Skip to main content
Erschienen in: European Journal of Clinical Microbiology & Infectious Diseases 10/2020

Open Access 06.06.2020 | Original Article

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

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

Erschienen in: European Journal of Clinical Microbiology & Infectious Diseases | Ausgabe 10/2020

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)
Hinweise

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Clostridioides difficile infection (CDI) is one of the most frequently reported hospital-acquired infections (HAIs) in European countries, accounting for 3.6% of all HAIs and 48% of all gastro-intestinal infections, both HA and non-HA (excluding hepatitis) [1]. With such high incidence rates, healthcare resource use and attributable financial burden of CDI are significant, mainly driven by length of hospital stay [24]. Retrospective cost analyses in various European countries estimated mean CDI attributable direct costs per hospital stay of €4396 to €14,023 [57]. The majority of primary CDI cases respond to antibiotic treatment; however, recurrence (rCDI) occurs in 25% of these cases with a 38% to 45% chance of subsequent recurrences [5, 812].
The phase 3 MODIFY I and II trials demonstrated that bezlotoxumab, a human monoclonal antibody against C. difficile toxin B, when given during standard of care (SOC) antibiotic treatment for an active CDI, significantly reduced rCDI in adults at high risk for rCDI [13, 14]. Healthcare practices and reimbursement methods differed across the countries that enrolled patients in the MODIFY trials. For example, European participants were more frequently treated in an inpatient setting compared with participants in North America. Additionally, the distribution of strains of C. difficile isolated from European participants differs from strains identified in North American participants, with a lower percentage of European participants infected with the 027 strain [1518]. Given these differences, the objective of this analysis was to assess the efficacy of bezlotoxumab in preventing rCDI, as well as other secondary efficacy outcomes, in the subgroup of participants enrolled in the European region.

Materials and methods

The MODIFY I/II (NCT01241552 and NCT01513239) trials were randomized, double-blind, placebo-controlled, multicenter, phase 3 studies conducted from November 2011 through May 2015 at 322 sites in 30 countries, including 17 countries in the European region (Fig. 1). Both MODIFY I and II were conducted in accordance with the Good Clinical Practice guidelines and the Declaration of Helsinki. The protocols and amendments were approved by the institutional review board or independent ethics committee at each study site. Written informed consent was provided by all participants before the trial began.
Adult participants with primary or rCDI were receiving antibiotic treatment for CDI (metronidazole, vancomycin, or fidaxomicin, selected by the treating physician) for 10–14 days. Eligibility criteria were described previously [14]. CDI was defined as diarrhea (≥ 3 unformed bowel movement [types 5 to 7 on the Bristol stool scale] [19] in 24 h) associated with a positive local laboratory stool test result for toxigenic C. difficile. Eligible participants received a single 60-min infusion of bezlotoxumab 10 mg/kg or a placebo (0.9% saline) while continuing to receive antibiotic treatment. Randomization was stratified by oral antibacterial treatment (metronidazole, vancomycin, or fidaxomicin) and hospitalization status (inpatient or outpatient). Participants recorded their daily loose stool counts for 12 weeks and provided a stool sample for testing if they had a return of diarrhea.

Population, endpoints, and statistical analysis

The analysis included the subpopulation of MODIFY I/II participants enrolled by investigators residing in the European region. The modified intent-to-treat (mITT) population was the analysis population for the initial clinical cure (ICC) and sustained clinical cure (SCC) endpoints and included all randomly assigned participants who received study infusion, had a positive baseline stool test for toxigenic C. difficile, and initiated standard of care (SOC) antibacterial therapy prior to or within 1 day after receiving study treatment. The population was further categorized based on known risk factors for rCDI or CDI-related adverse outcomes that were prespecified in the protocols: age ≥ 65 years, history of CDI in the previous 6 months, compromised immunity, severe CDI (Zar score ≥ 2 points) at the time of randomization, and isolation of a strain associated with poor outcomes (ribotypes 027, 078, or 244). ICC was defined as SOC given for ≤ 16 days and no diarrhea on the two consecutive days after SOC end. The primary endpoint was rCDI, defined as new diarrhea associated with toxigenic C. difficile in stool within 12 weeks following infusion in participants who had achieved ICC (clinical cure population). SCC was defined as ICC and no rCDI during the 12-week follow-up period. The rates of all-cause and CDI-related rehospitalization within 30 days of antibiotic treatment were assessed in mITT participants who were inpatients at the time of randomization, were subsequently discharged, and then were rehospitalized within 30 days of discharge. Rehospitalizations were characterized as CDI-associated if they were due to rCDI or if rCDI occurred during the rehospitalization. The all patients as treated (APaT) population consisted of all randomized patients who received an infusion of study medication and was used for the analysis of mortality. Participant disposition and baseline demographic and clinical characteristics of the mITT population were summarized descriptively using frequencies and percent for each treatment group unless specified otherwise. Observed rCDI rates among participants who achieved ICC, ICC rates, and SCC rates along with rate differences between treatment groups and their 95% confidence intervals (CI) are presented. The 95% CIs are based on Miettinen and Nurminen’s method [20]. The nonparametric Kaplan-Meier (KM) method was used to estimate the distribution of time to CDI recurrence for each treatment group. Other outcomes of interest during the 12-week follow-up period are summarized descriptively using frequencies and percent for each treatment group.

Results

There were 1554 participants in the MODIFY trials; 606 were enrolled at a European site, including 313 in the bezlotoxumab group (51.7%) and 293 in the placebo group (48.3%). Figure 1 shows the distribution of participants by European country. Baseline characteristics were generally similar across groups (Table 1), although compared with placebo, there were more immunocompromised participants (27.2% vs 20.1%) and participants with 2 prespecified risk factors for rCDI (43.5% vs 33.9%) in the bezlotoxumab group. Conversely, there were approximately 6% more participants with renal impairment, 1 prespecified risk factor for rCDI, and a hypervirulent strain (i.e., ribotypes 027, 078, or 244) in the placebo group compared with the bezlotoxumab group. Slightly more than half (54.8%) of the participants were female, and 86.0% of all enrolled patients were hospitalized at the time of randomization.
Table 1
Baseline demographics and clinical characteristics (mITT population)
 
Bezlotoxumab (n = 313)
Placebo (n = 293)
Demographics
  Mean age (years (SD))
65.7 (17.0)
66.9 (16.8)
    Median
68
70
    Range
19-97
18-96
  18 to < 50 years
47 (15.0)
45 (15.4)
  50 to < 65 years
82 (26.2)
63 (21.5)
  65 to < 80 years
113 (36.1)
109 (37.2)
  ≥ 80 years
71 (22.7)
76 (25.9)
  Female
169 (54.0)
163 (55.6)
Standard of care antibiotic
  Metronidazole
150 (47.9)
139 (47.4)
  Vancomycin
152 (48.6)
148 (50.5)
  Fidaxomicin
11 (3.5)
6 (2.0)
Clinical characteristics
  ≥ 65 years of agea
184 (58.8)
185 (63.1)
  Primary CDI
209 (66.8)
193 (65.9)
  ≥ 1 CDI episodes in past 6 monthsa
84 (26.8)
80 (27.3)
  1 previous CDI episode ever
57 (18.2)
40 (13.7)
  ≥ 2 previous CDI episodes ever
40 (13.1)
51 (18.0)
  Severe CDI (Zar score ≥ 2)a,b
57 (18.2)
57 (19.5)
  Immunocompromiseda,c
85 (27.2)
59 (20.1)
  Inpatient at time of randomization
265 (84.7)
256 (87.4)
  Antibiotic used during SOC
123 (39.3)
115 (39.2)
  Antibiotic used after SOC
102 (32.6)
95 (32.4)
  Charlson index ≥ 3
145 (46.3)
128 (43.7)
  Renal impairmente
60 (19.2)
40 (13.7)
  Hepatic impairmentf
25 (8.0)
16 (5.5)
  Albumin < 2.5 g/dL
57 (18.2)
48 (16.4)
C. difficile straing
  Participants with a positive baseline culture
191
179
  Ribotype 027, 078, or 244 straina
37 (19.4)
45 (25.1)
  Ribotype 027 strain
28 (14.7)
36 (20.1)
Prespecified risk factors
  0 risk factors
58 (18.5)
51 (17.4)
  Participants with ≥ 1 risk factor
255 (81.5)
242 (82.6)
    1 risk factor
107 (42.0)
116 (47.9)
    2 risk factors
111 (43.5)
82 (33.9)
    ≥3 risk factors
37 (14.5)
44 (18.2)
aPrespecified risk factor
bBased on the following: (1) age > 60 years (1 point), (2) body temperature > 38.3 °C (> 100 °F) (1 point), (3) albumin level < 2.5 g/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
dSystemic antibiotic other than SOC antibiotic given to treat CDI
eRenal impairment defined as serum creatinine ≥ 1.5 mg/dL
fHepatic impairment defined by two or more of the following: (1) albumin ≤ 3.1 g/dL, (2) ALT ≥ 2× ULN, (3) total bilirubin ≥ 1.3× ULN, or (4) mild, moderate, or severe liver disease (as reported on the Charlson index)
gDenominator is subjects in the mITT population with a positive baseline culture
The rate of ICC was similar between treatment groups (bezlotoxumab vs placebo 82.4% vs 81.2%; difference [95% CI] 1.2 [− 5.0, 7.4]; Fig. 2). The rate of rCDI was lower in the bezlotoxumab group compared with the placebo group overall (bezlotoxumab vs placebo 18.2% vs 29.8%; difference [95% CI] − 11.6 [− 19.1, − 4.1]), and among those with ≥ 1 risk factor for rCDI (bezlotoxumab vs placebo 19.2% vs 33.2%; difference [95% CI] − 13.9 [− 22.4, − 5.4]; Fig. 2). The rate of SCC was higher in the bezlotoxumab group compared with the placebo group overall (bezlotoxumab vs placebo 67.4% vs 57.0%; difference [95% CI] 10.4 [2.7, 18.0]), and among those with ≥ 1 risk factor for rCDI (bezlotoxumab vs placebo 65.9% vs 54.1%; difference [95% CI] 11.8 [3.1, 20.2]; Fig. 2).
Figure 3 shows the rCDI rates by risk factor subgroups. Compared with placebo, treatment with bezlotoxumab was associated with a lower rate of rCDI in participants with ≥ 1 prespecified risk factor for rCDI (33% vs 19%, respectively; difference − 14%) particularly in those ≥ 65 years of age (34% vs 18%, respectively; difference − 16%) and with severe CDI (36% vs 15%, respectively; difference − 21%). Compared with placebo, treatment with bezlotoxumab was associated with a lower rate of rCDI in all participants with primary CDI (25% vs 13%, respectively; difference − 12%), with a larger difference observed in participants with primary CDI who had at least 1 risk factor for rCDI (29% vs 12%, respectively; difference − 16%). Participants who were hospitalized at the time of infusion and those who were treated with vancomycin experienced a lower rate of rCDI when treated with bezlotoxumab compared with placebo (Fig. 3). In other subgroups, the difference favored bezlotoxumab; however, confidence intervals were wide and included zero, most likely due to the small sample sizes (Fig. 3).
The week 12 Kaplan-Meier CDI recurrence event rate was lower in the bezlotoxumab treatment group compared with the event rate in the placebo group (Fig. 4). The majority (approximately 82%) of all recurrences occurred within the first 4 weeks following the infusion in both treatment groups (Fig. 4). The differences in the distributions of times to CDI recurrences between the bezlotoxumab group and the placebo group were observed as early as 3 weeks post-infusion, and this continued to increase throughout the 12-week follow-up period (Fig. 4).
In participants who experienced an episode of rCDI during the 12-week follow-up period, diarrhea duration was generally similar between groups. However, a higher proportion of placebo participants had severe disease (13% in the placebo group vs 9% in the bezlotoxumab group), and a higher percentage of placebo participants were treated with a CDI antibiotic for the recurrence compared with bezlotoxumab participants (70% vs 66%; Table 2). Bezlotoxumab participants had a lower proportion of CDI-associated rehospitalization within 30 days of discharge compared with placebo participants (5% vs 15%; Table 3), while all-cause rehospitalizations and the proportion of participants who died within 30 days or 90 days of randomization were similar between groups (Table 3).
Table 2
Time to resolution, severity, and treatment of rCDI episode (clinical cure population who experienced rCDI during the 12-week follow-up period)
 
Bezlotoxumab (n (%))
Placebo (n (%))
 
n = 47
n = 71
Diarrhea severity and duration
Maximum number of loose stools during  CDI episode (median)
5 (10.6)
6 (8.5)
  Time to resolution of new episode (days)
    ≤ 2
24 (51.1)
34 (47.9)
    3 or more
23 (48.9)
37 (52.1)
  Severe CDI (Zar score ≥ 2)
4 (8.5)
9 (12.7)
Any CDI antibiotic treatmenta
31 (66.0)
50 (70.4)
aAny vancomycin, metronidazole, or fidaxomicin
Table 3
Additional outcomes in European participants
 
Bezlotoxumab (n (%))
Placebo (n (%))
 
n = 265
n = 256
30-day rehospitalizationa
  All-cause
61 (23.0)
68 (26.6)
  CDI-associated
13 (4.9)
38 (14.8)
Deathb
  30-day mortality
16 (5.1)
15 (5.1)
  90-day mortality
31 (9.9)
31 (10.5)
amITT patients who were inpatients at randomization
bAPaT, all patients as treated population

Discussion

The results of this post hoc analysis used pooled data to evaluate the efficacy of bezlotoxumab treatment in European participants enrolled in the MODIFY I and II trials. The results of this analysis were consistent with the primary results from MODIFY I and II, which demonstrated that treatment with bezlotoxumab reduced CDI recurrence over a 12-week period compared with placebo in participants overall and in those with at least 1 risk factor [14].
C. difficile has multiple strain types, some of which are associated with greater virulence, more severe disease, and increased complications and recurrences [2123]. The EUCLID surveillance study identified 125 different ribotypes in 19 countries in Europe [18], while a study conducted by the Centers for Disease Control identified 143 distinct ribotypes in the USA [15]. The most commonly identified strains in both Europe and North America include ribotypes 001, 014, 020, 027, and 078; however, the prevalence differs between the two continents [1518, 24]. The rates of ribotype 027 decreased in prevalence in the USA and Canada between 2009 and 2015, but ribotype 027 was more prevalent in Eastern European countries in 2015 compared with 2009 while the remaining unchanged in other parts of Europe [25]. Importantly, the differences in strain types and the differences in their distribution between North America and Europe did not appear to impact the efficacy of bezlotoxumab in reducing the rate of rCDI in the European population compared with the global population enrolled in the MODIFY trials.
Hospital costs account for the majority of costs in CDI and rCDI cases [26]. Therefore, the longer hospital stays and higher readmission rates associated with rCDI have a significant adverse economic impact in many European countries. In Germany, the mean length of hospital stay for a rCDI infection, often associated with admission to ICU, is 94 days at a cost of €73,900 (95% CI €50,340, €97,460) compared with 32 days for CDI at a cost of €18,460 (95% CI €14,660, €22,270) [5]. A second study conducted in 6 UK hospitals showed that the median cost of rCDI infection was £7539 (IQR £5617, £9730) for a mean length of stay of 21 days compared with £6294 (IQR £2700, £9216) for a length of stay of 15.5 days for a first CDI episode [26]. In the current analysis of European participants, there was a greater proportion of hospitalized patients compared with the overall population (86% vs 68%). In participants who were hospitalized at randomization, the rate of 30-day rehospitalizations associated with CDI was lower in the bezlotoxumab treatment group compared with the placebo group. This is consistent with a similar post hoc analysis of the MODIFY trials, which demonstrated that bezlotoxumab-treated inpatients experienced fewer CDI-associated readmissions during the 30 days after discharge compared with placebo-treated inpatients [27].
There were some limitations of this analysis: First, it was a post hoc analysis, so it was not powered to assess statistical significance. The results should be interpreted as hypothesis generating. Secondly, the possible effect of bezlotoxumab on the resolution of symptoms of the episode under treatment at the time of randomization could not be evaluated because the majority of participants had been receiving antibiotics for at least 3 days prior to receiving bezlotoxumab or placebo and symptoms had already resolved. In addition, not all regions in all countries across Europe were represented in the MODIFY trials; therefore, the results may not be generalizable to all patients living in the European region. However, the participants included in the analysis did represent the majority of the region and likely included the most common C. difficile strains found in the European region.
In conclusion, treatment with bezlotoxumab reduced CDI recurrence over a 12-week period and was associated with fewer CDI-associated rehospitalizations within 30 days of discharge in European participants, with the presumed reduction in the economic cost of patient management. Therefore, it is reasonable to consider the use of bezlotoxumab as an adjunct to standard of care antibacterial therapy in European patients with one or more risk factors for rCDI.

Acknowledgments

Carol Zecca, BS, of Merck & Co., Inc., Kenilworth, NJ, USA, provided editorial and submission support. We gratefully acknowledge the MODIFY I/II trial investigators and especially the individuals who consented to participate.

Code availability

N/A

Compliance with ethical standards

Conflict of interest

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.

Ethics approval

Both MODIFY I and II were conducted in accordance with the Good Clinical Practice guidelines and the Declaration of Helsinki. The protocols and amendments were approved by the institutional review board or independent ethics committee at each study site. Written informed consent was provided by all participants before the trial began.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Suetens C, Hopkins S, Kolman J et al (2013) Point prevalence survey of healthcare-associated infections and antimicrobial use in European acute care hospitals. ECDC, Stockholm, pp 1–216 Suetens C, Hopkins S, Kolman J et al (2013) Point prevalence survey of healthcare-associated infections and antimicrobial use in European acute care hospitals. ECDC, Stockholm, pp 1–216
2.
Zurück zum Zitat Asensio A, Di Bella S, Lo Vecchio A et al (2015) 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 36:31–38CrossRef Asensio A, Di Bella S, Lo Vecchio A et al (2015) 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 36:31–38CrossRef
3.
Zurück zum Zitat Tresman R, Goldenberg SD (2018) Healthcare resource use and attributable cost of Clostridium difficile infection: a micro-costing analysis comparing first and recurrent episodes. J Antimicrob Chemother 73:2851–2855CrossRef Tresman R, Goldenberg SD (2018) Healthcare resource use and attributable cost of Clostridium difficile infection: a micro-costing analysis comparing first and recurrent episodes. J Antimicrob Chemother 73:2851–2855CrossRef
4.
Zurück zum Zitat Leblanc S, Blein C, Andremont A et al (2017) Burden of Clostridium difficile infections in French hospitals in 2014 from the national health insurance perspective. Infect Control Hosp Epidemiol 38:906–911CrossRef Leblanc S, Blein C, Andremont A et al (2017) Burden of Clostridium difficile infections in French hospitals in 2014 from the national health insurance perspective. Infect Control Hosp Epidemiol 38:906–911CrossRef
5.
Zurück zum Zitat Heimann SM, Vehreschild JJ, Cornely OA et al (2015) Economic burden of Clostridium difficile associated diarrhoea: a cost-of-illness study from a German tertiary care hospital. Infection 43:707–714CrossRef Heimann SM, Vehreschild JJ, Cornely OA et al (2015) Economic burden of Clostridium difficile associated diarrhoea: a cost-of-illness study from a German tertiary care hospital. Infection 43:707–714CrossRef
6.
Zurück zum Zitat Le Monnier A, Duburcq A, Zahar JR et al (2015) Hospital cost of Clostridium difficile infection including the contribution of recurrences in French acute-care hospitals. J Hosp Infect 91:117–122CrossRef Le Monnier A, Duburcq A, Zahar JR et al (2015) Hospital cost of Clostridium difficile infection including the contribution of recurrences in French acute-care hospitals. J Hosp Infect 91:117–122CrossRef
7.
Zurück zum Zitat Poli A, Di Matteo S, Bruno GM et al (2015) Economic burden of Clostridium difficile in five hospitals of the Florence health care system in Italy. Risk Manag Healthc Policy 8:207–213PubMedPubMedCentral Poli A, Di Matteo S, Bruno GM et al (2015) Economic burden of Clostridium difficile in five hospitals of the Florence health care system in Italy. Risk Manag Healthc Policy 8:207–213PubMedPubMedCentral
8.
Zurück zum Zitat Cornely OA, Miller MA, Louie TJ et al (2012) Treatment of first recurrence of Clostridium difficile infection: fidaxomicin versus vancomycin. Clin Infect Dis 55(Suppl 2):S154–SS61CrossRef Cornely OA, Miller MA, Louie TJ et al (2012) Treatment of first recurrence of Clostridium difficile infection: fidaxomicin versus vancomycin. Clin Infect Dis 55(Suppl 2):S154–SS61CrossRef
9.
Zurück zum Zitat Johnson S, Louie TJ, Gerding DN et al (2014) Vancomycin, metronidazole, or tolevamer for Clostridium difficile infection: results from two multinational, randomized, controlled trials. Clin Infect Dis 59:345–354CrossRef Johnson S, Louie TJ, Gerding DN et al (2014) Vancomycin, metronidazole, or tolevamer for Clostridium difficile infection: results from two multinational, randomized, controlled trials. Clin Infect Dis 59:345–354CrossRef
10.
Zurück zum Zitat Louie TJ, Miller MA, Mullane KM et al (2011) Fidaxomicin versus vancomycin for Clostridium difficile infection. N Engl J Med 364:422–431CrossRef Louie TJ, Miller MA, Mullane KM et al (2011) Fidaxomicin versus vancomycin for Clostridium difficile infection. N Engl J Med 364:422–431CrossRef
11.
Zurück zum Zitat Sheitoyan-Pesant C, Abou Chakra CN, Pepin J et al (2016) Clinical and healthcare burden of multiple recurrences of Clostridium difficile infection. Clin Infect Dis 62:574–580CrossRef Sheitoyan-Pesant C, Abou Chakra CN, Pepin J et al (2016) Clinical and healthcare burden of multiple recurrences of Clostridium difficile infection. Clin Infect Dis 62:574–580CrossRef
12.
Zurück zum Zitat Surawicz CM (2004) Treatment of recurrent Clostridium difficile-associated disease. Nat Clin Pract Gastroenterol Hepatol 1:32–38CrossRef Surawicz CM (2004) Treatment of recurrent Clostridium difficile-associated disease. Nat Clin Pract Gastroenterol Hepatol 1:32–38CrossRef
13.
Zurück zum Zitat Gerding DN, Kelly CP, Rahav G et al (2018) Bezlotoxumab for prevention of recurrent Clostridium difficile infection in patients at increased risk for recurrence. Clin Infect Dis 67:649–656CrossRef Gerding DN, Kelly CP, Rahav G et al (2018) Bezlotoxumab for prevention of recurrent Clostridium difficile infection in patients at increased risk for recurrence. Clin Infect Dis 67:649–656CrossRef
14.
Zurück zum Zitat Wilcox MH, Gerding DN, Poxton IR et al (2017) Bezlotoxumab for prevention of recurrent Clostridium difficile infection. N Engl J Med 376:305–317CrossRef Wilcox MH, Gerding DN, Poxton IR et al (2017) Bezlotoxumab for prevention of recurrent Clostridium difficile infection. N Engl J Med 376:305–317CrossRef
15.
Zurück zum Zitat (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. (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.
16.
Zurück zum Zitat Freeman J, Vernon J, Pilling S et al (2018) 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 24:724–731CrossRef Freeman J, Vernon J, Pilling S et al (2018) 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 24:724–731CrossRef
17.
Zurück zum Zitat Chitnis AS, Holzbauer SM, Belflower RM et al (2011) Epidemiology of community-associated Clostridium difficile infection, 2009 through. JAMA Intern Med 2013:1–9 Chitnis AS, Holzbauer SM, Belflower RM et al (2011) Epidemiology of community-associated Clostridium difficile infection, 2009 through. JAMA Intern Med 2013:1–9
18.
Zurück zum Zitat 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 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
19.
Zurück zum Zitat Lewis SJ, Heaton KW (1997) Stool form scale as a useful guide to intestinal transit time. Scand J Gastroenterol 32:920–924CrossRef Lewis SJ, Heaton KW (1997) Stool form scale as a useful guide to intestinal transit time. Scand J Gastroenterol 32:920–924CrossRef
20.
Zurück zum Zitat Miettinen O, Nurminen M (1985) Comparative analysis of two rates. Stat Med 4:213–226CrossRef Miettinen O, Nurminen M (1985) Comparative analysis of two rates. Stat Med 4:213–226CrossRef
21.
Zurück zum Zitat Sundram F, Guyot A, Carboo I et al (2009) Clostridium difficile ribotypes 027 and 106: clinical outcomes and risk factors. J Hosp Infect 72:111–118CrossRef Sundram F, Guyot A, Carboo I et al (2009) Clostridium difficile ribotypes 027 and 106: clinical outcomes and risk factors. J Hosp Infect 72:111–118CrossRef
22.
Zurück zum Zitat Loo VG, Poirier L, Miller MA et al (2005) A predominantly clonal multi-institutional outbreak of Clostridium difficile-associated diarrhea with high morbidity and mortality. N Engl J Med 353:2442–2449CrossRef Loo VG, Poirier L, Miller MA et al (2005) A predominantly clonal multi-institutional outbreak of Clostridium difficile-associated diarrhea with high morbidity and mortality. N Engl J Med 353:2442–2449CrossRef
23.
Zurück zum Zitat McDonald LC, Killgore GE, Thompson A et al (2005) An epidemic, toxin gene-variant strain of Clostridium difficile. N Engl J Med 353:2433–2441CrossRef McDonald LC, Killgore GE, Thompson A et al (2005) An epidemic, toxin gene-variant strain of Clostridium difficile. N Engl J Med 353:2433–2441CrossRef
24.
Zurück zum Zitat Stubbs SL, Brazier JS, O’Neill GL et al (1999) 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 37:461–463CrossRef Stubbs SL, Brazier JS, O’Neill GL et al (1999) 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 37:461–463CrossRef
25.
Zurück zum Zitat Cheknis A, Johnson S, Chesnel L et al (2018) Molecular epidemiology of Clostridioides (Clostridium) difficile strains recovered from clinical trials in the US, Canada and Europe from 2006-2009 to 2012-2015. Anaerobe 53:38–42CrossRef Cheknis A, Johnson S, Chesnel L et al (2018) Molecular epidemiology of Clostridioides (Clostridium) difficile strains recovered from clinical trials in the US, Canada and Europe from 2006-2009 to 2012-2015. Anaerobe 53:38–42CrossRef
26.
Zurück zum Zitat Wilcox MH, Ahir H, Coia JE et al (2017) Impact of recurrent Clostridium difficile infection: hospitalization and patient quality of life. J Antimicrob Chemother 72:2647–2656CrossRef Wilcox MH, Ahir H, Coia JE et al (2017) Impact of recurrent Clostridium difficile infection: hospitalization and patient quality of life. J Antimicrob Chemother 72:2647–2656CrossRef
27.
Zurück zum Zitat Prabhu VS, Cornely OA, Golan Y et al (2017) Thirty-Day readmissions in hospitalized patients who received bezlotoxumab with antibacterial drug treatment for Clostridium difficile infection. Clin Infect Dis 65:1218–1221CrossRef Prabhu VS, Cornely OA, Golan Y et al (2017) Thirty-Day readmissions in hospitalized patients who received bezlotoxumab with antibacterial drug treatment for Clostridium difficile infection. Clin Infect Dis 65:1218–1221CrossRef
Metadaten
Titel
Analysis of C. difficile infection–related outcomes in European participants in the bezlotoxumab MODIFY I and II trials
verfasst von
Emilio Bouza
Oliver A. Cornely
Antonio Ramos-Martinez
Robert Plesniak
Misoo C. Ellison
Mary E. Hanson
Mary Beth Dorr
Publikationsdatum
06.06.2020
Verlag
Springer Berlin Heidelberg
Erschienen in
European Journal of Clinical Microbiology & Infectious Diseases / Ausgabe 10/2020
Print ISSN: 0934-9723
Elektronische ISSN: 1435-4373
DOI
https://doi.org/10.1007/s10096-020-03935-3

Weitere Artikel der Ausgabe 10/2020

European Journal of Clinical Microbiology & Infectious Diseases 10/2020 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.