Bezlotoxumab Is Associated With a Reduction in Cumulative Inpatient-Days: Analysis of the Hospitalization Data From the MODIFY I and II Clinical Trials

Anirban Basu, Vimalanand S Prabhu, Mary Beth Dorr, Yoav Golan, Erik R Dubberke, Oliver A Cornely, Sebastian M Heimann, Alison Pedley, Ruifeng Xu, Mary E Hanson, Stephen Marcella, Anirban Basu, Vimalanand S Prabhu, Mary Beth Dorr, Yoav Golan, Erik R Dubberke, Oliver A Cornely, Sebastian M Heimann, Alison Pedley, Ruifeng Xu, Mary E Hanson, Stephen Marcella

Abstract

Background: Patients with recurrent Clostridium difficile infection (rCDI) are more likely to have a hospital readmission and spend increased time in inpatient settings compared with patients with primary CDI. MODIFY I and II demonstrated that bezlotoxumab significantly reduced rCDI vs placebo. A post hoc within-trial analysis assessed whether bezlotoxumab was associated with a reduction in cumulative inpatient-days.

Methods: Data were pooled from the MODIFY trials to estimate the cumulative hospitalized days summed over the 84-day follow-up period. We adjusted inpatient use data from pooled MODIFY I and II for survival and censoring to estimate 84-day cumulative inpatient-days, overall and for subgroups. Treatment effects were obtained using recycled predictions based on trial protocol and rCDI risk, and 95% confidence intervals were obtained using 1000 bootstrap replicates.

Results: Mean cumulative inpatient-days were greater in the placebo arm (14.1 days) vs the bezlotoxumab arm (12.1 days) in the overall population. The mean difference between treatment groups was 2.1 days (95% confidence interval, -0.4 to -3.7). This was consistent in participants with risk factors for rCDI: age ≥65 years, compromised immunity, severe CDI, prior CDI, and ribotype 027/078/244 infection. As the number of risk factors increased, bezlotoxumab resulted in greater reductions in the number of inpatient-days compared with placebo (difference: -1.2 days, -2.3 days, -2.5 days, and -3.0 days for 0, 1, 2, and ≥3 risk factors, respectively).

Conclusions: Bezlotoxumab was associated with a reduction in cumulative inpatient-days, suggesting that treatment with bezlotoxumab may substantially reduce rCDI-associated health care resource use. Trial registrations. MODIFY I (MK-3415A-001, NCT01241552) and II (MK-3415A-002, NCT01513239).

Keywords: CDI burden of disease; Clostridium difficile infection; recurrence; rehospitalization.

Figures

Figure 1.
Figure 1.
Estimated mean cumulative inpatient-days summed over 84 days in the modified intent-to-treat population. aInpatients at the time of study randomization. bHistory of Clostridium difficile infection recurrence within the previous 6 months. cDefined on the basis of a subject’s medical history or use of immunosuppressive therapy. dZar 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 white blood cell count >15 000 cells/mm3 within 48 hours (1 point); (5) endoscopic evidence of pseudomembranous colitis (2 points); and (6) treatment in an intensive care unit (2 points). Abbreviations: CDI, Clostridium difficile infection; CI, confidence interval.

References

    1. Johnson S, Louie TJ, Gerding DN, et al. ; Polymer Alternative for CDI Treatment (PACT) investigators Vancomycin, metronidazole, or tolevamer for Clostridium difficile infection: results from two multinational, randomized, controlled trials. Clin Infect Dis 2014; 59:345–54.
    1. Louie TJ, Miller MA, Mullane KM, et al. ; OPT-80-003 Clinical Study Group Fidaxomicin versus vancomycin for Clostridium difficile infection. N Engl J Med 2011; 364:422–31.
    1. Sheitoyan-Pesant C, Abou Chakra CN, Pépin J, et al. . Clinical and healthcare burden of multiple recurrences of Clostridium difficile infection. Clin Infect Dis 2016; 62:574–80.
    1. Dubberke ER, Schaefer E, Reske KA, et al. . Attributable inpatient costs of recurrent Clostridium difficile infections. Infect Control Hosp Epidemiol 2014; 35:1400–7.
    1. Bauer MP, Notermans DW, van Benthem BH, et al. ; ECDIS Study Group Clostridium difficile infection in Europe: a hospital-based survey. Lancet 2011; 377:63–73.
    1. Morrison RH, Hall NS, Said M, et al. . Risk factors associated with complications and mortality in patients with Clostridium difficile infection. Clin Infect Dis 2011; 53:1173–8.
    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. Zar FA, Bakkanagari SR, Moorthi KM, Davis MB. A comparison of vancomycin and metronidazole for the treatment of Clostridium difficile-associated diarrhea, stratified by disease severity. Clin Infect Dis 2007; 45:302–7.
    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. Abou Chakra CN, Pepin J, Sirard S, Valiquette L. Risk factors for recurrence, complications and mortality in Clostridium difficile infection: a systematic review. PLoS One 2014; 9:e98400.
    1. Inns T, Gorton R, Berrington A, et al. . Effect of ribotype on all-cause mortality following Clostridium difficile infection. J Hosp Infect 2013; 84:235–41.
    1. See I, Mu Y, Cohen J, et al. . NAP1 strain type predicts outcomes from Clostridium difficile infection. Clin Infect Dis 2014; 58:1394–400.
    1. Olsen MA, Yan Y, Reske KA, et al. . Impact of Clostridium difficile recurrence on hospital readmissions. Am J Infect Control 2015; 43:318–22.
    1. Elixhauser A, Steiner C, Gould C. Readmissions following hospitalizations with Clostridium difficile infections, 2009. Statistical Brief #145. 2006. Available at: . Accessed August 14, 2018.
    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; 43:707–14.
    1. Wilcox MH, Gerding DN, Poxton IR, et al. ; MODIFY I and MODIFY II Investigators Bezlotoxumab for prevention of recurrent Clostridium difficile infection. N Engl J Med 2017; 376:305–17.
    1. Gerding DN, Kelly CP, Rahav G, et al. . Bezlotoxumab for prevention of recurrent C. difficile infection in patients at increased risk for recurrence. Clin Infect Dis 2018; 67:649–56.
    1. Lin DY. Linear regression analysis of censored medical costs. Biostatistics 2000; 1:35–47.
    1. Shah DN, Aitken SL, Barragan LF, et al. . Economic burden of primary compared with recurrent Clostridium difficile infection in hospitalized patients: a prospective cohort study. J Hosp Infect 2016; 93:286–9.
    1. Zhang D, Prabhu VS, Marcella SW. Attributable healthcare resource utilization and costs for patients with primary and recurrent Clostridium difficile infection in the United States. Clin Infect Dis 2018; 66:1326–32.
    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; 65:1218–21.
    1. Prabhu VS, Dubberke ER, Dorr MB, et al. . Cost-effectiveness of bezlotoxumab compared with placebo for the prevention of recurrent Clostridium difficile Infection. Clin Infect Dis 2018; 66:355–62.
    1. Bump S. Evaluation of the 2012 Health Care Cost Containment Law in Massachusetts. 2017. Available at: . Accessed August 14, 2018.
    1. Murphy CR, Avery TR, Dubberke ER, Huang SS. Frequent hospital readmissions for Clostridium difficile infection and the impact on estimates of hospital-associated C. difficile burden. Infect Control Hosp Epidemiol 2012; 33:20–8.

Source: PubMed

3
Tilaa