Next Article in Journal
The Risk and Clinical Implications of Antibiotic-Associated Acute Kidney Injury: A Review of the Clinical Data for Agents with Signals from the Food and Drug Administration’s Adverse Event Reporting System (FAERS) Database
Next Article in Special Issue
Bezlotoxumab in Patients with a Primary Clostridioides difficile Infection: A Literature Review
Previous Article in Journal
Long-Read Whole Genome Sequencing Elucidates the Mechanisms of Amikacin Resistance in Multidrug-Resistant Klebsiella pneumoniae Isolates Obtained from COVID-19 Patients
Previous Article in Special Issue
Clinical Determinants Predicting Clostridioides difficile Infection among Patients with Chronic Kidney Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Fidaxomicin for the Treatment of Clostridioides difficile Infection in Adult Patients: An Update on Results from Randomized Controlled Trials

by
Daniele Roberto Giacobbe
1,2,*,
Antonio Vena
1,2,
Marco Falcone
3,
Francesco Menichetti
3 and
Matteo Bassetti
1,2
1
Department of Health Sciences (DISSAL), University of Genoa, 16132 Genoa, Italy
2
Infectious Diseases Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
3
Infectious Diseases Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliera Universitaria Pisana, University of Pisa, 56126 Pisa, Italy
*
Author to whom correspondence should be addressed.
Antibiotics 2022, 11(10), 1365; https://doi.org/10.3390/antibiotics11101365
Submission received: 22 September 2022 / Revised: 2 October 2022 / Accepted: 4 October 2022 / Published: 6 October 2022
(This article belongs to the Special Issue Clostridioides difficile Infection, 2nd Edition)

Abstract

:
In recently updated international guidelines, fidaxomicin is preferentially recommended as first-line treatment over vancomycin both for the first episode of CDI and for rCDI, based on the results of different randomized controlled trials (RCTs). Although noninferiority was the rule in phase-3 RCTs with regard to the primary endpoint of clinical cure, for shaping these recommendations, particular attention was devoted to the improved global cure and reduced risk of recurrent CDI (rCDI) observed with fidaxomicin compared to vancomycin in RCTs. Overall, while the major driver of choice should remain the global benefit for the patient, consideration of available resources should be necessarily weighed in the balance, since fidaxomicin still remains more costly than vancomycin. Against this background, precisely stratifying risk groups for rCDI will represent a crucial research trajectory of future real-life studies on the treatment of first CDI episodes. In the current narrative review, we discuss the updated evidence from RCTs on the efficacy of fidaxomicin for the treatment of either the first CDI episode or rCDI, which eventually supports its positioning within current treatment algorithms and guidelines.

1. Introduction

Clostridioides difficile is the most common causative agent of infectious diarrhea in hospitalized patients; although, community-acquired C. difficile infection (CDI) has also become epidemiologically and clinically relevant during the last decade [1,2,3,4,5,6].
In the treatment approach to CDI, clinicians aim both to cure the index episode and to reduce the risk of recurrences. Indeed, recurrent CDI (rCDI) develops in 10–30% of cases after the first CDI episode, with the risk further increasing with each successive episode [7,8,9,10]. In the recently released updates of guidelines/guidance documents from the Infectious Diseases Society of America/Society for Healthcare Epidemiology of America (IDSA/SHEA) and from the European Society of Clinical Microbiology and Infectious Diseases (ESCMID), there have been changes in the recommendations pertaining to the use of fidaxomicin, a macrocyclic antibiotic approved both in the US and in Europe for the treatment of CDI [11,12].
In the present narrative review, we discuss the updated evidence from randomized controlled trials (RCTs) on the efficacy of fidaxomicin for the treatment of either the first CDI episode or rCDI, which eventually supports its positioning within current treatment algorithms and guidelines.

2. Methods

In August 2022, we performed a PubMed search using the keyword “fidaxomicin”. After title and abstract screening of the retrieved 657 records, 227 of them were selected for initial full-text assessment. In line with the narrative nature of the present review, relevant articles pertaining to the topic were further selected by the authors and organized in the following structure: (i) an introductory section on the characteristics, mechanism of action, and antimicrobial activity of fidaxomicin; (ii) a main section on the results from phase-3/4 RCTs; (iii) a conclusions section.

3. Characteristics, Mechanism of Action, and Antimicrobial Activity of Fidaxomicin

Fidaxomicin, administered orally, is the first member of the macrocycles class of antibiotics, and it shows bactericidal activity against C. difficile [13,14]. In addition, fidaxomicin has negligible activity against other bacteria constituting the gut microbiota [15,16]. This selective activity relies on the fact that the C. difficile RNA polymerase (inhibited by fidaxomicin [17]) has a specific residue (lysine 84) that is bound by fidaxomicin and acts as a crucial sensitizer allowing fidaxomicin killing activity [18]. This specific residue is absent in gut bacteria belonging to the phyla Bacteroides and Proteobacteria [18,19]. In line with the largely reported more favorable effect than other CDI treatments in terms of microbiota disruption [16,20,21,22,23], combined with its modest activity (although inhibitory at the achieved stool concentrations) against vancomycin-resistant enterococci (VRE) [24], fidaxomicin treatment resulted in a reduced frequency of novel stool culture positivity for vancomycin-resistant enterococci (VRE) and Candida spp. compared to vancomycin among patients with negative pre-treatment stool cultures enrolled in a phase-3 RCT (7% vs. 31% for VRE acquisition among 247 patients, p < 0.001; 19% vs. 29% for Candida spp. acquisition among 252 patients, p = 0.03) [25]. In patients with pre-treatment VRE colonization, a larger decrease in the mean stool concentration of VRE was observed with fidaxomicin therapy than with vancomycin therapy; although, selection of some subpopulations of VRE with high fidaxomicin minimum inhibitory concentration (MIC) was observed during fidaxomicin treatment [25]. Of note, in patients receiving vancomycin, the risk of colonization and subsequent bloodstream infections by Candida spp. or enterococci may be possibly higher among those receiving high vancomycin dosages (>500 mg/day) [26].
Another peculiar characteristic of fidaxomicin, not shared by other anti-C. difficile agents such as vancomycin and metronidazole, is its long post-antibiotic effect, which might be relevant considering the hastened intestinal transit and drug elimination in patients with diarrhea [14,27]. Following oral administration, fidaxomicin is poorly absorbed, reaching high intracolonic concentrations [28,29]. Together with the lack of cytochrome P450 metabolism, the very low bioavailability of fidaxomicin may explain its low potential for systemic adverse events and drug interactions [14,30,31]. The main metabolite of fidaxomicin, OP-1118, is produced in vivo by the action of an esterase, and retains antimicrobial activity against C. difficile [13,32,33].
The activity of fidaxomicin against C. difficile has been assessed in several in vitro studies. Among 403 non duplicate C. difficile isolates from Taiwan, fidaxomicin showed potent in vitro activity, with MIC90 of 0.5 mg/L (range ≤ 0.015 to 0.5 mg/L) [34]. An even lower MIC90 of 0.125 mg/L was measured among 188 C. difficile isolates from Hungary, with only four isolates displaying a MIC value of 0.5 mg/L [35]. In another surveillance study on 925 C. difficile isolates from the US, MIC90 for fidaxomicin was 0.5 mg/L, with a range from 0.004 to 4 mg/L [36]. The same MIC90 of 0.5 mg/L, with a range from 0.004 to 1 mg/L, was observed in a subsequent update on a larger sample of 1889 C. difficile isolates [37]. In a surveillance study from Japan, MIC90 for fidaxomicin was 0.25 mg/L among 100 C. difficile isolates (range 0.03 to 0.5 mg/L) [38]. A MIC90 of 0.25 mg/L for fidaxomicin was also observed in a surveillance study on 105 C. difficile isolates from Thailand (range 0.004 to 0.25 mg/L) [39]. Among 101 C. difficile isolates from China, MIC90 for fidaxomicin was 0.5 mg/L (range 0.032 to 1 mg/L), whereas it was 0.03 mg/L among 100 C. difficile isolates from the US in another study (range ≤ 0.008 to 8 mg/L) [40,41]. In a small study on 64 C. difficile isolates from the Czech Republic, MIC90 for fidaxomicin was 0.125 mg/L (range 0.06 to 0.25 mg/L) [42]. Fidaxomicin showed the greatest in vitro potency compared to the other seven antimicrobial agents tested against 1310 C. difficile isolates from Canada (with 027 being the most frequent ribotype, 24.5%), showing a MIC90 of 0.25 mg/L (range 0.055 to 2 mg/L) [43]. In a large pan-European surveillance study of 953 C. difficile isolates, MIC90 for fidaxomicin was 0.125 mg/L (range ≤ 0.002 to 0.25 mg/L), and all strains were considered susceptible according to an epidemiological cut-off of 1 mg/L [44]. In subsequent updates of the same surveillance study including up to 3499 C. difficile isolates, a fidaxomicin MIC ≥ 4 mg/L was observed only in a single case [45,46]. Low MIC90 values for fidaxomicin were also displayed by C. difficile isolates from phase-2 (38 isolates, MIC90 of 0.125 mg/L, range ≤ 0.008 to 0.25 mg/L) and phase-3 (719 isolates, MIC90 of 0.25 mg/L, range 0.03 to 1 mg/L) studies of fidaxomicin, with only one strain isolated from a patient from a phase-3 study who developed rCDI showing a fidaxomicin MIC of 16 mg/L at the time of rCDI [47,48].
According to the in vitro studies reported above, reduced susceptibility to fidaxomicin is very rare; although, it has seldom been described [49]. In vitro, reduced susceptibility to fidaxomicin was selected through serial passages in a medium over a range of drug concentrations [50]. C. difficile isolates with reduced fidaxomicin susceptibility selected through serial passages harbored mutations in rpoB, encoding the β-subunit of RNA polymerase, or in CD22120, encoding a homolog of the family of transcriptional regulators MarR [50,51]. In a subsequent study, three C. difficile mutants with reduced susceptibility to fidaxomicin (MIC of 2, 8, and >32 mg/L, respectively) after the introduction of non-synonymous single-nucleotide polymorphisms in rpoB by allelic exchange also showed attenuated growth and reduced sporulation capacity, toxin A/B production, and cytotoxicity compared with the parental strain [52]. In a hamster model, the three mutants had impaired virulence in comparison to the parental strain; although, caecum colonization capacity was similar to the parental strain [52]. In a more recent study, a V1143D mutation was characterized in the rpoB gene of a clinical C. difficile isolate with fidaxomicin MIC > 64 mg/L and was associated with a less marked fitness defect than previously reported [53].
Regarding other particular characteristics, fidaxomicin and its metabolite OP-1118, differently from vancomycin, are able to inhibit sporulation (spore formation) of C. difficile, a fact which is thought to contribute to the observed increased rates of sustained response and reduced risk of recurrence in comparisons with other treatments (see the following section), since spores may persist after completion of a successful treatment course and subsequently germinate and proliferate, leading to a novel CDI episode [54,55,56]. After spores are formed, fidaxomicin, like vancomycin, is unable to inhibit germination, but both agents are able to counteract the outgrowth of vegetative cells from germinating spores [57]. However, fidaxomicin, but not vancomycin, has been demonstrated to persist on C. difficile spores after washing in saline and fecal filtrate, with consequent higher inhibitory effect on the outgrowth of vegetative cells and toxin production [58,59]. A substantial direct inhibitory effect of fidaxomicin and OP-1118 on toxin production may also explain the less frequent detection of post-treatment toxin production in fidaxomicin-treated patients than in vancomycin-treated patients [56,60,61]. A reduction in toxin A- and toxin B-mediated inflammatory responses and colonic tissue damage has also been described following exposure to fidaxomicin [62,63]. Another effect of fidaxomicin reported in in vitro studies, not observed with vancomycin, is the inhibition of biofilm formation, which could have implications for reducing the risk of both C. difficile colonization and CDI [64,65,66,67]. Finally, reduced shedding and environmental contamination by C. difficile have been described with fidaxomicin treatment more than with metronidazole or, although to a lesser extent, vancomycin [68,69,70,71].

4. Results of Phase-3/4 Randomized Controlled Trials

A summary of the main results of phase-3/4 RCTs assessing the efficacy of fidaxomicin for the treatment of CDI in adult patients is available in Table 1.
The first two large phase-3 randomized controlled trials (RCTs) assessing the efficacy of fidaxomicin for the treatment of CDI were the OPT-80-003 and OPT-80-004 studies [72,73]. Of note, patients with life-threatening or fulminant CDI were excluded from these studies [72,73]. In the non-inferiority, double-blind OPT-80-003 RCT, fidaxomicin (200 mg orally twice daily for 10 days) was compared to vancomycin (125 mg orally four times daily for 10 days) for the treatment of CDI [73]. The primary endpoint was clinical cure (defined as resolution of the symptoms and no need for further CDI treatment), assessed on the second day after the end of treatment. The secondary endpoints were rCDI (defined as diarrhea plus toxin test positivity on stool within 4 weeks after treatment of the previous episodes) and global cure (defined as clinical cure plus lack of rCDI). The primary study populations were the modified intention to treat (mITT) population (patients with documented CDI who received at least one dose of the study drug) and the per-protocol population (patients of the mITT population who received at least 3 days of treatment in the case of failure and at least 8 days of treatment in the case of clinical cure). Regarding the primary endpoint, fidaxomicin was found to be noninferior to vancomycin in terms of clinical cure both in the mITT population (88.2% (253/287) vs. 85.8% (265/309) in the fidaxomicin and vancomycin arms, respectively; lower margin of the 97.5% confidence interval [CI] for difference equal to −3.1%) and in the per-protocol population (92.1% (244/265) vs. 89.8% (254/283) in the fidaxomicin and vancomycin arms, respectively; lower margin of the 97.5% CI for difference equal to −2.6%). Regarding secondary endpoints, a lower frequency of rCDI was observed in fidaxomicin-treated than vancomycin-treated patients, both in the mITT population (15.4% (39/253) vs. 25.3% (67/265) in the fidaxomicin and vancomycin arms, respectively, with 95% CI for the difference from −16.6% to −2.9%) and in the per-protocol population (13.3% (28/211) vs. 24.0% (53/221) in the fidaxomicin and vancomycin arms, respectively, with 95% CI for the difference from −17.9% to −3.3%). A reduced frequency of rCDI in fidaxomicin-treated than in vancomycin-treated patients was retained in most subgroups; although, not in the subgroup of patients with CDI due to the 027 ribotype (27.1% (16/59) vs. 20.9% (14/67) in the fidaxomicin and vancomycin arms, respectively, in the mITT population). Finally, a higher frequency of global cure was registered in fidaxomicin-treated than in vancomycin-treated patients, both in the mITT population (74.6% (214/287) vs. 64.1% (198/309) in the fidaxomicin and vancomycin arms, respectively, with 95% CI from 3.1% to 17.7%) and in the per-protocol population (77.7% (206/265) vs. 67.1% (190/283) in the fidaxomicin and vancomycin arms, respectively, with 95% CI for the difference from 3.1% to 17.9%) [73]. Dosing schedules, endpoints, and primary study populations of the OPT-80-004 study were defined as in the OPT-80-003 study [72,73]. With regard to clinical cure (primary endpoint), fidaxomicin achieved noninferiority to vancomycin also in the OPT-80-004 study, both in the mITT population (87.7% (221/252) vs. 86.8% (223/257) in the fidaxomicin and vancomycin arms, respectively; lower margin of the 97.5% CI for difference equal to −4.9%) and in the per-protocol population (91.7% (198/216) vs. 90.6% (213/235) in the fidaxomicin and vancomycin arms, respectively; lower margin of the 97.5% CI for difference equal to −4.3%). A lower frequency of rCDI was observed in fidaxomicin-treated than in vancomycin-treated patients also in the OPT-80-004 study, both in the mITT population (12.7% (28/221) vs. 26.9% (60/223) in the fidaxomicin and vancomycin arms, respectively, with 95% CI for the difference from −21.4% to −6.8%) and in the per-protocol population (12.8% (23/180) vs. 25.3% (46/182) in the fidaxomicin and vancomycin arms, respectively, with 95% CI for the difference from −20.3% to −4.4%). Differently from the OTP-80-003 study, in the OTP-80-004 study, a lower frequency of rCDI in the fidaxomicin arm was also registered in the subgroup of patients with CDI due to the 027 ribotype (22.2% (12/54) vs. 38.0% (19/50) in the fidaxomicin and vancomycin arms, respectively, in the mITT population). As in the OTP-80-003 study, a higher frequency of global cure (also defined as a sustained response) was registered in fidaxomicin-treated than in vancomycin-treated patients, both in the mITT population (76.6% (193/252) vs. 63.4% (163/257) in the fidaxomicin and vancomycin arms, respectively, with 95% CI from 5.2% to 20.9%) and in the per-protocol population (79.6% (172/216) vs. 65.5% (154/235) in the fidaxomicin and vancomycin arms, respectively, with 95% CI for the difference from 5.9% to 22.1%) [72].
Different meta-analyses were conducted by pooling data from OTP-80-003 and OTP-80-004 after the two RCTs were released. In one of them, an exploratory, post hoc time-to-event analysis was conducted by means of fixed-effect meta-analysis and Cox regression [74]. Overall, the analysis included 1164 patients (ITT population) from the two RCTs and showed a reduction of persistent diarrhea, rCDI, or death (composite endpoint) of 40% (95% CI from 26% to 51%) through day 40 in fidaxomicin-treated patients vs. vancomycin-treated patients [74]. In another meta-analysis pooling data from the two RCTs, the odds ratio (OR) for clinical cure was 1.17 for fidaxomicin vs. vancomycin as reference (95% CI from 0.82 to 1.66) [75]. In subgroup analyses, the OR for clinical cure was 1.45 (95% CI from 0.63 to 3.36) and 0.86 (95% CI from 0.50 to 1.47) in patients with non-severe CDI and severe CDI, respectively. The OR for rCDI was 0.47 for fidaxomicin vs. vancomycin as a reference (95% CI from 0.34 to 0.65). In subgroup analyses, the OR for rCDI was 0.49 (95% CI from 0.32 to 0.74) and 0.46 (95% CI from 0.26 to 0.79) in patients with non-severe CDI and severe CDI, respectively. The OR for global cure was 1.75 for fidaxomicin vs. vancomycin as a reference (95% CI from 1.35 to 2.27). In subgroup analyses, the pooled OR for global cure was 1.92 (95% CI from 1.37 to 2.69) and 1.49 (95% CI from 0.99 to 2.26) in patients with non-severe CDI and severe CDI, respectively [75]. In another meta-analysis with pooled data from OTP-80-003 and OTP-80-004, the OR for symptomatic cure (defined as initial resolution of diarrhea and no evidence of recurrence up to 4 weeks) was 1.17 for fidaxomicin vs. vancomycin as reference (95% CI from 1.07 to 1.27) [76].
Combining data from the OTP-80-003 and OTP-80-004 studies, the efficacy of fidaxomicin vs. vancomycin for the treatment of CDI was evaluated in the following subgroups of patients with CDI: (i) patients who were concomitantly receiving other antibiotics for concomitant infections; and (ii) patients who were not receiving other concomitant antibiotics [77]. In presence of other concomitant antibiotic treatments, the clinical cure was higher in fidaxomicin-treated than in vancomycin-treated patients (90.0% (81/90) vs. 79.4% (81/102) in the fidaxomicin and vancomycin arms, respectively, with 95% CI for the difference from 0.2% to 20.4%), whereas clinical cure was similar between the two arms in the absence of other concomitant antibiotic treatments (92.3% (361/391) vs. 92.8% (386/416) in the fidaxomicin and vancomycin arms, respectively, with 95% CI for the difference from −4.1% to 3.2%). With regard to secondary endpoints, rates of rCDI and global cure were lower and higher, respectively, in fidaxomicin-treated than in vancomycin-treated patients both in patients receiving concomitant antibiotics and in patients not receiving concomitant antibiotics, in line with the main results of OTP-80-003 and OTP-80-004 [77]. Another exploratory post hoc analysis of combined data from OTP-80-003 and OTP-80-004 was conducted in the subgroups of CDI patients with and without cancer [78]. In patients with cancer, the clinical cure was 85.1% (74/87) and 74.0% (71/96) in patients treated with fidaxomicin and in patients treated with vancomycin, respectively (OR 2.00, with 95% CI from 0.95 to 4.22). In patients without cancer, the clinical cure was 88.5% (400/452) and 88.7% (417/470) in patients treated with fidaxomicin and in patients treated with vancomycin, respectively (odds ratio (OR) 0.98, with 95% CI from 0.65 to 1.47). The rates of rCDI and global cure were lower and higher, respectively, in fidaxomicin-treated than in vancomycin-treated patients both in patients with cancer and in patients without cancer, again in line with the main results of OTP-80-003 and OTP-80-004. Of note, the median time to resolution of diarrhea was longer in patients with cancer than in those without cancer in the vancomycin arm (123 h vs. 58 h, log-rank p < 0.001), but not in the fidaxomicin arm (74 h vs. 54 h, log-rank p = 0.145) [78]. Another study combining data from OTP-80-003 and OTP-80-004 employed restriction endonuclease analysis (REA) typing on paired isolates from the index episode and recurrence (available from 90/146 patients with rCDI), to differentiate between relapse (identical REA type strain) and reinfection (different REA type strain) [79]. There was no comparison between fidaxomicin and vancomycin in terms of the study endpoints of the original RCTs, whereas a comparison between the two agents was made in terms of mean time to relapse and reinfection. The mean time to relapse in fidaxomicin-treated and in vancomycin-treated patients was 11.2 days (standard deviation (SD) ±6.1) and 14.3 days (SD ±6.2), respectively (t test, p = 0.044). The mean time to reinfection in fidaxomicin-treated and in vancomycin-treated patients was 13.9 days (SD ±7.5) and 16.8 days (SD ±4.6), respectively (t test, p = 0.497) [79]. In a further study combining data from OTP-80-003 and OTP-80-004 and employing whole-genome sequencing for distinguishing relapse (paired samples from CDI and rCDI ≤ 2 single-nucleotide variants apart) from reinfection (paired samples from CDI and rCDI > 2 single-nucleotide variants apart), the reduction in the risks of relapse and reinfection in fidaxomicin-treated vs. vancomycin-treated patients was explored using competing risk models (subdistribution hazard ratio (sHR) 0.40 for relapse, with 95% CI from 0.25 to 0.66; sHR 0.33 for reinfection, with 95% CI from 0.11 to 1.01) [80]. Regarding patients with rCDI, their possible differential risk of developing further recurrences based on fidaxomicin vs. vancomycin treatment was explored in a subset analysis of combined data from the OTP-80-003 and OTP-80-004 studies, including 128 patients who had a recent CDI episode before the index episode leading to enrollment [81]. In this analysis, the frequency of clinical cure was similar in fidaxomicin-treated and vancomycin-treated patients (93.7% (74/79) vs. 91.6% (76/83) in the fidaxomicin and vancomycin arms, respectively), whereas the frequency of rCDI (in this subgroup representing a second occurrence of rCDI) was lower in fidaxomicin-treated than in vancomycin-treated patients (19.7% (13/66) vs. 35.5% (22/62) in the fidaxomicin and vancomycin arms, respectively, with 95% CI for the difference from −30.4% to −0.3%) [81]. Finally, treatment with fidaxomicin was associated with a 60% reduced risk of recurrence in comparison with vancomycin in a logistic regression model adjusted for C. difficile strain, age, and concomitant antibiotics in 567 patients from OTP-80-003 and OTP-80-004 studies [82].
Subsequently, a phase-3 study was also conducted in Japan to assess the efficacy of fidaxomicin vs. vancomycin for the treatment of CDI. The drugs were administered at the same dosages of OTP-80-003 and OTP-80-004 [83]. The primary endpoint was global cure, which was assessed in the full analysis set (FAS) population and achieved in 67.3% (70/104) and 65.7% (71/108) of fidaxomicin-treated and vancomycin-treated patients, respectively (95% CI for the difference from −11.3 to 13.7, thereby not allowing demonstration of noninferiority). In a post hoc analysis of FAS patients who received at least 3 days of treatment, the global cure was 72.2% (70/97) and 67.0% (71/106) in fidaxomicin-treated and vancomycin-treated patients, respectively (95% CI for the difference from −7.9% to 17.1%). The frequency of rCDI in the FAS for recurrence (FAS-R) population, composed of FAS patients who achieved clinical cure during the index episode, was 19.5% (17/87) and 25.3% (24/95) in fidaxomicin-treated and vancomycin-treated patients, respectively (95% CI for the difference from −16.7% to 7.0%) [83]. In a network meta-analysis including pooled data from the RCT conducted in Japan, the OTP-80-003 RCT, and the OTP-80-004 RCT, the clinical cure was found to be similar between fidaxomicin and vancomycin (OR 1.17 with vancomycin as a reference, with 95% credible intervals from 0.78 to 1.48), whereas fidaxomicin showed a favorable association both with rCDI (OR 0.50 with vancomycin as a reference, with 95% credible intervals from 0.37 to 0.68) and global cure (OR 1.61 with vancomycin as a reference, with 95% credible intervals from 1.27 to 2.05) [84].
Three other small RCTs assessing the efficacy of fidaxomicin at standard dosage (200 mg twice daily for 10 days) for the treatment of CDI were recently published [61,85,86]. In one of them, the standard dosage of fidaxomicin was compared with vancomycin (125 mg four times daily for 10 days) for the treatment of first CDI episodes [85]. The primary endpoint was the percentage of subjects achieving a reduction of at least 2 log10 colony-forming units (CFU)/g of spores in stools from screening to the end of therapy, and was achieved more frequently in fidaxomicin-treated than vancomycin-treated patients (67% (8/12) vs. 14% (1/7)) [85]. In another small, pilot RCT of 12 patients, the standard dosage of fidaxomicin was compared with vancomycin (125 mg four times daily for 10 days) with respect to the reduction in toxin concentrations in stools from baseline, with results suggesting a favorable association between fidaxomicin and a sustained reduction in toxins A and B up to day 30 after therapy [61]. Finally, 64 patients with rCDI were randomized into three arms (standard dosage fidaxomicin, standard dosage vancomycin, and fecal microbiota transplant (FMT)) in a third small RCT [86]. The primary endpoint was a combination of clinical resolution and a negative toxin polymerase chain reaction at 8 weeks after allocation, and was achieved in 33% (8/24), 19% (3/16), and 71% (17/24) of patients receiving fidaxomicin, vancomycin, and FMT, respectively [86]. A recent meta-analysis pooling data also from these three small RCTs in addition to OTP-80-003, OTP-80-004, and the RCT conducted in Japan, showed a comparable clinical cure between fidaxomicin and vancomycin (risk ratio (RR) 1.02, with 95% CI from 0.98 to 1.06), and favorable associations between fidaxomicin and reduced risk of rCDI (RR 0.59, with 95% CI from 0.47 to 0.75) and improved global cure (RR 1.18, with 95% CI from 1.09 to 1.26) [87].
The results of the EXTEND RCT were published in 2018 [88]. EXTEND was an open-label phase-3b/4 RCT comparing extended-pulsed fidaxomicin (administered orally at 200 mg twice daily on days 1–5, and then only once daily on alternate days from day 7 to day 25) vs. vancomycin (at the standard dosage of 125 mg four times daily for 10 days) in inpatients aged 60 years or older. The primary endpoint was sustained clinical cure at 30 days after the end of treatment in the modified FAS population (all randomized patients who received at least one dose of the study drug), and was achieved in 70% (124/177) and 59% (106/179) of patients in extended-pulsed fidaxomicin and vancomycin arms, respectively (95% CI for the difference from 1.0% to 20.7%), thereby demonstrating superiority (a fact which is in line with the enhanced suppression of C. difficile by a pulsed fidaxomicin regimen in preclinical studies [89]; although, the limitations of the lack of comparison vs. the standard fidaxomicin dosage and an extended-pulsed vancomycin regimen were acknowledged in the EXTEND study). With regard to rCDI (one of the study’s secondary endpoints), lower rates of recurrences were registered in the extended-pulsed fidaxomicin arm than in the vancomycin arm at day 40 (3/124 (2.4%) vs. 22/125 (17.6%)), day55 (7/124 (5.6%) vs. 23/125 (18.4%)), and day 90 (11/124 (8.8%) vs. 23/125 (18.4%)) [88]. Of note, pharmacokinetic/pharmacodynamic data from patients enrolled in EXTEND revealed that fidaxomicin concentrations in stools were above the MIC90 of C. difficile isolates (inferred from in vitro studies) until day 26 ± 1 [90]. The subgroup analyses of the EXTEND study showed higher clinical cure rates in the extended-pulsed fidaxomicin arm independent of age, prior CDI, infection with PCR-ribotype 027, CDI severity, or presence of cancer [91]. A post hoc analysis of the EXTEND study conducted after testing stools of enrolled patients at screening, also with the BioFire FilmArray Gastrointestinal Panel (BioMérieux, Basingstoke, UK), suggested that co-infection with other pathogens could possibly explain clinical failures [92]. In a meta-analysis pooling data from five of the RCTs discussed above plus the EXTEND study, fidaxomicin was associated with improved sustained symptomatic cure compared to vancomycin (OR 0.67, with 95% CI from 0.55 to 0.82) [93].
In hematopoietic stem cell transplantation (HSCT) recipients, the development of CDI is more frequent than in the general population of hospitalized patients, and it has been associated with an increased risk of bloodstream infections, new-onset graft versus host disease, and non-relapse mortality [94,95,96,97]. In the double-blind DEFLECT-1 RCT, fidaxomicin was compared to a placebo for the prophylaxis of CDI in HSCT recipients (either allo-HSCT or auto-HSCT) undergoing fluoroquinolone prophylaxis [98]. The primary composite endpoint, assessed in the mITT population (subjects receiving at least one dose of study drug/placebo) was prophylaxis failure, defined as confirmed CDI, receipt of anti-C. difficile drugs for any indication, or missed assessment of CDI for any reason. Fidaxomicin was administered at the dosage of 200 mg daily, starting from 2 days after conditioning or initiation of prophylaxis with fluoroquinolones, and continuing until 7 days after neutrophil engraftment or completion of prophylaxis with fluoroquinolones/treatment with other antimicrobials, for up to 40 days. Prophylaxis failure was similar in patients receiving fidaxomicin and in patients receiving placebo (28.6% (86/301) vs. 30.8% (92/299), respectively, with 95% CI for the difference from −5.1% to 9.5%); although, it is of note that most failures occurred because of non-CDI events and confirmed CDI was less frequent in the fidaxomicin arm than in the placebo arm in a sensitivity analysis (4.3% (13/301) vs. 10.7% (32/299), with 95% CI for the difference from 2.2% to 10.6%) [98].
Among currently ongoing RCT comparing fidaxomicin vs. other treatments for CDI is OpTION, a double-blind study that is being conducted in patients with rCDI and is comparing the efficacy of three different treatment regimens: (i) 200 mg of fidaxomicin twice daily, for 10 days; (ii) 125 mg of vancomycin four times daily, for 10 days; and (iii) 125 mg of vancomycin four times daily, for 10 days, followed by a taper/pulse regimen of vancomycin for 3 weeks [99]. Other ongoing phase-3/4 RCTs are comparing fidaxomicin vs. FMT in patients with rCDI (NCT05266807, NCT05201079). The results of an open-label RCT comparing standard dosage fidaxomicin vs. standard dosage vancomycin in patients with CDI receiving concurrent antibiotics for other infections have been recently released, with the primary endpoint of clinical cure having been registered in 73% (54/74) and 62.9% (44/70) of patients in the fidaxomicin and vancomycin arms, respectively [100]. Among secondary endpoints, rCDI developed in 3.3% (2/60) and 4.0% (2/50) of patients in the fidaxomicin and vancomycin arms, respectively [100].
In RCTs, fidaxomicin was overall well tolerated. In the OPT-80-003 and OPT-80-004 studies, its safety profile was similar to oral vancomycin, and there were no differences between the two drugs in the frequency of death or serious adverse events [101]. The only numerical imbalances in these studies were related to gastrointestinal hemorrhage (4.1% vs. 3.1% in fidaxomicin-treated and vancomycin-treated patients, respectively) and leukopenia (4.1% vs. 1.7% in fidaxomicin-treated and vancomycin-treated patients, respectively); although, there was no evidence of a causal relationship between fidaxomicin administration and the occurrence of these events [101]. Similar tolerability profiles of fidaxomicin and vancomycin were observed in the phase-3 study conducted in Japan [83]. The incidence of treatment-emergent adverse events was similar in the extended-pulsed fidaxomicin arm and in the vancomycin arm in the EXTEND study [88]. One death in the vancomycin arm was deemed as being related to the study drug by the investigators [88]. The registered drug-related adverse events were 15% and 20% in the fidaxomicin and placebo arms in the DEFLECT-1 RCT [98].

5. Conclusions

In both the recently updated IDSA/SHEA guidelines and the updated ESCMID guidance document, fidaxomicin is preferentially recommended as first-line treatment over vancomycin both for the first episode of CDI and for rCDI (see Table 2 for more details) [11,12]. Although vancomycin remains a suitable alternative to fidaxomicin (noninferiority was indeed the rule in phase-3 RCTs with regard to the primary endpoint of clinical response), for shaping these recommendations particular attention was devoted to the improved global cure and reduced risk of rCDI observed with fidaxomicin compared to vancomycin in RCTs. The overall scenario is thus shifting from “administer vancomycin first, because of reduced cost and similar efficacy” to “consider fidaxomicin first, in view of the global benefits for the patient, if feasible”. With regard to feasibility, fidaxomicin still remains more costly than vancomycin, and, while the major driver of choice should solidly remain the global benefit for the patient, consideration of available resources should also be necessarily weighed in the balance. Against this background, a clear mistake would be that of continuing to administer vancomycin for any first CDI episode only because of reduced costs, thereby ignoring the evidence arising from RCTs about the improved global benefits following fidaxomicin treatment. Rather, risk models for rCDI should be used for selecting patients to preferentially receive fidaxomicin (i.e., to clearly identify those patients for whom fidaxomicin-driven global benefits are relevant). In our opinion, precisely stratifying risk groups for rCDI will represent a crucial research trajectory of future real-life studies on the treatment of initial CDI episodes. In addition, after reviewing the results of existing RCTs summarized in the previous sections, we also consider some other remaining grey areas as relevant fields for current and future research: (i) the exact positioning of the extended-pulsed fidaxomicin regimen, and its comparative efficacy with an extended-pulsed vancomycin regimen; (ii) the comparative efficacy of fidaxomicin vs. vancomycin in severe and severe-complicated CDI; (iii) the efficacy of fidaxomicin plus bezlotoxumab in preventing rCDI in comparison to other bezlotoxumab-including regimens; and (iv) when to precisely consider FMT instead of treatment with oral drugs, including fidaxomicin. Elucidating all of these remaining areas could further optimize the current positioning of fidaxomicin within CDI and rCDI treatment algorithms and, in turn, patients’ health.

Author Contributions

Conceptualization, D.R.G., A.V. and M.B.; methodology, D.R.G.; writing—original draft preparation, D.R.G.; writing—review and editing, D.R.G., A.V., M.F., F.M. and M.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Conflicts of Interest

Outside the submitted work, Daniele Roberto Giacobbe reports investigator-initiated grants from Pfizer, Shionogi, and Gilead Italia, and speaker fees and/or advisory board fees from Pfizer and Tillotts Pharma. Outside the submitted work, Matteo Bassetti reports research grants and/or personal fees for advisor/consultant and/or speaker/chairman from Bayer, BioMérieux, Cidara, Cipla, Gilead, Menarini, MSD, Pfizer, and Shionogi. The other authors have no conflict of interest to disclose.

References

  1. Guh, A.Y.; Mu, Y.; Winston, L.G.; Johnston, H.; Olson, D.; Farley, M.M.; Wilson, L.E.; Holzbauer, S.M.; Phipps, E.C.; Dumyati, G.K.; et al. Trends in U.S. Burden of Clostridioides difficile Infection and Outcomes. N. Engl. J. Med. 2020, 382, 1320–1330. [Google Scholar] [CrossRef] [PubMed]
  2. Khanna, S. My Treatment Approach to Clostridioides difficile Infection. Mayo Clin. Proc. 2021, 96, 2192–2204. [Google Scholar] [CrossRef] [PubMed]
  3. Khanna, S.; Pardi, D.S.; Aronson, S.L.; Kammer, P.P.; Orenstein, R.; St Sauver, J.L.; Harmsen, W.S.; Zinsmeister, A.R. The epidemiology of community-acquired Clostridium difficile infection: A population-based study. Am. J. Gastroenterol. 2012, 107, 89–95. [Google Scholar] [CrossRef] [Green Version]
  4. Lessa, F.C. Community-associated Clostridium difficile infection: How real is it? Anaerobe 2013, 24, 121–123. [Google Scholar] [CrossRef]
  5. Alicino, C.; Giacobbe, D.R.; Durando, P.; Bellina, D.; AM, D.I.B.; Paganino, C.; Del Bono, V.; Viscoli, C.; Icardi, G.; Orsi, A. Increasing incidence of Clostridium difficile infections: Results from a 5-year retrospective study in a large teaching hospital in the Italian region with the oldest population. Epidemiol. Infect. 2016, 144, 2517–2526. [Google Scholar] [CrossRef] [Green Version]
  6. Finn, E.; Andersson, F.L.; Madin-Warburton, M. Burden of Clostridioides difficile infection (CDI)—A systematic review of the epidemiology of primary and recurrent CDI. BMC Infect. Dis. 2021, 21, 456. [Google Scholar] [CrossRef]
  7. Fekety, R.; McFarland, L.V.; Surawicz, C.M.; Greenberg, R.N.; Elmer, G.W.; Mulligan, M.E. Recurrent Clostridium difficile diarrhea: Characteristics of and risk factors for patients enrolled in a prospective, randomized, double-blinded trial. Clin. Infect. Dis. 1997, 24, 324–333. [Google Scholar] [CrossRef] [PubMed]
  8. Granata, G.; Petrosillo, N.; Adamoli, L.; Bartoletti, M.; Bartoloni, A.; Basile, G.; Bassetti, M.; Bonfanti, P.; Borromeo, R.; Ceccarelli, G.; et al. Prospective Study on Incidence, Risk Factors and Outcome of Recurrent Clostridioides difficile Infections. J. Clin. Med. 2021, 10, 1127. [Google Scholar] [CrossRef]
  9. McFarland, L.V.; Surawicz, C.M.; Rubin, M.; Fekety, R.; Elmer, G.W.; Greenberg, R.N. Recurrent Clostridium difficile disease: Epidemiology and clinical characteristics. Infect. Control Hosp. Epidemiol. 1999, 20, 43–50. [Google Scholar] [CrossRef]
  10. Falcone, M.; Tiseo, G.; Iraci, F.; Raponi, G.; Goldoni, P.; Delle Rose, D.; Santino, I.; Carfagna, P.; Murri, R.; Fantoni, M.; et al. Risk factors for recurrence in patients with Clostridium difficile infection due to 027 and non-027 ribotypes. Clin. Microbiol. Infect. 2019, 25, 474–480. [Google Scholar] [CrossRef]
  11. Johnson, S.; Lavergne, V.; Skinner, A.M.; Gonzales-Luna, A.J.; Garey, K.W.; Kelly, C.P.; Wilcox, M.H. 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, 73, 755–757. [Google Scholar] [CrossRef] [PubMed]
  12. van Prehn, J.; Reigadas, E.; Vogelzang, E.H.; Bouza, E.; Hristea, A.; Guery, B.; Krutova, M.; Noren, T.; Allerberger, F.; Coia, J.E.; et al. European Society of Clinical Microbiology and Infectious Diseases: 2021 update on the treatment guidance document for Clostridioides difficile infection in adults. Clin. Microbiol. Infect. 2021, 27 (Suppl. S2), S1–S21. [Google Scholar] [CrossRef] [PubMed]
  13. Babakhani, F.; Gomez, A.; Robert, N.; Sears, P. Killing kinetics of fidaxomicin and its major metabolite, OP-1118, against Clostridium difficile. J. Med. Microbiol. 2011, 60, 1213–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Cornely, O.A. Current and emerging management options for Clostridium difficile infection: What is the role of fidaxomicin? Clin. Microbiol. Infect. 2012, 18 (Suppl. S6), 28–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Louie, T.J.; Emery, J.; Krulicki, W.; Byrne, B.; Mah, M. OPT-80 eliminates Clostridium difficile and is sparing of bacteroides species during treatment of C. difficile infection. Antimicrob. Agents Chemother. 2009, 53, 261–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Tannock, G.W.; Munro, K.; Taylor, C.; Lawley, B.; Young, W.; Byrne, B.; Emery, J.; Louie, T. A new macrocyclic antibiotic, fidaxomicin (OPT-80), causes less alteration to the bowel microbiota of Clostridium difficile-infected patients than does vancomycin. Microbiology 2010, 156, 3354–3359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Artsimovitch, I.; Seddon, J.; Sears, P. Fidaxomicin is an inhibitor of the initiation of bacterial RNA synthesis. Clin. Infect. Dis. 2012, 55 (Suppl. S2), S127–S131. [Google Scholar] [CrossRef] [Green Version]
  18. Cao, X.; Boyaci, H.; Chen, J.; Bao, Y.; Landick, R.; Campbell, E.A. Basis of narrow-spectrum activity of fidaxomicin on Clostridioides difficile. Nature 2022, 604, 541–545. [Google Scholar] [CrossRef]
  19. How the antibiotic fidaxomicin targets an intestinal pathogen. Nature 2022. online ahead of print. [CrossRef]
  20. Ajami, N.J.; Cope, J.L.; Wong, M.C.; Petrosino, J.F.; Chesnel, L. Impact of Oral Fidaxomicin Administration on the Intestinal Microbiota and Susceptibility to Clostridium difficile Colonization in Mice. Antimicrob. Agents Chemother. 2018, 62, e02112-17. [Google Scholar] [CrossRef]
  21. Yamaguchi, T.; Konishi, H.; Aoki, K.; Ishii, Y.; Chono, K.; Tateda, K. The gut microbiome diversity of Clostridioides difficile-inoculated mice treated with vancomycin and fidaxomicin. J. Infect. Chemother. 2020, 26, 483–491. [Google Scholar] [CrossRef] [PubMed]
  22. Deshpande, A.; Hurless, K.; Cadnum, J.L.; Chesnel, L.; Gao, L.; Chan, L.; Kundrapu, S.; Polinkovsky, A.; Donskey, C.J. Effect of Fidaxomicin versus Vancomycin on Susceptibility to Intestinal Colonization with Vancomycin-Resistant Enterococci and Klebsiella pneumoniae in Mice. Antimicrob. Agents Chemother. 2016, 60, 3988–3993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Louie, T.J.; Cannon, K.; Byrne, B.; Emery, J.; Ward, L.; Eyben, M.; Krulicki, W. Fidaxomicin preserves the intestinal microbiome during and after treatment of Clostridium difficile infection (CDI) and reduces both toxin reexpression and recurrence of CDI. Clin. Infect. Dis. 2012, 55 (Suppl. S2), S132–S142. [Google Scholar] [CrossRef] [PubMed]
  24. Biedenbach, D.J.; Ross, J.E.; Putnam, S.D.; Jones, R.N. In vitro activity of fidaxomicin (OPT-80) tested against contemporary clinical isolates of Staphylococcus spp. and Enterococcus spp. Antimicrob. Agents Chemother. 2010, 54, 2273–2275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Nerandzic, M.M.; Mullane, K.; Miller, M.A.; Babakhani, F.; Donskey, C.J. Reduced acquisition and overgrowth of vancomycin-resistant enterococci and Candida species in patients treated with fidaxomicin versus vancomycin for Clostridium difficile infection. Clin. Infect. Dis. 2012, 55 (Suppl. S2), S121–S126. [Google Scholar] [CrossRef]
  26. Falcone, M.; Russo, A.; Iraci, F.; Carfagna, P.; Goldoni, P.; Vullo, V.; Venditti, M. Risk Factors and Outcomes for Bloodstream Infections Secondary to Clostridium difficile Infection. Antimicrob. Agents Chemother. 2016, 60, 252–257. [Google Scholar] [CrossRef] [Green Version]
  27. Babakhani, F.; Gomez, A.; Robert, N.; Sears, P. Postantibiotic effect of fidaxomicin and its major metabolite, OP-1118, against Clostridium difficile. Antimicrob. Agents Chemother. 2011, 55, 4427–4429. [Google Scholar] [CrossRef] [Green Version]
  28. Sears, P.; Crook, D.W.; Louie, T.J.; Miller, M.A.; Weiss, K. Fidaxomicin attains high fecal concentrations with minimal plasma concentrations following oral administration in patients with Clostridium difficile infection. Clin. Infect. Dis. 2012, 55 (Suppl. S2), S116–S120. [Google Scholar] [CrossRef]
  29. Soriano, M.M.; Liao, S.; Danziger, L.H. Fidaxomicin: A minimally absorbed macrocyclic antibiotic for the treatment of Clostridium difficile infections. Expert Rev. Anti-Infect. Ther. 2013, 11, 767–776. [Google Scholar] [CrossRef]
  30. Mullane, K.M.; Gorbach, S. Fidaxomicin: First-in-class macrocyclic antibiotic. Expert Rev. Anti-Infect. Ther. 2011, 9, 767–777. [Google Scholar] [CrossRef]
  31. Shue, Y.K.; Sears, P.S.; Shangle, S.; Walsh, R.B.; Lee, C.; Gorbach, S.L.; Okumu, F.; Preston, R.A. Safety, tolerance, and pharmacokinetic studies of OPT-80 in healthy volunteers following single and multiple oral doses. Antimicrob. Agents Chemother. 2008, 52, 1391–1395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Zhanel, G.G.; Walkty, A.J.; Karlowsky, J.A. Fidaxomicin: A novel agent for the treatment of Clostridium difficile infection. Can. J. Infect. Dis. Med. Microbiol. 2015, 26, 305–312. [Google Scholar] [CrossRef] [Green Version]
  33. Goldstein, E.J.; Babakhani, F.; Citron, D.M. Antimicrobial activities of fidaxomicin. Clin. Infect. Dis. 2012, 55 (Suppl. S2), S143–S148. [Google Scholar] [CrossRef] [Green Version]
  34. Liao, C.H.; Ko, W.C.; Lu, J.J.; Hsueh, P.R. Characterizations of clinical isolates of Clostridium difficile by toxin genotypes and by susceptibility to 12 antimicrobial agents, including fidaxomicin (OPT-80) and rifaximin: A multicenter study in Taiwan. Antimicrob. Agents Chemother. 2012, 56, 3943–3949. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Eitel, Z.; Terhes, G.; Soki, J.; Nagy, E.; Urban, E. Investigation of the MICs of fidaxomicin and other antibiotics against Hungarian Clostridium difficile isolates. Anaerobe 2015, 31, 47–49. [Google Scholar] [CrossRef] [PubMed]
  36. Snydman, D.R.; McDermott, L.A.; Jacobus, N.V.; Thorpe, C.; Stone, S.; Jenkins, S.G.; Goldstein, E.J.; Patel, R.; Forbes, B.A.; Mirrett, S.; et al. U.S.-Based National Sentinel Surveillance Study for the Epidemiology of Clostridium difficile-Associated Diarrheal Isolates and Their Susceptibility to Fidaxomicin. Antimicrob. Agents Chemother. 2015, 59, 6437–6443. [Google Scholar] [CrossRef] [Green Version]
  37. Thorpe, C.M.; McDermott, L.A.; Tran, M.K.; Chang, J.; Jenkins, S.G.; Goldstein, E.J.C.; Patel, R.; Forbes, B.A.; Johnson, S.; Gerding, D.N.; et al. U.S.-Based National Surveillance for Fidaxomicin Susceptibility of Clostridioides difficile-Associated Diarrheal Isolates from 2013 to 2016. Antimicrob. Agents Chemother. 2019, 63, e00391-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Yamagishi, Y.; Nishiyama, N.; Koizumi, Y.; Matsukawa, Y.; Suematsu, H.; Hagihara, M.; Katsumata, K.; Mikamo, H. Antimicrobial activity of fidaxomicin against Clostridium difficile clinical isolates in Aichi area in Japan. J. Infect. Chemother. 2017, 23, 724–726. [Google Scholar] [CrossRef] [PubMed]
  39. Putsathit, P.; Maneerattanaporn, M.; Piewngam, P.; Knight, D.R.; Kiratisin, P.; Riley, T.V. Antimicrobial susceptibility of Clostridium difficile isolated in Thailand. Antimicrob. Resist. Infect. Control. 2017, 6, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Cheng, J.W.; Yang, Q.W.; Xiao, M.; Yu, S.Y.; Zhou, M.L.; Kudinha, T.; Kong, F.; Liao, J.W.; Xu, Y.C. High in vitro activity of fidaxomicin against Clostridium difficile isolates from a university teaching hospital in China. J. Microbiol. Immunol. Infect. 2018, 51, 411–416. [Google Scholar] [CrossRef]
  41. Wolfe, C.; Pagano, P.; Pillar, C.M.; Shinabarger, D.L.; Boulos, R.A. Comparison of the in vitro antibacterial activity of Ramizol, fidaxomicin, vancomycin, and metronidazole against 100 clinical isolates of Clostridium difficile by broth microdilution. Diagn. Microbiol. Infect. Dis. 2018, 92, 250–252. [Google Scholar] [CrossRef] [Green Version]
  42. Beran, V.; Kuijper, E.J.; Harmanus, C.; Sanders, I.M.; van Dorp, S.M.; Knetsch, C.W.; Janeckova, J.; Seidelova, A.; Barekova, L.; Tvrdik, J.; et al. Molecular typing and antimicrobial susceptibility testing to six antimicrobials of Clostridium difficile isolates from three Czech hospitals in Eastern Bohemia in 2011-2012. Folia Microbiol. 2017, 62, 445–451. [Google Scholar] [CrossRef]
  43. Karlowsky, J.A.; Adam, H.J.; Kosowan, T.; Baxter, M.R.; Nichol, K.A.; Laing, N.M.; Golding, G.; Zhanel, G.G. PCR ribotyping and antimicrobial susceptibility testing of isolates of Clostridium difficile cultured from toxin-positive diarrheal stools of patients receiving medical care in Canadian hospitals: The Canadian Clostridium difficile Surveillance Study (CAN-DIFF) 2013-2015. Diagn. Microbiol. Infect. Dis. 2018, 91, 105–111. [Google Scholar] [CrossRef]
  44. Freeman, J.; Vernon, J.; Morris, K.; Nicholson, S.; Todhunter, S.; Longshaw, C.; Wilcox, M.H.; Pan-European Longitudinal Surveillance of Antibiotic Resistance among Prevalent Clostridium difficile Ribotypes’ Study Group. Pan-European longitudinal surveillance of antibiotic resistance among prevalent Clostridium difficile ribotypes. Clin. Microbiol. Infect. 2015, 21, 248.E9–248.E16. [Google Scholar] [CrossRef] [Green Version]
  45. Freeman, J.; Vernon, J.; Pilling, S.; Morris, K.; Nicholson, S.; Shearman, S.; Longshaw, C.; Wilcox, M.H.; Pan-European Longitudinal Surveillance of Antibiotic Resistance among Prevalent Clostridium difficile Ribotypes Study Group. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Freeman, J.; Vernon, J.; Pilling, S.; Morris, K.; Nicolson, S.; Shearman, S.; Clark, E.; Palacios-Fabrega, J.A.; Wilcox, M.; Pan-European Longitudinal Surveillance of Antibiotic Resistance among Prevalent Clostridium difficile Ribotypes’ Study Group. Five-year Pan-European, longitudinal surveillance of Clostridium difficile ribotype prevalence and antimicrobial resistance: The extended ClosER study. Eur. J. Clin. Microbiol. Infect. Dis. 2020, 39, 169–177. [Google Scholar] [CrossRef] [Green Version]
  47. Citron, D.M.; Babakhani, F.; Goldstein, E.J.; Nagaro, K.; Sambol, S.; Sears, P.; Shue, Y.K.; Gerding, D.N. Typing and susceptibility of bacterial isolates from the fidaxomicin (OPT-80) phase II study for C. difficile infection. Anaerobe 2009, 15, 234–236. [Google Scholar] [CrossRef]
  48. Goldstein, E.J.; Citron, D.M.; Sears, P.; Babakhani, F.; Sambol, S.P.; Gerding, D.N. Comparative susceptibilities to fidaxomicin (OPT-80) of isolates collected at baseline, recurrence, and failure from patients in two phase III trials of fidaxomicin against Clostridium difficile infection. Antimicrob. Agents Chemother. 2011, 55, 5194–5199. [Google Scholar] [CrossRef] [Green Version]
  49. Peng, Z.; Jin, D.; Kim, H.B.; Stratton, C.W.; Wu, B.; Tang, Y.W.; Sun, X. Update on Antimicrobial Resistance in Clostridium difficile: Resistance Mechanisms and Antimicrobial Susceptibility Testing. J. Clin. Microbiol. 2017, 55, 1998–2008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Leeds, J.A.; Sachdeva, M.; Mullin, S.; Barnes, S.W.; Ruzin, A. In vitro selection, via serial passage, of Clostridium difficile mutants with reduced susceptibility to fidaxomicin or vancomycin. J. Antimicrob. Chemother. 2014, 69, 41–44. [Google Scholar] [CrossRef] [PubMed]
  51. Leeds, J.A. Antibacterials Developed to Target a Single Organism: Mechanisms and Frequencies of Reduced Susceptibility to the Novel Anti-Clostridium difficile Compounds Fidaxomicin and LFF571. Cold Spring Harb. Perspect. Med. 2016, 6, a025445. [Google Scholar] [CrossRef] [Green Version]
  52. Kuehne, S.A.; Dempster, A.W.; Collery, M.M.; Joshi, N.; Jowett, J.; Kelly, M.L.; Cave, R.; Longshaw, C.M.; Minton, N.P. Characterization of the impact of rpoB mutations on the in vitro and in vivo competitive fitness of Clostridium difficile and susceptibility to fidaxomicin. J. Antimicrob. Chemother. 2018, 73, 973–980. [Google Scholar] [CrossRef] [Green Version]
  53. Schwanbeck, J.; Riedel, T.; Laukien, F.; Schober, I.; Oehmig, I.; Zimmermann, O.; Overmann, J.; Gross, U.; Zautner, A.E.; Bohne, W. Characterization of a clinical Clostridioides difficile isolate with markedly reduced fidaxomicin susceptibility and a V1143D mutation in rpoB. J. Antimicrob. Chemother. 2019, 74, 6–10. [Google Scholar] [CrossRef] [PubMed]
  54. Babakhani, F.; Bouillaut, L.; Gomez, A.; Sears, P.; Nguyen, L.; Sonenshein, A.L. Fidaxomicin inhibits spore production in Clostridium difficile. Clin. Infect. Dis. 2012, 55 (Suppl. S2), S162–S169. [Google Scholar] [CrossRef] [Green Version]
  55. Chilton, C.H.; Crowther, G.S.; Freeman, J.; Todhunter, S.L.; Nicholson, S.; Longshaw, C.M.; Wilcox, M.H. Successful treatment of simulated Clostridium difficile infection in a human gut model by fidaxomicin first line and after vancomycin or metronidazole failure. J. Antimicrob. Chemother. 2014, 69, 451–462. [Google Scholar] [CrossRef] [Green Version]
  56. Aldape, M.J.; Packham, A.E.; Heeney, D.D.; Rice, S.N.; Bryant, A.E.; Stevens, D.L. Fidaxomicin reduces early toxin A and B production and sporulation in Clostridium difficile in vitro. J. Med. Microbiol. 2017, 66, 1393–1399. [Google Scholar] [CrossRef]
  57. Allen, C.A.; Babakhani, F.; Sears, P.; Nguyen, L.; Sorg, J.A. Both fidaxomicin and vancomycin inhibit outgrowth of Clostridium difficile spores. Antimicrob. Agents Chemother. 2013, 57, 664–667. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Chilton, C.H.; Crowther, G.S.; Ashwin, H.; Longshaw, C.M.; Wilcox, M.H. Association of Fidaxomicin with C. difficile Spores: Effects of Persistence on Subsequent Spore Recovery, Outgrowth and Toxin Production. PLoS ONE 2016, 11, e0161200. [Google Scholar] [CrossRef] [Green Version]
  59. Basseres, E.; Endres, B.T.; Montes-Bravo, N.; Perez-Soto, N.; Rashid, T.; Lancaster, C.; Begum, K.; Alam, M.J.; Paredes-Sabja, D.; Garey, K.W. Visualization of fidaxomicin association with the exosporium layer of Clostridioides difficile spores. Anaerobe 2021, 69, 102352. [Google Scholar] [CrossRef] [PubMed]
  60. Babakhani, F.; Bouillaut, L.; Sears, P.; Sims, C.; Gomez, A.; Sonenshein, A.L. Fidaxomicin inhibits toxin production in Clostridium difficile. J. Antimicrob. Chemother. 2013, 68, 515–522. [Google Scholar] [CrossRef] [PubMed]
  61. Thabit, A.K.; Alam, M.J.; Khaleduzzaman, M.; Garey, K.W.; Nicolau, D.P. A pilot study to assess bacterial and toxin reduction in patients with Clostridium difficile infection given fidaxomicin or vancomycin. Ann. Clin. Microbiol. Antimicrob. 2016, 15, 22. [Google Scholar] [CrossRef] [Green Version]
  62. Koon, H.W.; Wang, J.; Mussatto, C.C.; Ortiz, C.; Lee, E.C.; Tran, D.H.; Chen, X.; Kelly, C.P.; Pothoulakis, C. Fidaxomicin and OP-1118 Inhibit Clostridium difficile Toxin A- and B-Mediated Inflammatory Responses via Inhibition of NF-kappaB Activity. Antimicrob. Agents Chemother. 2018, 62, e01513-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Koon, H.W.; Ho, S.; Hing, T.C.; Cheng, M.; Chen, X.; Ichikawa, Y.; Kelly, C.P.; Pothoulakis, C. Fidaxomicin inhibits Clostridium difficile toxin A-mediated enteritis in the mouse ileum. Antimicrob. Agents Chemother. 2014, 58, 4642–4650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Dapa, T.; Unnikrishnan, M. Biofilm formation by Clostridium difficile. Gut Microbes 2013, 4, 397–402. [Google Scholar] [CrossRef] [Green Version]
  65. Hamada, M.; Yamaguchi, T.; Ishii, Y.; Chono, K.; Tateda, K. Inhibitory effect of fidaxomicin on biofilm formation in Clostridioides difficile. J. Infect. Chemother. 2020, 26, 685–692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. James, G.A.; Chesnel, L.; Boegli, L.; deLancey Pulcini, E.; Fisher, S.; Stewart, P.S. Analysis of Clostridium difficile biofilms: Imaging and antimicrobial treatment. J. Antimicrob. Chemother. 2018, 73, 102–108. [Google Scholar] [CrossRef] [PubMed]
  67. Pantaleon, V.; Bouttier, S.; Soavelomandroso, A.P.; Janoir, C.; Candela, T. Biofilms of Clostridium species. Anaerobe 2014, 30, 193–198. [Google Scholar] [CrossRef] [PubMed]
  68. Biswas, J.S.; Patel, A.; Otter, J.A.; Wade, P.; Newsholme, W.; van Kleef, E.; Goldenberg, S.D. Reduction in Clostridium difficile environmental contamination by hospitalized patients treated with fidaxomicin. J. Hosp. Infect. 2015, 90, 267–270. [Google Scholar] [CrossRef] [Green Version]
  69. Davies, K.; Mawer, D.; Walker, A.S.; Berry, C.; Planche, T.; Stanley, P.; Goldenberg, S.; Sandoe, J.; Wilcox, M.H. An Analysis of Clostridium difficile Environmental Contamination During and After Treatment for C difficile Infection. Open Forum Infect. Dis. 2020, 7, ofaa362. [Google Scholar] [CrossRef]
  70. Turner, N.A.; Warren, B.G.; Gergen-Teague, M.F.; Addison, R.M.; Addison, B.; Rutala, W.A.; Weber, D.J.; Sexton, D.J.; Anderson, D.J. Impact of Oral Metronidazole, Vancomycin, and Fidaxomicin on Host Shedding and Environmental Contamination with Clostridioides difficile. Clin. Infect. Dis. 2022, 74, 648–656. [Google Scholar] [CrossRef]
  71. Cataldo, M.A.; Granata, G.; Petrosillo, N. Clostridium difficile infection: New approaches to prevention, non-antimicrobial treatment, and stewardship. Expert Rev. Anti-Infect. Ther. 2017, 15, 1027–1040. [Google Scholar] [CrossRef]
  72. Cornely, O.A.; Crook, D.W.; Esposito, R.; Poirier, A.; Somero, M.S.; Weiss, K.; Sears, P.; Gorbach, S.; Group, O.P.T.C.S. Fidaxomicin versus vancomycin for infection with Clostridium difficile in Europe, Canada, and the USA: A double-blind, non-inferiority, randomised controlled trial. Lancet Infect. Dis. 2012, 12, 281–289. [Google Scholar] [CrossRef]
  73. Louie, T.J.; Miller, M.A.; Mullane, K.M.; Weiss, K.; Lentnek, A.; Golan, Y.; Gorbach, S.; Sears, P.; Shue, Y.K.; OPT-80-003 Clinical Study Group. Fidaxomicin versus vancomycin for Clostridium difficile infection. N. Engl. J. Med. 2011, 364, 422–431. [Google Scholar] [CrossRef] [Green Version]
  74. Crook, D.W.; Walker, A.S.; Kean, Y.; Weiss, K.; Cornely, O.A.; Miller, M.A.; Esposito, R.; Louie, T.J.; Stoesser, N.E.; Young, B.C.; et al. Fidaxomicin versus vancomycin for Clostridium difficile infection: Meta-analysis of pivotal randomized controlled trials. Clin. Infect. Dis. 2012, 55 (Suppl. S2), S93–S103. [Google Scholar] [CrossRef]
  75. Cornely, O.A.; Nathwani, D.; Ivanescu, C.; Odufowora-Sita, O.; Retsa, P.; Odeyemi, I.A. Clinical efficacy of fidaxomicin compared with vancomycin and metronidazole in Clostridium difficile infections: A meta-analysis and indirect treatment comparison. J. Antimicrob. Chemother. 2014, 69, 2892–2900. [Google Scholar] [CrossRef] [Green Version]
  76. Nelson, R.L.; Suda, K.J.; Evans, C.T. Antibiotic treatment for Clostridium difficile-associated diarrhoea in adults. Cochrane Database Syst. Rev. 2017, 3, CD004610. [Google Scholar] [CrossRef]
  77. Mullane, K.M.; Miller, M.A.; Weiss, K.; Lentnek, A.; Golan, Y.; Sears, P.S.; Shue, Y.K.; Louie, T.J.; Gorbach, S.L. Efficacy of fidaxomicin versus vancomycin as therapy for Clostridium difficile infection in individuals taking concomitant antibiotics for other concurrent infections. Clin. Infect. Dis. 2011, 53, 440–447. [Google Scholar] [CrossRef]
  78. Cornely, O.A.; Miller, M.A.; Fantin, B.; Mullane, K.; Kean, Y.; Gorbach, S. Resolution of Clostridium difficile-associated diarrhea in patients with cancer treated with fidaxomicin or vancomycin. J. Clin. Oncol. 2013, 31, 2493–2499. [Google Scholar] [CrossRef]
  79. Figueroa, I.; Johnson, S.; Sambol, S.P.; Goldstein, E.J.; Citron, D.M.; Gerding, D.N. Relapse versus reinfection: Recurrent Clostridium difficile infection following treatment with fidaxomicin or vancomycin. Clin. Infect. Dis. 2012, 55 (Suppl. S2), S104–S109. [Google Scholar] [CrossRef]
  80. Eyre, D.W.; Babakhani, F.; Griffiths, D.; Seddon, J.; Del Ojo Elias, C.; Gorbach, S.L.; Peto, T.E.; Crook, D.W.; Walker, A.S. Whole-genome sequencing demonstrates that fidaxomicin is superior to vancomycin for preventing reinfection and relapse of infection with Clostridium difficile. J. Infect. Dis. 2014, 209, 1446–1451. [Google Scholar] [CrossRef]
  81. Cornely, O.A.; Miller, M.A.; Louie, T.J.; Crook, D.W.; Gorbach, S.L. Treatment of first recurrence of Clostridium difficile infection: Fidaxomicin versus vancomycin. Clin. Infect. Dis. 2012, 55 (Suppl. S2), S154–S161. [Google Scholar] [CrossRef] [PubMed]
  82. Louie, T.J.; Miller, M.A.; Crook, D.W.; Lentnek, A.; Bernard, L.; High, K.P.; Shue, Y.K.; Gorbach, S.L. Effect of age on treatment outcomes in Clostridium difficile infection. J. Am. Geriatr. Soc. 2013, 61, 222–230. [Google Scholar] [CrossRef] [PubMed]
  83. Mikamo, H.; Tateda, K.; Yanagihara, K.; Kusachi, S.; Takesue, Y.; Miki, T.; Oizumi, Y.; Gamo, K.; Hashimoto, A.; Toyoshima, J.; et al. Efficacy and safety of fidaxomicin for the treatment of Clostridioides (Clostridium) difficile infection in a randomized, double-blind, comparative Phase III study in Japan. J. Infect. Chemother. 2018, 24, 744–752. [Google Scholar] [CrossRef]
  84. Okumura, H.; Fukushima, A.; Taieb, V.; Shoji, S.; English, M. Fidaxomicin compared with vancomycin and metronidazole for the treatment of Clostridioides (Clostridium) difficile infection: A network meta-analysis. J. Infect. Chemother. 2020, 26, 43–50. [Google Scholar] [CrossRef]
  85. Housman, S.T.; Thabit, A.K.; Kuti, J.L.; Quintiliani, R.; Nicolau, D.P. Assessment of Clostridium difficile Burden in Patients Over Time with First Episode Infection Following Fidaxomicin or Vancomycin. Infect. Control Hosp. Epidemiol. 2016, 37, 215–218. [Google Scholar] [CrossRef] [PubMed]
  86. Hvas, C.L.; Dahl Jorgensen, S.M.; Jorgensen, S.P.; Storgaard, M.; Lemming, L.; Hansen, M.M.; Erikstrup, C.; Dahlerup, J.F. Fecal Microbiota Transplantation Is Superior to Fidaxomicin for Treatment of Recurrent Clostridium difficile Infection. Gastroenterology 2019, 156, 1324–1332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Tashiro, S.; Mihara, T.; Sasaki, M.; Shimamura, C.; Shimamura, R.; Suzuki, S.; Yoshikawa, M.; Hasegawa, T.; Enoki, Y.; Taguchi, K.; et al. Oral fidaxomicin versus vancomycin for the treatment of Clostridioides difficile infection: A systematic review and meta-analysis of randomized controlled trials. J. Infect. Chemother. 2022, 28, 1536–1545. [Google Scholar] [CrossRef]
  88. Guery, B.; Menichetti, F.; Anttila, V.J.; Adomakoh, N.; Aguado, J.M.; Bisnauthsing, K.; Georgopali, A.; Goldenberg, S.D.; Karas, A.; Kazeem, G.; et al. Extended-pulsed fidaxomicin versus vancomycin for Clostridium difficile infection in patients 60 years and older (EXTEND): A randomised, controlled, open-label, phase 3b/4 trial. Lancet Infect. Dis. 2018, 18, 296–307. [Google Scholar] [CrossRef] [Green Version]
  89. Chilton, C.H.; Crowther, G.S.; Todhunter, S.L.; Ashwin, H.; Longshaw, C.M.; Karas, A.; Wilcox, M.H. Efficacy of alternative fidaxomicin dosing regimens for treatment of simulated Clostridium difficile infection in an in vitro human gut model. J. Antimicrob. Chemother. 2015, 70, 2598–2607. [Google Scholar] [CrossRef] [Green Version]
  90. Guery, B.; Georgopali, A.; Karas, A.; Kazeem, G.; Michon, I.; Wilcox, M.H.; Cornely, O.A. Pharmacokinetic analysis of an extended-pulsed fidaxomicin regimen for the treatment of Clostridioides (Clostridium) difficile infection in patients aged 60 years and older in the EXTEND randomized controlled trial. J. Antimicrob. Chemother. 2020, 75, 1014–1018. [Google Scholar] [CrossRef]
  91. Cornely, O.A.; Vehreschild, M.; Adomakoh, N.; Georgopali, A.; Karas, A.; Kazeem, G.; Guery, B. Extended-pulsed fidaxomicin versus vancomycin for Clostridium difficile infection: EXTEND study subgroup analyses. Eur. J. Clin. Microbiol. Infect. Dis. 2019, 38, 1187–1194. [Google Scholar] [CrossRef] [Green Version]
  92. Wilcox, M.H.; Cornely, O.A.; Guery, B.; Longshaw, C.; Georgopali, A.; Karas, A.; Kazeem, G.; Palacios-Fabrega, J.A.; Vehreschild, M. Microbiological Characterization and Clinical Outcomes After Extended-Pulsed Fidaxomicin Treatment for Clostridioides difficile Infection in the EXTEND Study. Open Forum Infect. Dis. 2019, 6, ofz436. [Google Scholar] [CrossRef]
  93. Beinortas, T.; Burr, N.E.; Wilcox, M.H.; Subramanian, V. Comparative efficacy of treatments for Clostridium difficile infection: A systematic review and network meta-analysis. Lancet Infect. Dis. 2018, 18, 1035–1044. [Google Scholar] [CrossRef]
  94. Vehreschild, M.J.; Weitershagen, D.; Biehl, L.M.; Tacke, D.; Waldschmidt, D.; Tox, U.; Wisplinghoff, H.; Von Bergwelt-Baildon, M.; Cornely, O.A.; Vehreschild, J.J. Clostridium difficile infection in patients with acute myelogenous leukemia and in patients undergoing allogeneic stem cell transplantation: Epidemiology and risk factor analysis. Biol. Blood Marrow Transplant. 2014, 20, 823–828. [Google Scholar] [CrossRef] [Green Version]
  95. Trifilio, S.M.; Pi, J.; Mehta, J. Changing epidemiology of Clostridium difficile-associated disease during stem cell transplantation. Biol. Blood Marrow Transplant. 2013, 19, 405–409. [Google Scholar] [CrossRef] [Green Version]
  96. Dubberke, E.R.; Reske, K.A.; Srivastava, A.; Sadhu, J.; Gatti, R.; Young, R.M.; Rakes, L.C.; Dieckgraefe, B.; DiPersio, J.; Fraser, V.J. Clostridium difficile-associated disease in allogeneic hematopoietic stem-cell transplant recipients: Risk associations, protective associations, and outcomes. Clin. Transplant. 2010, 24, 192–198. [Google Scholar] [CrossRef] [Green Version]
  97. Alonso, C.D.; Treadway, S.B.; Hanna, D.B.; Huff, C.A.; Neofytos, D.; Carroll, K.C.; Marr, K.A. Epidemiology and outcomes of Clostridium difficile infections in hematopoietic stem cell transplant recipients. Clin. Infect. Dis. 2012, 54, 1053–1063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Mullane, K.M.; Winston, D.J.; Nooka, A.; Morris, M.I.; Stiff, P.; Dugan, M.J.; Holland, H.; Gregg, K.; Adachi, J.A.; Pergam, S.A.; et al. A Randomized, Placebo-controlled Trial of Fidaxomicin for Prophylaxis of Clostridium difficile-associated Diarrhea in Adults Undergoing Hematopoietic Stem Cell Transplantation. Clin. Infect. Dis. 2019, 68, 196–203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Johnson, S.; Gerding, D.N.; Li, X.; Reda, D.J.; Donskey, C.J.; Gupta, K.; Goetz, M.B.; Climo, M.W.; Gordin, F.M.; Ringer, R.; et al. Defining optimal treatment for recurrent Clostridioides difficile infection (OpTION study): A randomized, double-blind comparison of three antibiotic regimens for patients with a first or second recurrence. Contemp. Clin. Trials 2022, 116, 106756. [Google Scholar] [CrossRef] [PubMed]
  100. A Comparison of Fidaxomicin and Vancomycin in Patients with CDI Receiving Antibiotics for Concurrent. Available online: https://clinicaltrials.gov/ct2/show/study/NCT02692651 (accessed on 9 September 2022).
  101. Weiss, K.; Allgren, R.L.; Sellers, S. Safety analysis of fidaxomicin in comparison with oral vancomycin for Clostridium difficile infections. Clin. Infect. Dis. 2012, 55 (Suppl. S2), S110–S115. [Google Scholar] [CrossRef] [PubMed]
Table 1. Main efficacy data from phase-3/4 randomized controlled trials of fidaxomicin for the treatment of CDI in adult patients.
Table 1. Main efficacy data from phase-3/4 randomized controlled trials of fidaxomicin for the treatment of CDI in adult patients.
Author, YearFidaxomicinComparator/sStudy PopulationFrequency% Difference a (95% CI)
Study Name [Ref]Regimen(Dosage)Endpoint (Primary/Secondary)(Events/Treated)
(Dosage)
Louie et al., 2011
OTP-80-003 [73]
Standard regimen
(200 mg orally twice daily for 10 days)
Vancomycin
(125 mg orally four times daily for 10 days)
mITT populationb
Clinical cure (primary)
Fidaxomicin88.2% (253/287)2.4 (−3.1 d)
Vancomycin85.8% (265/309)Reference
rCDI (secondary)
Fidaxomicin15.4% (39/253)−9.9 (−16.6 to −2.9)
Vancomycin25.3% (67/265)Reference
Global cure (secondary)
Fidaxomicin74.6% (214/287)10.5 (3.1 to 17.7)
Vancomycin64.1% (198/309)Reference
Per-protocol populationc
Clinical cure (primary)
Fidaxomicin92.1% (244/265)2.3 (−2.6 d)
Vancomycin89.8% (254/283)Reference
rCDI (secondary)
Fidaxomicin13.3% (28/211)−10.7 (−17.9 to −3.3)
Vancomycin24.0% (53/221)Reference
Global cure (secondary)
Fidaxomicin77.7% (206/265)10.6 (3.1 to 17.9)
Vancomycin67.1% (190/283)Reference
Cornely et al., 2012
OTP-80-004 [72]
Standard regimen
(200 mg orally twice daily for 10 days)
Vancomycin
(125 mg orally four times daily for 10 days)
mITT populationb
Clinical cure (primary)
Fidaxomicin87.7% (221/252)0.9 (−4.9 d)
Vancomycin86.8% (223/257)Reference
rCDI (secondary)
Fidaxomicin12.7% (28/221)−14.2 (−21.4 to −6.8)
Vancomycin26.9% (60/223)Reference
Sustained response (secondary)
Fidaxomicin76.6% (193/252)13.2 (5.3 to 21.0)
Vancomycin63.4% (163/257)Reference
Per-protocol populationc
Clinical cure (primary)
Fidaxomicin91.7% (198/216)1.1 (−4.3 d)
Vancomycin90.6% (213/235)Reference
rCDI (secondary)
Fidaxomicin12.8% (23/180)−12.5 (−20.5 to −4.5)
Vancomycin25.3% (46/182)Reference
Global cure (secondary)
Fidaxomicin79.6% (172/216)14.1 (6.0 to 22.2)
Vancomycin65.5% (154/235)Reference
Mikamo et al., 2018
[83]
Standard regimen
(200 mg orally twice daily for 10 days)
Vancomycin
(125 mg orally four times daily for 10 days)
FAS population
Global cure (primary)
Fidaxomicin67.3% (70/104)1.2 (−11.3 to 13.7)
Vancomycin65.7% (71/108)Reference
FAS-R populatione
rCDI (secondary)
Fidaxomicin19.5% (17/87)−4.9 (−16.7 to 7.0)
Vancomycin25.3% (24/95)Reference
Housman et al., 2016
[85]
Standard regimen
(200 mg orally twice daily for 10 days)
Vancomycin
(125 mg orally four times daily for 10 days)
Patients with CDI
Reduction of spores (primary)f
Fidaxomicin66.7% (8/12)52.4 (NA)
Vancomycin14.3% (1/7)Reference
Hvas et al., 2019
[86]
Standard regimen
(200 mg orally twice daily for 10 days)
Vancomycin
(125 mg orally four times daily for 10 days) or FMT
Patients with rCDI
Clinical resolution (primary)g
Fidaxomicin33.3% (8/24)14.5% (NA)
FMT70.8% (17/24)52.0% NA)
Vancomycin18.8% (3/16)Reference
Guery et al., 2018
EXTEND
[88]
Extended-pulsed regimen (200 mg twice daily on days 1–5, and then only once daily on alternate days from day 7 to day 25)Vancomycin
(125 mg orally four times daily for 10 days)
Modified FAS populationh
Sustained clinical cure (primary)
Fidaxomicin70.1% (124/177)OR 1.62 (1.04 to 2.54)
Vancomycin59.2% (106/179)Reference
Per-protocol population
rCDI at day 40 (secondary)
Fidaxomicin2.4% (3/124)OR 0.12 (0.04 to 0.41)
Vancomycin17.6% (22/125)Reference
rCDI at day 55 (secondary)
Fidaxomicin5.6% (7/124)OR 0.31 (0.13 to 0.73)
Vancomycin18.4% (23/125)Reference
rCDI at day 90 (secondary)
Fidaxomicin8.8% (11/124)OR 0.49 (0.23 to 1.04)
Vancomycin18.4% (23/125)Reference
CDI, Clostridioides difficile infection; CI, confidence interval; FAS, full analysis set; FMT, fecal microbiota transplant; mITT, modified intention-to-treat; NA, not available; OR, odds ratio; rCDI, recurrent CDI. a Unless otherwise indicated. b Including patients with documented CDI who received at least one dose of study drug. c Including patients of the mITT population who received at least 3 days of treatment in the case of failure and at least 8 days of treatment in the case of clinical cure. d One-sided 97.5% CI. e FAS patients who achieved clinical cure during the index episode. f Defined as percentage of subjects achieving a reduction of at least 2 log10 colony-forming units (CFU)/g of spores in stools from screening to the end of therapy. g Defined as combination of clinical resolution and a negative toxin polymerase chain reaction at 8 weeks after allocation. h Including all randomized patients who received at least one dose of study drug.
Table 2. Current IDSA/SHEA and ESCMID recommendations regarding fidaxomicin for the treatment of CDI and rCDI.
Table 2. Current IDSA/SHEA and ESCMID recommendations regarding fidaxomicin for the treatment of CDI and rCDI.
Guidelines/Guidance DocumentRecommended Treatment for First CDI Episode *Recommended Treatment for rCDI *
ESCMID guidance document [12]
  • The use of a standard regimen of fidaxomicin is suggested over vancomycin
    (Strong recommendation, with moderate level of evidence)
  • Risk stratification should be considered when access to fidaxomicin is limited (e.g., older age >65 years plus one or more of the following: healthcare-associated CDI; hospitalization; in the previous 3 months, administration of concomitant antibiotics, initiation of PPIs during or after diagnosis of CDI; previous CDI episode)
    (Good practice statement)
  • When fidaxomicin is unavailable or unfeasible, vancomycin is a suitable alternative
    (Strong recommendation, with high level of evidence)
  • An extended-pulsed regiment of fidaxomicin could be considered in case of risk of rCDI, especially in old inpatients
    (Weak recommendation, with low level of evidence)
  • For severe or severe-complicated CDI, a standard regimen of fidaxomicin or vancomycin is suggested
    (good practice statement)
  • If the initial CDI episode was treated with metronidazole or vancomycin, the use of a standard regimen of fidaxomicin is preferentially recommended
    (Strong recommendation, with low level of evidence)
  • If the initial CDI episode was treated with fidaxomicin, considered bezlotoxumab in addition to fidaxomicin
    (Weak recommendation, with moderate level of evidence; “addition to fidaxomicin” as a good practice statement)
  • When fidaxomicin and bezlotoxumab are unavailable or unfeasible, consider a tapered/pulsed regimen of vancomycin
    (Weak recommendation, with very low level of evidence)
  • For multiple recurrences, FMT or bezlotoxumab in addition to standard of care is suggested
    (Weak recommendation, with moderate level of evidence for FMT and low level of evidence for bezlotoxumab)
IDSA/SHEA guidelines [11]
  • The use of a standard regimen of fidaxomicin is suggested over a standard course of vancomycin. A high value is placed on the beneficial effects and the safety of fidaxomicin, with implementations depending on available resources and with vancomycin remaining an acceptable alternative
    (Conditional recommendation with moderate certainty of evidence)
  • The use of a standard or extended-pulsed regimen of fidaxomicin is suggested over a standard regimen of vancomycin. For a first rCDI episode, vancomycin in a standard or tapered/pulsed regimen is an acceptable alternative. For multiple recurrences, possible options are fidaxomicin (standard or extended-pulsed regimen), vancomycin in a tapered/pulsed regimen, vancomycin followed by rifaximin, and FMT
    (Conditional recommendation with low certainty of evidence)
CDI, Clostridioides difficile infection; ESCMID, European Society of Clinical Microbiology and Infectious Diseases; FMT, fecal microbiota transplant; IDSA, Infectious Diseases Society of America; PPIs, proton pump inhibitors; rCDI, recurrent CDI; SHEA, Society for Healthcare Epidemiology of America. * For other recommendations about the use of other agents (e.g., bezlotoxumab) or FMT and not directly involving a decision about fidaxomicin please refer to the original guidelines/guidance documents [11,12]. For a fulminant CDI episode (hypotension or shock, ileus, or megacolon), IDSA/SHEA guidelines recommend oral/nasogastric tube vancomycin 500 mg four times daily plus intravenous metronidazole 500 mg thrice daily plus rectal instillation of vancomycin if ileus.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Giacobbe, D.R.; Vena, A.; Falcone, M.; Menichetti, F.; Bassetti, M. Fidaxomicin for the Treatment of Clostridioides difficile Infection in Adult Patients: An Update on Results from Randomized Controlled Trials. Antibiotics 2022, 11, 1365. https://doi.org/10.3390/antibiotics11101365

AMA Style

Giacobbe DR, Vena A, Falcone M, Menichetti F, Bassetti M. Fidaxomicin for the Treatment of Clostridioides difficile Infection in Adult Patients: An Update on Results from Randomized Controlled Trials. Antibiotics. 2022; 11(10):1365. https://doi.org/10.3390/antibiotics11101365

Chicago/Turabian Style

Giacobbe, Daniele Roberto, Antonio Vena, Marco Falcone, Francesco Menichetti, and Matteo Bassetti. 2022. "Fidaxomicin for the Treatment of Clostridioides difficile Infection in Adult Patients: An Update on Results from Randomized Controlled Trials" Antibiotics 11, no. 10: 1365. https://doi.org/10.3390/antibiotics11101365

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop