Nat Clin Pract Gastroenterol Hepatol. 2004;1:32–38. doi: 10.1038/ncpgasthep0018.
    1. Gerding DN, Kelly CP, Rahav G, et al. Bezlotoxumab for prevention of recurrent Clostridium difficile infection in patients at increased risk for recurrence. Clin Infect Dis. 2018;67:649–656. doi: 10.1093/cid/ciy171.
    1. 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: 10.1056/NEJMoa1602615.
    1. (2019) 2017 annual report for the emerging infections program for Clostridioides difficile infection. Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases.
    1. Freeman J, Vernon J, Pilling S, et al. The ClosER study: results from a three-year pan-European longitudinal surveillance of antibiotic resistance among prevalent Clostridium difficile ribotypes, 2011-2014. Clin Microbiol Infect. 2018;24:724–731. doi: 10.1016/j.cmi.2017.10.008.
    1. Chitnis AS, Holzbauer SM, Belflower RM, et al. Epidemiology of community-associated Clostridium difficile infection, 2009 through. JAMA Intern Med. 2011;2013:1–9.
    1. Davies KA, Ashwin H, Longshaw CM et al (2016) Diversity of Clostridium difficile PCR ribotypes in Europe: results from the European, multicentre, prospective, biannual, point-prevalence study of Clostridium difficile infection in hospitalised patients with diarrhoea (EUCLID), 2012 and 2013. Euro Surveill 21
    1. Lewis SJ, Heaton KW. Stool form scale as a useful guide to intestinal transit time. Scand J Gastroenterol. 1997;32:920–924. doi: 10.3109/00365529709011203.
    1. Miettinen O, Nurminen M. Comparative analysis of two rates. Stat Med. 1985;4:213–226. doi: 10.1002/sim.4780040211.
    1. Sundram F, Guyot A, Carboo I, et al. Clostridium difficile ribotypes 027 and 106: clinical outcomes and risk factors. J Hosp Infect. 2009;72:111–118. doi: 10.1016/j.jhin.2009.02.020.
    1. Loo VG, Poirier L, Miller MA, et al. A predominantly clonal multi-institutional outbreak of Clostridium difficile-associated diarrhea with high morbidity and mortality. N Engl J Med. 2005;353:2442–2449. doi: 10.1056/NEJMoa051639.
    1. McDonald LC, Killgore GE, Thompson A, et al. An epidemic, toxin gene-variant strain of Clostridium difficile. N Engl J Med. 2005;353:2433–2441. doi: 10.1056/NEJMoa051590.
    1. Stubbs SL, Brazier JS, O’Neill GL, et al. PCR targeted to the 16S-23S rRNA gene intergenic spacer region of Clostridium difficile and construction of a library consisting of 116 different PCR ribotypes. J Clin Microbiol. 1999;37:461–463. doi: 10.1128/JCM.37.2.461-463.1999.
    1. Cheknis A, Johnson S, Chesnel L, et al. Molecular epidemiology of Clostridioides (Clostridium) difficile strains recovered from clinical trials in the US, Canada and Europe from 2006-2009 to 2012-2015. Anaerobe. 2018;53:38–42. doi: 10.1016/j.anaerobe.2018.05.009.
    1. Wilcox MH, Ahir H, Coia JE, et al. Impact of recurrent Clostridium difficile infection: hospitalization and patient quality of life. J Antimicrob Chemother. 2017;72:2647–2656. doi: 10.1093/jac/dkx174.
    1. Prabhu VS, Cornely OA, Golan Y, et al. Thirty-Day readmissions in hospitalized patients who received bezlotoxumab with antibacterial drug treatment for Clostridium difficile infection. Clin Infect Dis. 2017;65:1218–1221. doi: 10.1093/cid/cix523.

Source: PubMed

3
구독하다