Molnupiravir versus placebo in unvaccinated and vaccinated patients with early SARS-CoV-2 infection in the UK (AGILE CST-2): a randomised, placebo-controlled, double-blind, phase 2 trial

Saye H Khoo, Richard FitzGerald, Geoffrey Saunders, Calley Middleton, Shazaad Ahmad, Christopher J Edwards, Dennis Hadjiyiannakis, Lauren Walker, Rebecca Lyon, Victoria Shaw, Pavel Mozgunov, Jimstan Periselneris, Christie Woods, Katie Bullock, Colin Hale, Helen Reynolds, Nichola Downs, Sean Ewings, Amanda Buadi, David Cameron, Thomas Edwards, Emma Knox, I'ah Donovan-Banfield, William Greenhalf, Justin Chiong, Lara Lavelle-Langham, Michael Jacobs, Josh Northey, Wendy Painter, Wayne Holman, David G Lalloo, Michelle Tetlow, Julian A Hiscox, Thomas Jaki, Thomas Fletcher, Gareth Griffiths, AGILE CST-2 Study Group, Nicholas Paton, Fred Hayden, Janet Darbyshire, Amy Lucas, Ulrika Lorch, Andrew Freedman, Richard Knight, Stevan Julious, Rachel Byrne, Ana Cubas Atienzar, Jayne Jones, Chris Williams, Anna Song, Jan Dixon, Anja Alexandersson, Parys Hatchard, Emma Tilt, Andrew Titman, Ale Doce Carracedo, Vatsi Chandran Gorner, Andrea Davies, Louis Woodhouse, Nicola Carlucci, Emmanuel Okenyi, Marcin Bula, Kate Dodd, Jennifer Gibney, Lesley Dry, Zalina Rashid Gardner, Amin Sammour, Christine Cole, Tim Rowland, Maria Tsakiroglu, Vincent Yip, Rostam Osanlou, Anna Stewart, Ben Parker, Tolga Turgut, Afshan Ahmed, Kay Starkey, Sujamole Subin, Jennifer Stockdale, Lisa Herring, Jonathon Baker, Abigail Oliver, Mihaela Pacurar, Dan Owens, Alistair Munro, Gavin Babbage, Saul Faust, Matthew Harvey, Danny Pratt, Deepak Nagra, Aashish Vyas, Saye H Khoo, Richard FitzGerald, Geoffrey Saunders, Calley Middleton, Shazaad Ahmad, Christopher J Edwards, Dennis Hadjiyiannakis, Lauren Walker, Rebecca Lyon, Victoria Shaw, Pavel Mozgunov, Jimstan Periselneris, Christie Woods, Katie Bullock, Colin Hale, Helen Reynolds, Nichola Downs, Sean Ewings, Amanda Buadi, David Cameron, Thomas Edwards, Emma Knox, I'ah Donovan-Banfield, William Greenhalf, Justin Chiong, Lara Lavelle-Langham, Michael Jacobs, Josh Northey, Wendy Painter, Wayne Holman, David G Lalloo, Michelle Tetlow, Julian A Hiscox, Thomas Jaki, Thomas Fletcher, Gareth Griffiths, AGILE CST-2 Study Group, Nicholas Paton, Fred Hayden, Janet Darbyshire, Amy Lucas, Ulrika Lorch, Andrew Freedman, Richard Knight, Stevan Julious, Rachel Byrne, Ana Cubas Atienzar, Jayne Jones, Chris Williams, Anna Song, Jan Dixon, Anja Alexandersson, Parys Hatchard, Emma Tilt, Andrew Titman, Ale Doce Carracedo, Vatsi Chandran Gorner, Andrea Davies, Louis Woodhouse, Nicola Carlucci, Emmanuel Okenyi, Marcin Bula, Kate Dodd, Jennifer Gibney, Lesley Dry, Zalina Rashid Gardner, Amin Sammour, Christine Cole, Tim Rowland, Maria Tsakiroglu, Vincent Yip, Rostam Osanlou, Anna Stewart, Ben Parker, Tolga Turgut, Afshan Ahmed, Kay Starkey, Sujamole Subin, Jennifer Stockdale, Lisa Herring, Jonathon Baker, Abigail Oliver, Mihaela Pacurar, Dan Owens, Alistair Munro, Gavin Babbage, Saul Faust, Matthew Harvey, Danny Pratt, Deepak Nagra, Aashish Vyas

Abstract

Background: The antiviral drug molnupiravir was licensed for treating at-risk patients with COVID-19 on the basis of data from unvaccinated adults. We aimed to evaluate the safety and virological efficacy of molnupiravir in vaccinated and unvaccinated individuals with COVID-19.

Methods: This randomised, placebo-controlled, double-blind, phase 2 trial (AGILE CST-2) was done at five National Institute for Health and Care Research sites in the UK. Eligible participants were adult (aged ≥18 years) outpatients with PCR-confirmed, mild-to-moderate SARS-CoV-2 infection who were within 5 days of symptom onset. Using permuted blocks (block size 2 or 4) and stratifying by site, participants were randomly assigned (1:1) to receive either molnupiravir (orally; 800 mg twice daily for 5 days) plus standard of care or matching placebo plus standard of care. The primary outcome was the time from randomisation to SARS-CoV-2 PCR negativity on nasopharyngeal swabs and was analysed by use of a Bayesian Cox proportional hazards model for estimating the probability of a superior virological response (hazard ratio [HR]>1) for molnupiravir versus placebo. Our primary model used a two-point prior based on equal prior probabilities (50%) that the HR was 1·0 or 1·5. We defined a priori that if the probability of a HR of more than 1 was more than 80% molnupiravir would be recommended for further testing. The primary outcome was analysed in the intention-to-treat population and safety was analysed in the safety population, comprising participants who had received at least one dose of allocated treatment. This trial is registered in ClinicalTrials.gov, NCT04746183, and the ISRCTN registry, ISRCTN27106947, and is ongoing.

Findings: Between Nov 18, 2020, and March 16, 2022, 1723 patients were assessed for eligibility, of whom 180 were randomly assigned to receive either molnupiravir (n=90) or placebo (n=90) and were included in the intention-to-treat analysis. 103 (57%) of 180 participants were female and 77 (43%) were male and 90 (50%) participants had received at least one dose of a COVID-19 vaccine. SARS-CoV-2 infections with the delta (B.1.617.2; 72 [40%] of 180), alpha (B.1.1.7; 37 [21%]), omicron (B.1.1.529; 38 [21%]), and EU1 (B.1.177; 28 [16%]) variants were represented. All 180 participants received at least one dose of treatment and four participants discontinued the study (one in the molnupiravir group and three in the placebo group). Participants in the molnupiravir group had a faster median time from randomisation to negative PCR (8 days [95% CI 8-9]) than participants in the placebo group (11 days [10-11]; HR 1·30, 95% credible interval 0·92-1·71; log-rank p=0·074). The probability of molnupiravir being superior to placebo (HR>1) was 75·4%, which was less than our threshold of 80%. 73 (81%) of 90 participants in the molnupiravir group and 68 (76%) of 90 participants in the placebo group had at least one adverse event by day 29. One participant in the molnupiravir group and three participants in the placebo group had an adverse event of a Common Terminology Criteria for Adverse Events grade 3 or higher severity. No participants died (due to any cause) during the trial.

Interpretation: We found molnupiravir to be well tolerated and, although our predefined threshold was not reached, we observed some evidence that molnupiravir has antiviral activity in vaccinated and unvaccinated individuals infected with a broad range of SARS-CoV-2 variants, although this evidence is not conclusive.

Funding: Ridgeback Biotherapeutics, the UK National Institute for Health and Care Research, the Medical Research Council, and the Wellcome Trust.

Conflict of interest statement

Declaration of interests SHK has received research funding from ViiV Healthcare and Merck for the Liverpool HIV Drug Interactions programme and for unrelated clinical studies. GG has received funding from Janssen-Cilag, AstraZeneca, Novartis, Astex, Roche, Heartflow, Celldex, BMS, BionTech, Cancer Research UK, the NIHR, the British Lung Foundation, Unitaid, and GSK for unrelated academic clinical trials and programme funding. WG has received funding from the Wellcome Trust. WH is a cofounder and owner of, and adviser for, Ridgeback Biotherapeutics. WP is employed by Ridgeback Biotherapeutics. JP has received honoraria for lectures from Gilead and London International Patient Services UK. JAH has received research funding from the US Food and Drug Administration and payment for expert testimony from DAC Beachcroft and Clyde and Co. TE has received unrelated research funding from the Medical Research Council, the Wellcome Trust, and Bloomsbury SET and travel grants from the European Society of Clinical Microbiology and Infectious Diseases. All other authors declare no competing interests.

Copyright © 2023 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Trial profile
Figure 2
Figure 2
Kaplan–Meier plots for time from randomisation to negative PCR (A) All patients. (B) Vaccinated patients. (C) Unvaccinated patients. Shaded areas represent 95% CIs.
Figure 3
Figure 3
Forest plot for time from randomisation to negative PCR by subgroup For our analysis by variant, we did not include patients with an unknown or unique (B.1.1.1 and XE) variant type. *People could identify as White English, Welsh, Scottish, Northern Irish, or British.

References

    1. Khoo SH, Fitzgerald R, Fletcher T, et al. Optimal dose and safety of molnupiravir in patients with early SARS-CoV-2: a phase I, open-label, dose-escalating, randomized controlled study. J Antimicrob Chemother. 2021;76:3286–3295.
    1. Jayk Bernal A, Gomes da Silva MM, Musungaie DB, et al. Molnupiravir for oral treatment of Covid-19 in nonhospitalized patients. N Engl J Med. 2022;386:509–520.
    1. Kumarasamy N, Saha B, Jindal A, et al. Phase 3 trial of molnupiravir in adults with mild SARS-CoV-2 infection in India. Conference on Retroviruses and Opportunistic Infections; Feb 15, 2022 (abstract O-9).
    1. Troth S, Butterton J, DeAnda CS, et al. Letter to the editor in response to Zhou et al. J Infect Dis. 2021;224:1442–1443.
    1. US Food and Drug Administration Center for Drug Evaluation and Research. FDA briefing document. Antimicrobial drugs advisory committee meeting. Nov 30, 2021.
    1. Griffiths GO, FitzGerald R, Jaki T, et al. AGILE: a seamless phase I/IIa platform for the rapid evaluation of candidates for COVID-19 treatment: an update to the structured summary of a study protocol for a randomised platform trial letter. Trials. 2021;22:487.
    1. WHO Working Group on the Clinical Characterisation and Management of COVID-19 infection A minimal common outcome measure set for COVID-19 clinical research. Lancet Infect Dis. 2020;20:e192–e197.
    1. Coolen JPM, Wolters F, Tostmann A, et al. SARS-CoV-2 whole-genome sequencing using reverse complement PCR: for easy, fast and accurate outbreak and variant analysis. J Clin Virol. 2021;144:104993.
    1. O'Toole Á, Scher E, Underwood A, et al. Assignment of epidemiological lineages in an emerging pandemic using the Pangolin tool. Virus Evol. 2021;7:veab064.
    1. Ewings S, Saunders G, Jaki T, Mozgunov P. Practical recommendations for implementing a Bayesian adaptive phase I design during a pandemic. BMC Med Res Methodol. 2022;22:25.
    1. He X, Lau EHY, Wu P, et al. Temporal dynamics in viral shedding and transmissibility of COVID-19. Nat Med. 2020;26:672–675.
    1. US Food and Drug Administration Guidance document. COVID-19: developing drugs and biological products for treatment or prevention. May, 2020.
    1. Wong CKH, Au ICH, Lau KTK, Lau EHY, Cowling BJ, Leung GM. Real-world effectiveness of early molnupiravir or nirmatrelvir–ritonavir in hospitalised patients with COVID-19 without supplemental oxygen requirement on admission during Hong Kong's omicron BA.2 wave: a retrospective cohort study. Lancet Infect Dis. 2022 doi: 10.1016/S1473-3099(22)00507-2. published online Aug 24.

Source: PubMed

3
Abonner