Open access
Antimicrobial Chemotherapy
Research Article
25 March 2024

Outcomes by Candida spp. in the ReSTORE Phase 3 trial of rezafungin versus caspofungin for candidemia and/or invasive candidiasis

ABSTRACT

Rezafungin is a long-acting, intravenously administered echinocandin for the treatment of candidemia and invasive candidiasis (IC). Non-inferiority of rezafungin vs caspofungin for the treatment of adults with candidemia and/or IC was demonstrated in the Phase 3 ReSTORE study based on the primary endpoints of day 14 global cure and 30-day all-cause mortality. Here, an analysis of ReSTORE data evaluating efficacy outcomes by baseline Candida species is described. Susceptibility testing was performed for Candida species using the Clinical and Laboratory Standards Institute reference broth microdilution method. There were 93 patients in the modified intent-to-treat population who received rezafungin; 94 received caspofungin. Baseline Candida species distribution was similar in the two treatment groups; C. albicans (occurring in 41.9% and 42.6% of patients in the rezafungin and caspofungin groups, respectively), C. glabrata (25.8% and 26.6%), and C. tropicalis (21.5% and 18.1%) were the most common pathogens. Rates of global cure and mycological eradication at day 14 and day 30 all-cause mortality by Candida species were comparable in the rezafungin and caspofungin treatment groups and did not appear to be impacted by minimal inhibitory concentration (MIC) values for either rezafungin or caspofungin. Two patients had baseline isolates with non-susceptible MIC values (both in the rezafungin group: one non-susceptible to rezafungin and one to caspofungin, classified as intermediate); both were candidemia-only patients in whom rezafungin treatment was successful based on the day 30 all-cause mortality endpoint. This analysis of ReSTORE demonstrated the efficacy of rezafungin for candidemia and IC in patients infected with a variety of Candida species.

INTRODUCTION

Commensal Candida species (spp.) are present on the skin and mucosa of 50%–70% of healthy individuals, but invasive candidiasis (IC), encompassing candidemia and infection of deep tissues, can occur as an opportunistic infection in immunocompromised or immunosuppressed individuals (1, 2). In healthcare settings, IC and candidemia are among the most frequently seen fungal diseases and bloodstream infections, respectively, and are associated with substantial morbidity and mortality (13). Five spp. account for most cases—C. albicans, Nakaseomyces glabratus (the previous classification as C. glabrata is retained in this report), Pichia kudriavzevii (termed C. krusei herein), C. tropicalis, and C. parapsilosis—although C. auris has also demonstrated high potential for nosocomial transmission since its emergence (1, 2).
IC is treated with systemic antifungals comprising azoles, amphotericin B, and echinocandins, with guidelines recommending echinocandin therapy for first-line use (4, 5). Echinocandins target 1,3-β-D-glucan synthase, resulting in destabilization of the fungal cell wall; however, mutations within FKS gene “hotspot” (HS) regions give rise to resistance (6). There has also been a rise in IC cases caused by non-albicans Candida spp., such as C. glabrata and C. auris, which have a higher intrinsic resistance potential (7). Increased rates of fluconazole resistance have also been reported, with intrinsic resistance seen in C. krusei (8, 9). As such, there is an urgent need for novel, effective antifungal agents.
Rezafungin received US Food and Drug Administration (FDA) approval in March 2023 for the treatment of candidemia and IC in patients aged ≥18 years with limited or no alternative treatment options (10) and was approved in the European Union for the treatment of IC in adults in December 2023 (11). This next-generation echinocandin is also in development for the prevention of invasive fungal diseases caused by Candida, Aspergillus, and Pneumocystis spp. in patients undergoing allogeneic blood and marrow transplantation (12). Compared with other echinocandins, rezafungin has an increased molecular stability, which results in a longer half-life that translates to higher, front-loaded exposure and allows weekly, rather than daily, administration (6, 13). The front-loaded exposure enhances antifungal activity early in treatment, potentially reducing the opportunity for FKS resistance-conferring mutations to arise (6, 13). Rezafungin has shown activity against a broad range of isolates, including azole-resistant Candida spp., and requires lower pharmacokinetic/pharmacodynamic (PK/PD) target exposures than other echinocandins (14, 15), which could enable treatment of some isolates with elevated MICs (13, 14). A study using 2021 provisional Clinical and Laboratory Standards Institute (CLSI) clinical susceptible-only breakpoints showed that a global 2019–2020 panel of Candida spp. had high susceptibility to rezafungin based on MICs (16). The inclusion of a CLSI MIC susceptibility breakpoint for rezafungin against C. auris (≤0.5 µg/mL) in the most recent CLSI Performance Standards for Antifungal Susceptibility Testing of Yeasts document (the provisional CLSI breakpoints were approved as of 20 January 2024, unpublished data) also highlights the promise of rezafungin, given this is the first C. auris susceptibility breakpoint defined for an antifungal agent (17).
The Phase 3 ReSTORE trial in patients with candidemia and/or IC, on which FDA approval was based, compared weekly treatment with rezafungin vs daily treatment with caspofungin, an established echinocandin (12). Rezafungin was non-inferior to caspofungin for the efficacy endpoints of all-cause mortality at day 30 (primary endpoint for FDA) and global cure at day 14 (primary endpoint for European Medicines Agency), with a similar safety profile. Here, we report an analysis of efficacy outcomes from the ReSTORE study in subgroups defined by baseline pathogen and susceptibility. Isolates demonstrating reduced echinocandin susceptibility were characterized for the presence of FKS mutations, and findings were evaluated in light of CLSI breakpoints and those breakpoints recently granted by the FDA (18).

RESULTS

Patients

The disposition of patients in ReSTORE was reported previously (12). Briefly, 222 patients were screened, and 199 were randomized to receive treatment (100 to rezafungin and 99 to caspofungin). Nine patients did not have a documented Candida infection in the 96 hours prior to randomization (n = 5 in the rezafungin group; n = 4 in the caspofungin group), and three did not receive study drug (two assigned to the rezafungin group, and one to caspofungin). The modified intent-to-treat (mITT) population, therefore, consisted of 187 patients (n = 93 patients in the rezafungin arm; n = 94 patients in the caspofungin arm) from whom 204 isolates were submitted for identification and susceptibility testing.

Baseline Candida spp.

Distribution of Candida spp. at baseline in the rezafungin and caspofungin treatment groups (Fig. 1) was well balanced, with the exception of C. parapsilosis, which was detected in 8.6% (8/93) of patients in the rezafungin group and 18.1% (17/94) of patients in the caspofungin group. MIC breakpoints for rezafungin and caspofungin by Candida pathogen are reported in Table 1, along with those for anidulafungin and micafungin for additional context. FDA MIC breakpoints for rezafungin are also listed in Table 1 (18). These breakpoints were defined based primarily on clinical and mycological data, with less emphasis on PK/PD, and are, therefore, more conservative than the CLSI values.
Fig 1
Fig 1 Baseline Candida spp. pathogens in the rezafungin and caspofungin treatment groups (mITT population). Data were calculated from 204 identified infections in 187 patients (rezafungin, n = 93; caspofungin, n = 94). All unique pathogens from the cultures collected within 96 hours prior to randomization or prior to the first dose of the study drug after randomization were summarized. Seventeen patients had multiple isolates at baseline: rezafungin treatment group (no. of patients): C. glabrata and C. tropicalis (3); C. albicans and C. glabrata (1); Candida dubliniensis and C. glabrata (1), Candida guilliermondii and C. tropicalis (1); C. parapsilosis and C. tropicalis (1); caspofungin treatment group: C. albicans and C. glabrata (3); C. albicans and C. tropicalis (3); C. glabrata and C. tropicalis (2); C. albicans and C. dubliniensis (1); C. glabrata and C. krusei (1).
TABLE 1
TABLE 1 Echinocandin MIC breakpoints
Candida spp.Breakpoint MIC (µg/mL)
FDAaCLSIb
 RezafunginAnidulafunginMicafunginCaspofungin
SSIRSIRSIRSIR
C. albicans≤0.12≤0.25  ≤0.250.5≥1≤0.250.5≥1≤0.250.5≥1
C. glabrata≤0.12≤0.5  ≤0.120.25≥0.5≤0.060.12≥0.25≤0.120.25≥0.5
C. tropicalis≤0.12≤0.25  ≤0.250.5≥1≤0.250.5≥1≤0.250.5≥1
C. parapsilosis≤2≤2  ≤24≥8≤24≥8≤24≥8
C. krusei ≤0.25  ≤0.250.5≥1≤0.250.5≥1≤0.250.5≥1
C. guilliermondii    ≤24≥8≤24≥8≤24≥8
C. dubliniensis ≤0.12           
C. auris ≤0.5           
a
Current FDA-approved breakpoints (18).
b
CLSI 2022 (17); rezafungin breakpoints are approved by CLSI as of 20 January 2024. I, intermediate; R, resistant; S, susceptible.

Fluconazole susceptibility

Although susceptibility to fluconazole was not a focus of the ReSTORE microbiological analyses, MIC values were generated for all isolates. The rate of fluconazole resistance at baseline was comparable between the rezafungin and caspofungin treatment groups. Pooled baseline isolate resistance rates were 0% (0/78) for C. albicans, 8.2% (4/49) for C. glabrata, 19.4% (7/36) for C. tropicalis, and 25.0% (6/24) for C. parapsilosis.

Efficacy outcomes

Global cure and mycological eradication

Global cure and mycological eradication at day 14 and treatment-specific MIC values by pathogen spp. at baseline for rezafungin and caspofungin are summarized in Table 2, with the antimicrobial activity against baseline Candida spp. detailed in Table S1. Rates of global cure and mycological eradication at day 14 were generally comparable between the two treatment groups.
TABLE 2
TABLE 2 Global cure and mycological eradication at day 14 by baseline Candida species
Candida spp.CharacteristicRezafungin (400/200 mg)Caspofungin (70/50 mg)
C. albicansGlobal cure at day 14, n/Na (%)21/39 (53.8)23/40 (57.5)
 Mycological eradication at day 14, n/Na (%)23/39 (59.0)24/40 (60.0)
 MIC90b (MIC range), μg/mL0.06 (0.008–0.12)0.06 (0.008–0.12)
 nc3939
C. glabrataGlobal cure at day 14, n/Na (%)16/24 (66.7)14/25 (56.0)
 Mycological eradication at day 14, n/Na (%)20/24 (83.3)15/25 (60.0)
 MIC90b (MIC range), μg/mL0.12 (0.03–0.5)0.06 (0.03–0.12)
 nc2425
C. tropicalisGlobal cure at day 14, n/Na (%)14/20 (70.0)10/17 (58.8)
 Mycological eradication at day 14, n/Na (%)15/20 (75.0)10/17 (58.8)
 MIC90b (MIC range), μg/mL0.06 (0.015–0.12)0.06 (0.015–0.12)
 nc2016
C. parapsilosisGlobal cure at day 14, n/Na (%)6/8 (75.0)11/17 (64.7)
 Mycological eradication at day 14, n/Na (%)6/8 (75.0)14/17 (82.4)
 MIC90b (MIC range), μg/mLN/A (0.5–2)0.5 (0.25–0.5)
 nc816
a
n/N is the number of patients with the listed response at day 14 per number of patients with the corresponding Candida pathogen at baseline.
b
For pathogens isolated ≥10 times in a treatment group.
c
Number of patients with baseline pathogens and susceptibility data available. MIC90, minimal inhibitory concentration required to inhibit 90% of isolates tested; N/A, not available.
Treatment-specific MIC90 values were also comparable between the two treatment groups (within a twofold difference) for all baseline Candida spp. where values could be determined (note, for C. parapsilosis, the MIC90 for rezafungin could not be estimated due to the small number of isolates; Table 2). Outcomes at day 14 by pathogen spp. and by rezafungin and caspofungin treatment-specific MIC values are given in Table 3. There was no trend for increasing MICs affecting day 14 mycological outcomes for either rezafungin or caspofungin for any of the Candida spp.
TABLE 3
TABLE 3 Outcomes at day 14 by baseline Candida spp. and rezafungin and caspofungin CLSI MIC values
Candida spp.
Treatment (isolatesb)
n/N (%) by treatment-specific MIC value, µg/mLa
0.0080.0150.030.060.120.250.512
C. albicans
Rezafungin [39]
 Global cure4/7 (57.1)11/20 (55.0)4/6 (66.7)1/4 (25.0)1/2 (50.0)    
 Mycological eradication5/7 (71.4)11/20 (55.0)5/6 (83.3)1/4 (25.0)1/2 (50.0)    
Caspofungin [39]
 Global cure1/2 (50.0)5/9 (55.6)11/21 (52.4)5/6 (83.3)0/1 (0)    
 Mycological eradication1/2 (50.0)5/9 (55.6)12/21 (57.1)5/6 (83.3)0/1 (0)    
C. glabrata
Rezafungin [24]
 Global cure  6/8 (75.0)4/6 (66.7)6/9 (66.7) 0/1 (0)  
 Mycological eradication  7/8 (87.5)6/6 (100)6/9 (66.7) 1/1 (100)  
Caspofungin [25]
 Global cure  2/5 (40.0)11/19 (57.9)1/1 (100)    
 Mycological eradication  2/5 (40.0)12/19 (63.2)1/1 (100)    
C. tropicalis
Rezafungin [20]
 Global cure 3/3 (100)5/8 (62.5)5/7 (71.4)1/2 (50.0)    
 Mycological eradication 3/3 (100)5/8 (62.5)5/7 (71.4)2/2 (100)    
Caspofungin [16]
 Global cure 1/1 (100)3/7 (42.9)4/7 (57.1)1/1 (100)    
 Mycological eradication 0/1 (0)4/7 (57.1)4/7 (57.1)1/1 (100)    
C. parapsilosis
Rezafungin [8]
 Global cure      1/1 (100)2/4 (50.0)3/3 (100)
 Mycological eradication      1/1 (100)2/4 (50.0)3/3 (100)
Caspofungin [16]
 Global cure     5/8 (62.5)6/8 (75.0)  
 Mycological eradication     5/8 (62.5)8/8 (100)  
a
n/N is the number of patients with the listed response at day 14 per number of patients with the corresponding Candida pathogen and MIC value at baseline; not all isolates had MIC data.
b
Number of isolates.

All-cause mortality

The observed pattern of day 30 all-cause mortality by baseline pathogen spp. was broadly similar across the rezafungin and caspofungin treatment groups, with most patients who died having C. albicans, C. glabrata, C. tropicalis, or C. parapsilosis isolates at baseline (Table 4). Day 30 all-cause mortality and treatment-specific MIC values by baseline pathogen spp. for each group are presented in Table S2.
TABLE 4
TABLE 4 Day 30 all-cause mortality by baseline Candida species (mITT population)
Candida spp.Rezafungin (400/200 mg; N = 93)Caspofungin (70/50 mg; N = 94)
n/N1aDay 30 ACM, %n/N1aDay 30 ACM, %
C. albicans11/3928.29/4022.5
C. glabrata4/2416.72/258.0
C. tropicalis5/2025.04/1723.5
C. parapsilosis1/812.56/1735.3
C. krusei1/250.00/20
C. guilliermondii0/200/00
C. dubliniensis0/300/10
C. lusitaniae0/100/10
C. metapsilosis0/100/00
C. nivariensis0/000/10
a
n/N1 is the number of patients with the corresponding Candida pathogen who died on or before day 30, or with unknown survival status/the number of patients with the corresponding Candida pathogen at baseline. ACM, all-cause mortality.

Clinical outcomes in patients with baseline isolates that were non-susceptible to rezafungin or caspofungin

Baseline isolates identified in ReSTORE were largely wild-type (WT) and susceptible to rezafungin and caspofungin (based on CLSI interpretation). Breakpoint data are available from two patients (in the rezafungin treatment group) who had baseline isolates with echinocandin non-susceptible MIC values in ReSTORE. One had a C. dubliniensis isolate that was non-susceptible to rezafungin, and the other had a C. glabrata isolate that was classed as intermediate for caspofungin but was susceptible to rezafungin. Clinical outcomes for these two patients are presented in Table 5. Both were candidemia-only patients, achieved mycological eradication at day 14, and were treatment successes based on the 30-day all-cause mortality endpoint. The patient with C. dubliniensis had an indeterminate global cure response at day 14, and the patient with C. glabrata failed to achieve a global cure response at day 14. The patient with C. glabrata had an FKS mutant isolate harboring the F659V Fks2 HS1 alteration, while the isolate from the patient withC. dubliniensis had no identified mutations in FKS HSs.
TABLE 5
TABLE 5 Outcomes of patients in ReSTORE with baseline Candida isolates that were non-susceptible to rezafungin or caspofungin
Patient (treatment group)SpeciesMycological response day 14Investigator response day 14Global response day 14ACM day 30MIC, µg/mL/CLSI interpretationaFks sequence
RezafunginCaspofungin
1 (rezafungin)C. dubliniensisEradicationIndeterminateIndeterminateAlive0.25/NS0.12/N/AWT
2 (rezafungin)C. glabrataEradicationFailureFailureAlive0.5/S0.25/IF659V (Fks2 HS1)
a
CLSI 2022 (17). ACM, all-cause mortality; HS1, hotspot 1; I, intermediate; N/A, not available; NS, non-susceptible; S, susceptible.

Clinical outcomes in patients with post-baseline isolates demonstrating reduced susceptibility to rezafungin or caspofungin

Isolates were recovered from two patients (both from blood cultures) who showed greater than equal to fourfold increases in rezafungin or caspofungin MIC values from baseline. One patient in the rezafungin treatment group had a C. glabrata isolate with a rezafungin MIC of 0.06 µg/mL at baseline. Treatment successes for day 5 and day 14 mycological response, day 14 global response, and day 30 all-cause mortality were achieved in this patient. However, at an unscheduled visit on day 35, the C. glabrata isolated from this patient had a rezafungin MIC value of 0.5 µg/mL and harbored a mutation in FKS2 (S663P, Fks2 HS1). The caspofungin MIC value for the isolate at day 35 was also elevated relative to baseline (0.25 vs 0.06 µg/mL, respectively). Based on multilocus sequencing type analysis of whole genome sequence data, both isolates were determined to be type 10, although it is unknown whether this day 35 isolate was isogenic to the FKS WT baseline C. glabrata or represented infection with a new strain. One patient in the caspofungin treatment group had a C. albicans isolate with a rezafungin MIC value of 0.03 µg/mL at baseline, rising to 0.12 µg/mL at day 5, with a concomitant increase in caspofungin MIC values from 0.008 µg/mL (baseline) to 0.06 µg/mL (day 5). Sequencing revealed that both the baseline and day 5 isolate were FKS1 WT. This patient died and had failed treatment outcomes based on all endpoints: day 5 and day 14 mycological response, day 14 Global Response, and day 30 ACM (i.e., deceased by day 30).

DISCUSSION

In this analysis of the Phase 3 ReSTORE study, rezafungin demonstrated efficacy based on rates of global cure and mycological eradication at day 14 and the day 30 all-cause mortality rate, regardless of baseline Candida spp. Efficacy outcomes did not appear to be impacted by MIC values across Candida spp. for either rezafungin or caspofungin. Although the distribution of Candida isolates in ReSTORE was largely WT and susceptible to rezafungin based on CLSI interpretation, rezafungin treatment was also successful in the limited number of patients from ReSTORE who had non-susceptible baseline Candida isolates and FKS mutant isolates.
The MIC data from this study were consistent with the worldwide reported antimicrobial activity (2019–2020) of rezafungin and caspofungin against Candida spp. (16). The rates of all-cause mortality at day 30 by pathogen spp. at baseline and treatment-specific MIC value were comparable between the two treatment groups. There were some numerical differences in global cure and mycological eradication between the treatments for certain species; however, the small sample sizes prohibited drawing broad conclusions on comparative efficacy by Candida pathogen. In other studies, correlations between MIC values and patient outcomes have not consistently been seen. For example, higher caspofungin MICs correlated with poor treatment outcomes among patients with C. glabrata, IC, and prior echinocandin exposure in a retrospective study (19), whereas an analysis of caspofungin clinical trial data found no such correlation (20). In addition, a correlation between high MIC and poor treatment outcomes was found in an analysis of 32 clinical isolates from candidemia patients treated with fluconazole (21) but was not observed in a larger population-based cohort (22). These findings suggest a multifactorial relationship between MIC values and treatment outcomes and the need for additional factors to be considered (23). Catheter placement is one such clinical risk factor affecting treatment outcomes that should be taken into account (24, 25).
A relationship between echinocandin exposure (area under the curve), MIC, and efficacy has been found for micafungin (26), underlining the importance of considering PK/PD parameters when evaluating breakpoints in the context of candidemia and IC. PK/PD simulations of the older echinocandins found that caspofungin and micafungin were likely to achieve therapeutic drug exposures in the majority of simulated patients relative to C. glabrata MIC90 values, whereas anidulafungin was not likely to achieve therapeutic drug exposures (27). Similar PK/PD simulations with a once-weekly 400 mg rezafungin regimen found a 100% probability of PK/PD target attainment across weeks 1–6 for the C. glabrata MIC90 of 0.12 µg/mL (28).
A clinical “susceptible” breakpoint for C. glabrata of ≤0.5 µg/mL was approved by the CLSI Subcommittee on Antifungal Susceptibility Tests on 20 January 2024; this breakpoint is higher than those established for anidulafungin (≤0.12 µg/mL), caspofungin (≤0.12 µg/mL), and micafungin (≤0.06 µg/mL) (17). As a drug that provides high plasma drug concentrations early in therapy, rezafungin may be better positioned to treat infections caused by isolates with higher MICs, as the epidemiology of C. glabrata moves toward reduced susceptibility to treatment with echinocandins. In the rezafungin clinical program, there were three patients (one in the ReSTORE study and two in the expanded access program) who had infections caused by FKS mutant C. glabrata isolates exhibiting reduced in vitro susceptibility to the approved echinocandins and rezafungin; however, all had positive treatment outcomes with rezafungin (29, 30). Of note, the ReSTORE trial FKS mutant C. glabrata isolate that was a mycological eradication success possessed the same Fks alteration (Fks2 HS1 F659V) as an isolate used in a neutropenic mouse model where rezafungin was also efficacious (11).
Despite a limited sample size, the prevalence of fluconazole-resistant isolates in the ReSTORE trial is reflective of increasing rates observed clinically over time for C. glabrata, C. tropicalis (particularly in the Asia-Pacific region), and C. parapsilosis (8, 9). Notably, all fluconazole-resistant isolates in the trial were susceptible to rezafungin and comparator echinocandins (data not shown), further supporting the role of echinocandins as first-line therapy for candidemia and IC.
As with the primary analysis of the ReSTORE trial (12), potential limitations of this analysis are that the study excluded those with specific forms of IC typically requiring long courses of antifungal treatment or occurring at sites where echinocandin penetration is poor, such as the urinary tract and the central nervous system. The study also excluded pediatric patients. These exclusion criteria limit the generalizability of the results of this analysis to these specific patient subgroups. The relatively small sample size in some of the baseline Candida pathogen groups is a further limitation specific to this analysis, as is the small number of samples with elevated and non-WT MICs, and the lack of echinocandin-resistant pathogens and C. auris isolates.

Conclusions

Overall, these data further support the efficacy of rezafungin in candidemia and IC. We found that rezafungin demonstrated efficacy for a global cure, mycological eradication, and day 30 all-cause mortality regardless of baseline Candida spp. Efficacy outcomes across Candida spp. did not appear to be impacted by MIC values for either rezafungin or caspofungin; assessment of other clinical factors may be warranted.

MATERIALS AND METHODS

Study design

ReSTORE (NCT03667690) (12) was a global, double-blind, double-dummy, randomized, Phase 3, non-inferiority trial of rezafungin vs caspofungin. Patients (≥18 years old) with candidemia or IC were randomly assigned (1:1) to receive either rezafungin 400/200 mg intravenously (IV) once weekly or caspofungin 70/50 mg IV once daily for ≥14 days (up to 4 weeks). Patients who met relevant criteria could be switched to oral step-down therapy after ≥3 days of IV therapy (placebo for the rezafungin group and fluconazole for the caspofungin group). The study was performed at 66 tertiary care centers across 15 countries between October 2018 and October 2021. The study was performed in compliance with the International Conference on Harmonisation Good Clinical Practice and the Declaration of Helsinki. All patients provided written informed consent. For full study details, please refer to the primary publication (12).

Eligibility criteria

Key inclusion criteria were an established mycological diagnosis of candidemia and/or IC ≤96 hours before randomization (≥1 blood culture positive for yeast or Candida, a positive test for Candida from a rapid in vitro diagnostic test, or a positive Gram stain for yeast or a positive culture for Candida spp. from a specimen obtained from a normally sterile site) and ≥1 systemic signs attributable to candidemia or IC appearing from ≤12 hours prior to the qualifying positive culture through to the time of randomization. If the positive blood culture was drawn >12 hours prior to randomization, an additional set of blood cultures was obtained≤12 hours before randomization to confirm Candida spp. status at enrollment. Results of the blood cultures obtained ≤12 hours before randomization were not required for the patient to be enrolled in the study. Patients with prosthetic joint septic arthritis, osteomyelitis, endocarditis, myocarditis, meningitis, endophthalmitis, central nervous system infection, chronic disseminated candidiasis, or urinary tract candidiasis were excluded, as were patients who had received systemic treatment with an antifungal agent for >48 hours in the 96 hours before randomization or who had an indwelling vascular catheter/device that could not be removed and that was likely to be the source of candidemia.

Assessments and outcomes

This analysis assessed outcomes among patients treated with rezafungin or caspofungin in the ReSTORE mITT in subgroups defined by Candida spp. and in vitro susceptibility at baseline. The mITT population was defined as patients who had a documented Candida infection based on central laboratory evaluation of culture from blood or another normally sterile site obtained ≤4 days (96 hours) before randomization and who received ≥1 dose of the study drug.
The genus and spp. of Candida pathogens were identified using mass spectrometry (matrix-assisted laser desorption/ionization–time of flight mass spectrometry, Bruker Daltonics, Bremen, Germany) by central laboratory evaluation of baseline blood and sterile site cultures. For susceptibility testing, CLSI (31) broth microdilution methodology was used to determine MIC values to rezafungin, caspofungin, anidulafungin, micafungin, and fluconazole for each Candida spp. Susceptibility interpretations for rezafungin and caspofungin were determined using the CLSI breakpoints (17) and those recently granted for rezafungin by the FDA (18).
Echinocandin non-susceptible isolates were submitted for whole-genome sequencing to screen for FKS mutations. Total genomic DNA was used as the input material for the library, which was sequenced using a MiSeq Sequencer (Illumina, San Diego, CA, USA). Reads were trimmed with Sickle, version 1.33 (32), with the error corrected using BayesHammer (33) in SPAdes 3.11.1 (34). Each sample was assembled using a reference-guided assembly in SeqMan NGen, version 16.0 (DNASTAR, Madison, WI, USA). DNA regions encoding FKS genes were compared to sequences available in the literature (35).
ReSTORE included two different primary efficacy endpoints; one (global cure at day 14) required by the European Medicines Agency and one (all-cause mortality at day 30) by the FDA. Global cure (defined as achieving clinical cure, mycological eradication, and, for patients with IC diagnosed by radiology, radiological cure; all confirmed by the independent data review committee) was assessed at day 14 (±1 day); and all-cause mortality was evaluated through day 30. Mycological response (eradication, failure, or indeterminate) was assessed at day 14 (±1 day) and at other timepoints; day 14 results are presented here unless otherwise stated. Mycological eradication was defined in these patients as a negative post-baseline blood culture or a negative post-baseline culture from another normally sterile site. If a follow-up culture from the normally sterile site other than blood was not accessible, the patient had to have achieved a successful clinical outcome, as assessed by the investigator, without a change of antifungal therapy for the treatment of candidemia and/or IC.
For this analysis, only descriptive data are presented, as counts and percentages.

ACKNOWLEDGMENTS

We thank all participants and investigators involved in ReSTORE. Medical writing support (including development of a draft outline and subsequent drafts in consultation with the authors, assembling tables and figures, collating author comments, copyediting, fact checking, and referencing) was provided by Caroline Greenwood, BSc (Hons), of Aspire Scientific (Bollington, UK) and funded by Melinta Therapeutics (Parsippany, NJ, USA).
The analysis described in this manuscript was funded by Melinta Therapeutics. The ReSTORE study was co-funded by Cidara Therapeutics and Mundipharma. Rezafungin is being developed by Cidara Therapeutics (San Diego, CA, USA) in partnership with Mundipharma (Cambridge, UK). Melinta Therapeutics (Parsippany, NJ, USA) holds the license to commercialize rezafungin in the USA.
A.D. reports consulting fees from Cidara. C.A. reports being an employee of Melinta. C.G.C. and M.C. report being an employee of JMI Laboratories. In 2022, JMI Laboratories was contracted to provide services for AbbVie, Inc., AimMax Therapeutics, Amicrobe, Inc., Appili Therapeutics, Armata Pharmaceuticals, Astellas Pharma, Inc., Basilea Pharmaceutica AG, Becton, Dickinson and Company, bioMérieux, Biosergen AB, Bugworks, Cerba Research NV, Cidara Therapeutics, Cipla USA Inc., ContraFect Corporation, CorMedix Inc., Crestone, Inc., Curza Global, LLC, Diamond V, Discuva Ltd., Entasis Therapeutics, Enveda Biosciences, Evopoint Biosciences, Fedora Pharmaceuticals, Fox Chase Chemical Diversity Center, Genentech, Gilead Sciences, Inc., GSK plc, Institute for Clinical Pharmacodynamics, Iterum Therapeutics plc, Janssen Biopharma, Johnson & Johnson, Kaleido Biosciences, LifeMine Therapeutics, Medpace, Inc, Lysovant Sciences, Inc, Meiji Seika Pharma, Melinta Therapeutics, Menarini Group, Merck & Co., MicuRx Pharmaceutical Inc., Mundipharma International Ltd., Mutabilis, Nabriva Therapeutics, National Cancer Institute, National Institutes of Health, Ohio State University, Omnix Medical Ltd., Paratek Pharmaceuticals, Pfizer, PolyPid Ltd., PPD, Prokaryotics, Inc., Pulmocide Ltd, Qpex Biopharma, Revagenix, Roche Holding AG, Roivant Sciences, Scynexis, Inc., SeLux Diagnostics, Shionogi & Co., Ltd., Sinovent Pharmaceuticals, Inc., Spero Therapeutics, Sumitovant Biopharma, Inc., TenNor Therapeutics, ThermoFisher Scientific, US Food and Drug Administration, VenatoRx Pharmaceuticals, Washington University, Watershed Medical, LLC, Wockhardt, and Zoetis, Inc. G.R.T. reports grants and consulting fees from Amplyx, Astellas, Cidara, F2G, and Manye; grants from Merck; and data safety monitoring board membership for Pfizer, outside of the submitted work. J.A.A. reports being an employee of Melinta. J.B.L. reports being an employee and shareholder of Cidara. K.B. reports being an employee and shareholder of Cidara. P.G.P. reports grants from and data review committee membership for Cidara and Melinta; grants from Astellas, Scynexis, and Merck; and advisory board membership for F2G and Matinas, outside of the submitted work. T.S. reports being an employee and stockholder of Cidara. All other authors (C.P. and D.A.) declare no competing interests.

SUPPLEMENTAL MATERIAL

Supplementary Tables - aac.01584-23-s0001.pdf
Table S1, Table S2.
ASM does not own the copyrights to Supplemental Material that may be linked to, or accessed through, an article. The authors have granted ASM a non-exclusive, world-wide license to publish the Supplemental Material files. Please contact the corresponding author directly for reuse.

REFERENCES

1.
Barantsevich N, Barantsevich E. 2022. Diagnosis and treatment of invasive candidiasis. Antibiotics (Basel) 11:718.
2.
Soriano A, Honore PM, Puerta-Alcalde P, Garcia-Vidal C, Pagotto A, Gonçalves-Bradley DC, Verweij PE. 2023. Invasive candidiasis: current clinical challenges and unmet needs in adult populations. J Antimicrob Chemother 78:1569–1585.
3.
Tsay SV, Mu Y, Williams S, Epson E, Nadle J, Bamberg WM, Barter DM, Johnston HL, Farley MM, Harb S, et al. 2020. Burden of candidemia in the United States, 2017. Clin Infect Dis 71:e449–e453.
4.
Pappas PG, Kauffman CA, Andes DR, Clancy CJ, Marr KA, Ostrosky-Zeichner L, Reboli AC, Schuster MG, Vazquez JA, Walsh TJ, Zaoutis TE, Sobel JD. 2016. Clinical practice guideline for the management of candidiasis: 2016 update by the Infectious Diseases Society of America. Clin Infect Dis 62:e1–50.
5.
Cornely OA, Bassetti M, Calandra T, Garbino J, Kullberg BJ, Lortholary O, Meersseman W, Akova M, Arendrup MC, Arikan-Akdagli S, et al. 2012. ESCMID guideline for the diagnosis and management of Candida diseases 2012: non-neutropenic adult patients. Clin Microbiol Infect 18 Suppl 7:19–37.
6.
Murphy SE, Bicanic T. 2021. Drug resistance and novel therapeutic approaches in invasive candidiasis. Front Cell Infect Microbiol 11:759408.
7.
Pappas PG, Lionakis MS, Arendrup MC, Ostrosky-Zeichner L, Kullberg BJ. 2018. Invasive candidiasis. Nat Rev Dis Primers 4:18026.
8.
Escribano P, Guinea J. 2022. Fluconazole-resistant Candida parapsilosis: a new emerging threat in the fungi arena. Front Fungal Biol 3:1010782.
9.
Pfaller MA, Diekema DJ, Turnidge JD, Castanheira M, Jones RN. 2019. Twenty years of the SENTRY antifungal surveillance program: results for Candida species from 1997-2016. Open Forum Infect Dis 6:S79–S94.
10.
US Food and Drug Administration. 2023. Rezzayo prescribing information. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/217417s000lbl.pdf. Retrieved 12 Jun 2023.
11.
European Medicines Agency. 2024. Rezzayo (rezafungin) summary of product characteristics. Available from: https://www.ema.europa.eu/en/documents/product-information/rezzayo-epar-product-information_en.pdf. Retrieved 1 Feb 2024.
12.
Thompson GR 3rd, Soriano A, Cornely OA, Kullberg BJ, Kollef M, Vazquez J, Honore PM, Bassetti M, Pullman J, Chayakulkeeree M, Poromanski I, Dignani C, Das AF, Sandison T, Pappas PG, ReSTORE trial investigators. 2023. Rezafungin versus caspofungin for treatment of candidaemia and invasive candidiasis (ReSTORE): a multicentre, double-blind, double-dummy, randomised phase 3 trial. Lancet 401:49–59.
13.
Ham YY, Lewis JS 2nd, Thompson GR 3rd. 2021. Rezafungin: a novel antifungal for the treatment of invasive candidiasis. Future Microbiol 16:27–36.
14.
Lepak AJ, Zhao M, VanScoy B, Ambrose PG, Andes DR. 2018. Pharmacodynamics of a long-acting echinocandin, CD101, in a neutropenic invasive-candidiasis murine model using an extended-interval dosing design. Antimicrob Agents Chemother 62:e02154-17.
15.
Pfaller MA, Messer SA, Rhomberg PR, Jones RN, Castanheira M. 2016. Activity of a long-acting echinocandin, CD101, determined using CLSI and EUCAST reference methods, against Candida and Aspergillus spp., including echinocandin- and azole-resistant isolates. J Antimicrob Chemother 71:2868–2873.
16.
Carvalhaes CG, Klauer AL, Rhomberg PR, Pfaller MA, Castanheira M. 2022. Evaluation of rezafungin provisional CLSI clinical breakpoints and epidemiological cutoff values tested against a worldwide collection of contemporaneous invasive fungal isolates (2019 to 2020). J Clin Microbiol 60:e0244921.
17.
Clinical and Laboratory Standards Institute. 2022. Performance standards for antifungal susceptibility testing of yeasts [M27M44S]. 3rd ed. Clinical and Laboratory Standards Institute, Wayne, PA.
18.
US Food and Drug Administration. 2023. Rezafungin injection. Available from: https://www.fda.gov/drugs/development-resources/rezafungin-injection. Retrieved 13 Jul 2023.
19.
Shields RK, Nguyen MH, Press EG, Updike CL, Clancy CJ. 2013. Caspofungin MICs correlate with treatment outcomes among patients with Candida glabrata invasive candidiasis and prior echinocandin exposure. Antimicrob Agents Chemother 57:3528–3535.
20.
Kartsonis N, Killar J, Mixson L, Hoe CM, Sable C, Bartizal K, Motyl M. 2005. Caspofungin susceptibility testing of isolates from patients with esophageal candidiasis or invasive candidiasis: relationship of MIC to treatment outcome. Antimicrob Agents Chemother 49:3616–3623.
21.
Clancy CJ, Yu VL, Morris AJ, Snydman DR, Nguyen MH. 2005. Fluconazole MIC and the fluconazole dose/MIC ratio correlate with therapeutic response among patients with candidemia. Antimicrob Agents Chemother 49:3171–3177.
22.
Fernández-Ruiz M, Guinea J, Lora-Pablos D, Zaragoza Ó, Puig-Asensio M, Almirante B, Cuenca-Estrella M, Aguado JM, Padilla B, Muñoz P, et al. 2017. Impact of fluconazole susceptibility on the outcome of patients with candidaemia: data from a population-based surveillance. CMI 23:672.
23.
Rex JH, Pfaller MA, Barry AL, Nelson PW, Webb CD, NIAID Mycoses Study Group, Candidemia Study Group. 1995. Antifungal susceptibility testing of isolates from a randomized, multicenter trial of fluconazole versus amphotericin B as treatment of nonneutropenic patients with candidemia. Antimicrob Agents Chemother 39:40–44.
24.
Bilal H, Zhang D, Shafiq M, Khan MN, Chen C, Khan S, Wang Q, Cai L, Islam R, Hu H, Zeng Y. 2023. Six-year retrospective analysis of epidemiology, risk factors, and antifungal susceptibilities of candidiasis from a tertiary care hospital in South China. Microbiol Spectr 11:e0070823.
25.
Hoenigl M, Salmanton-García J, Egger M, Gangneux J-P, Bicanic T, Arikan-Akdagli S, Alastruey-Izquierdo A, Klimko N, Barac A, Özenci V, et al. 2023. Guideline adherence and survival of patients with candidaemia in Europe: results from the ECMM Candida III multinational European observational cohort study. Lancet Infect Dis 23:751–761.
26.
Andes D, Ambrose PG, Hammel JP, Van Wart SA, Iyer V, Reynolds DK, Buell DN, Kovanda LL, Bhavnani SM. 2011. Use of pharmacokinetic-pharmacodynamic analyses to optimize therapy with the systemic antifungal micafungin for invasive candidiasis or candidemia. Antimicrob Agents Chemother 55:2113–2121.
27.
Bader JC, Bhavnani SM, Andes DR, Ambrose PG. 2018. We can do better: a fresh look at echinocandin dosing. J Antimicrob Chemother 73:i44–i50.
28.
Bader JC, Lakota EA, Flanagan S, Ong V, Sandison T, Rubino CM, Bhavnani SM, Ambrose PG. 2018. Overcoming the resistance hurdle: pharmacokinetic-pharmacodynamic target attainment analyses for rezafungin (Cd101) against Candida albicans and Candida glabrata. Antimicrob Agents Chemother 62:e02614-17.
29.
Adeel A, Qu MD, Siddiqui E, Levitz SM, Ellison RT. 2021. Expanded access use of rezafungin for salvage therapy of invasive Candida glabrata infection: a case report. Open Forum Infect Dis 8:ofab431.
30.
US Food and Drug Administration. 2023. Rezafungin injection for treatment of candidemia and invasive candidiasis. Sponsor Briefing Document. Antimicrobial Drugs Advisory Committee. Available from: https://www.fda.gov/media/164668/download. Retrieved 13 Jul 2023.
31.
Clinical and Laboratory Standards Institute. 2017. Reference method for broth dilution antifungal susceptibility testing of yeasts [M27]. 4th ed. Clinical and Laboratory Standards Institute, Wayne, PA.
32.
Joshi NA, Fass JN. 2011. Sickle: a sliding-window, adaptive, quality-based trimming tool for FastQ files (version 1.33). Software. Available from: https://github.com/najoshi/sickle. Retrieved 21 Aug 2023.
33.
Nikolenko SI, Korobeynikov AI, Alekseyev MA. 2013. Bayeshammer: bayesian clustering for error correction in single-cell sequencing. BMC Genomics 14 Suppl 1:S7.
34.
Nurk S, Bankevich A, Antipov D, Gurevich AA, Korobeynikov A, Lapidus A, Prjibelski AD, Pyshkin A, Sirotkin A, Sirotkin Y, Stepanauskas R, Clingenpeel SR, Woyke T, McLean JS, Lasken R, Tesler G, Alekseyev MA, Pevzner PA. 2013. Assembling single-cell genomes and mini-metagenomes from chimeric MDA products. J Comput Biol 20:714–737.
35.
Castanheira M, Deshpande LM, Davis AP, Rhomberg PR, Pfaller MA. 2017. Monitoring antifungal resistance in a global collection of invasive yeasts and molds: application of CLSI epidemiological cutoff values and whole-genome sequencing analysis for detection of azole resistance in Candida albicans. Antimicrob Agents Chemother 61:e00906-17.

Information & Contributors

Information

Published In

cover image Antimicrobial Agents and Chemotherapy
Antimicrobial Agents and Chemotherapy
Volume 68Number 52 May 2024
eLocator: e01584-23
Editor: Andreas H. Groll, University Children's Hospital Münster, Münster, Germany
PubMed: 38526046

History

Received: 6 December 2023
Accepted: 16 February 2024
Published online: 25 March 2024

Keywords

  1. candidemia
  2. echinocandin
  3. invasive candidiasis
  4. Candida species
  5. antifungal therapy

Contributors

Authors

Cidara Therapeutics, Inc., San Diego, California, USA
Chris M. Pillar
Science and Operations, Microbiologics, Kalamazoo, Michigan, USA
JMI Laboratories, North Liberty, Iowa, USA
Cecilia G. Carvalhaes
JMI Laboratories, North Liberty, Iowa, USA
Present address: bioMérieux, St Louis, Missouri, USA
Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
Medical Affairs, Melinta Therapeutics, LLC, Parsippany, New Jersey, USA
Christina Andrzejewski
Medical Affairs, Melinta Therapeutics, LLC, Parsippany, New Jersey, USA
Ken Bartizal
Cidara Therapeutics, Inc., San Diego, California, USA
Anita F. Das
Cidara Therapeutics, Inc., San Diego, California, USA
Taylor Sandison
Cidara Therapeutics, Inc., San Diego, California, USA
George R. Thompson, III
Division of Infectious Diseases, Department of Internal Medicine and Department of Medical Microbiology and Immunology, University of California Davis Medical Center, Sacramento, California, USA
Peter G. Pappas
Division of Infectious Diseases, Department of Internal Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA

Editor

Andreas H. Groll
Editor
University Children's Hospital Münster, Münster, Germany

Metrics & Citations

Metrics

Note:

  • For recently published articles, the TOTAL download count will appear as zero until a new month starts.
  • There is a 3- to 4-day delay in article usage, so article usage will not appear immediately after publication.
  • Citation counts come from the Crossref Cited by service.

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. For an editable text file, please select Medlars format which will download as a .txt file. Simply select your manager software from the list below and click Download.

View Options

Figures

Tables

Media

Share

Share

Share the article link

Share with email

Email a colleague

Share on social media

American Society for Microbiology ("ASM") is committed to maintaining your confidence and trust with respect to the information we collect from you on websites owned and operated by ASM ("ASM Web Sites") and other sources. This Privacy Policy sets forth the information we collect about you, how we use this information and the choices you have about how we use such information.
FIND OUT MORE about the privacy policy