Efficacy, Safety, Pharmacokinetics, and Microbiome Changes of Ibezapolstat in Adults with Clostridioides difficile Infection: A Phase 2a Multicenter Clinical Trial

Kevin W Garey, Jacob McPherson, An Q Dinh, Chenlin Hu, Jinhee Jo, Weiqun Wang, Chris K Lancaster, Anne J Gonzales-Luna, Caroline Loveall, Khurshida Begum, M Jahangir Alam, Michael H Silverman, Blake M Hanson, Kevin W Garey, Jacob McPherson, An Q Dinh, Chenlin Hu, Jinhee Jo, Weiqun Wang, Chris K Lancaster, Anne J Gonzales-Luna, Caroline Loveall, Khurshida Begum, M Jahangir Alam, Michael H Silverman, Blake M Hanson

Abstract

Background: This study was the first human validation of the gram-positive bacterial DNA polymerase IIIC target in patients with Clostridioides difficile infection. The primary objectives were to assess clinical cure rates and adverse events (AEs). Secondary objectives were to evaluate plasma/fecal pharmacokinetics, microbiologic eradication, microbiome and bile acid effects, and sustained clinical cure (SCC) with ibezapolstat.

Methods: This single-arm, open-label, phase 2a study enrolled adults with C. difficile infection at 4 US centers. Patients received ibezapolstat 450 mg orally every 12 hours for 10 days and followed for an additional 28 days to assess study objectives.

Results: Ten patients with a mean (standard deviation [SD]) age of 49 [15] years were enrolled. Seven AEs were reported classified as mild-moderate. Plasma levels of ibezapolstat ranged from 233 to 578 ng/mL while mean (SD) fecal levels were 416 (494) µg/g stool by treatment day 3 and >1000 µg/g stool by days 8-10. A rapid increase in alpha diversity in the fecal microbiome was noted after starting ibezapolstat therapy, which was maintained after completion of therapy. A proportional decrease in Bacteroidetes phylum was observed (mean change [SD], -10.0% [4.8%]; P = .04) with a concomitantly increased proportion of Firmicutes phylum (+14.7% [5.4%]; P = .009). Compared with baseline, total primary bile acids decreased by a mean (SD) of 40.1 (9.6) ng/mg stool during therapy (P < .001) and 40.5 (14.1) ng/mg stool after completion of therapy (P = .007). Rates of both initial clinical cure and SCC at 28 days were 100% (10 of 10 patients).

Conclusions: In this phase 2a study, 10 of 10 patients achieved SCC, demonstrated favorable pharmacokinetics, minimal AEs, and beneficial microbiome and bile acids results. These results support continued clinical development.

Trial registration: ClinicalTrials.gov NCT04247542.

Keywords: Clostridioides difficile; DNA polymerase IIIC; Enterocolitis; Female; Male; humans.

Conflict of interest statement

Potential conflicts of interest. K. W. G. received research grant support from Acurx Pharmaceuticals, Paratek, and Summit Pharmaceuticals. M. H. S. is a shareholder and paid consultant for Acurx Pharmaceuticals and received consulting fees from Summit, Paratek, Tetraphase, and Seres. M. H. S. is a shareholder of and paid consultant to Acurx Pharmaceuticals, Inc. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.

© The Author(s) 2022. Published by Oxford University Press for the Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
Ibezapolstat pharmacokinetics in plasma (A) and stool (B) samples. Values represent means with standard deviations.
Figure 2.
Figure 2.
Summary estimates of changes in alpha diversity over time with the Shannon diversity (A) and inverse Simpson (B) indexes.
Figure 3.
Figure 3.
Effects of ibezapolstat on relative abundance of taxa by phylum (A), class (B), order (C), and family (D).
Figure 4.
Figure 4.
Changes over time in primary (A) and secondary (B) bile acid concentrations and the ratio of secondary to primary bile acid concentrations (C). Values represent means with standard errors.

References

    1. Magill SS, O’Leary E, Janelle SJ, et al. . Changes in prevalence of health care-associated infections in U.S. Hospitals. N Engl J Med 2018; 379:1732–44.
    1. Lessa FC, Winston LG, McDonald LC; Emerging Infections Program C. difficile Surveillance Team. Burden of Clostridium difficile infection in the United States. N Engl J Med 2015; 372:2369–70.
    1. Johnson S, Lavergne V, Skinner AM, et al. . 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; 73:755–7.
    1. Shen WJ, Deshpande A, Hevener KE, et al. . Constitutive expression of the cryptic vanGCd operon promotes vancomycin resistance in Clostridioides difficile clinical isolates. J Antimicrob Chemother 2020; 75:859–67.
    1. Gonzales-Luna AJ, Spinler JK, Oezguen N, et al. . Systems biology evaluation of refractory Clostridioides difficile infection including multiple failures of fecal microbiota transplantation. Anaerobe 2021; 70:102387.
    1. Oka D, Yamaya N, Kuno T, et al. . In vitro and in vivo antibacterial activities of a novel quinolone compound, OPS-2071, against Clostridioides difficile. Antimicrob Agents Chemother 2021; 65:e01170-20.
    1. Khoruts A, Dicksved J, Jansson JK, Sadowsky MJ.. Changes in the composition of the human fecal microbiome after bacteriotherapy for recurrent Clostridium difficile-associated diarrhea. J Clin Gastroenterol 2010; 44:354–60.
    1. Alam MJ, Walk ST, Endres BT, et al. . Community environmental contamination of toxigenic Clostridium difficile. Open Forum Infect Dis 2017; 4:ofx018.
    1. Basseres E, Endres BT, Dotson KM, Alam MJ, Garey KW.. Novel antibiotics in development to treat Clostridium difficile infection. Curr Opin Gastroenterol 2017; 33:1–7.
    1. Xu WC, Silverman MH, Yu XY, Wright G, Brown N.. Discovery and development of DNA polymerase IIIC inhibitors to treat gram-positive infections. Bioorg Med Chem 2019; 27:3209–17.
    1. van Eijk E, Boekhoud IM, Kuijper EJ, Bos-Sanders I, Wright G, Smits WK.. Genome location dictates the transcriptional response to PolC inhibition in Clostridium difficile. Antimicrob Agents Chemother 2019; 63:e01263-18.
    1. Garey KW, Begum K, Lancaster C, et al. . A randomized, double-blind, placebo-controlled, single and multiple ascending dose phase 1 study to determine the safety, pharmacokinetics and food and faecal microbiome effects of ibezapolstat administered orally to healthy subjects. J Antimicrob Chemother 2020; 75:3635–43.
    1. McDonald LC, Gerding DN, Johnson S, et al. . Clinical practice guidelines for Clostridium difficile infection in adults and children: 2017 update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA). Clin Infect Dis 2018; 66:e1–48.
    1. Gonzales-Luna AJ, Carlson TJ, Dotson KM, et al. . PCR ribotypes of Clostridioides difficile across Texas from 2011 to 2018 including emergence of ribotype 255. Emerg Microbes Infect 2020; 9:341–7.
    1. Begum K, Basseres E, Miranda J, et al. . In vitro activity of omadacycline, a new tetracycline analog, and comparators against Clostridioides difficile. Antimicrob Agents Chemother 2020; 64:e00522-20.
    1. Fadrosh DW, Ma B, Gajer P, et al. . An improved dual-indexing approach for multiplexed 16S rRNA gene sequencing on the Illumina MiSeq platform. Microbiome 2014; 2:6.
    1. Walker JN, Hanson BM, Pinkner CL, et al. . Insights into the microbiome of breast implants and periprosthetic tissue in breast implant-associated anaplastic large cell lymphoma. Sci Rep 2019; 9:10393.
    1. Scherer M, Gnewuch C, Schmitz G, Liebisch G.. Rapid quantification of bile acids and their conjugates in serum by liquid chromatography-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2009; 877:3920–5.
    1. R Core Team. R: a language and environment for statistical computing. Vienna, Austria: R Foundation for Statistical Computing,2013. Available at: . Accessed 22 February 2022.
    1. Ridlon JM, Kang DJ, Hylemon PB.. Bile salt biotransformations by human intestinal bacteria. J Lipid Res 2006; 47:241–59.
    1. Kullar R, Tran MN, Goldstein EJC.. Investigational treatment agents for recurrent Clostridioides difficile infection (rCDI). J Exp Pharmacol 2020; 12:371–84.
    1. Winston JA, Theriot CM.. Impact of microbial derived secondary bile acids on colonization resistance against Clostridium difficile in the gastrointestinal tract. Anaerobe 2016; 41:44–50.

Source: PubMed

3
Se inscrever