Hyperimmune immunoglobulin for hospitalised patients with COVID-19 (ITAC): a double-blind, placebo-controlled, phase 3, randomised trial

ITAC (INSIGHT 013) Study Group, Mark N Polizzotto, Jacqueline Nordwall, Abdel G Babiker, Andrew Phillips, David M Vock, Nnakelu Eriobu, Vivian Kwaghe, Roger Paredes, Lourdes Mateu, Srikanth Ramachandruni, Rajeev Narang, Mamta K Jain, Susana M Lazarte, Jason V Baker, Anne E P Frosch, Garyfallia Poulakou, Konstantinos N Syrigos, Gretchen S Arnoczy, Natalie A McBride, Philip A Robinson, Farjad Sarafian, Sanjay Bhagani, Hassan S Taha, Thomas Benfield, Sean T H Liu, Anastasia Antoniadou, Jens Ulrik Stæhr Jensen, Ioannis Kalomenidis, Adityo Susilo, Prasetyo Hariadi, Tomas O Jensen Md, Jose Luis Morales-Rull, Marie Helleberg, Sreenath Meegada, Isik S Johansen, Daniel Canario, Eduardo Fernández-Cruz, Simeon Metallidis, Amish Shah, Aki Sakurai, Nikolaos G Koulouris, Robin Trotman, Amy C Weintrob, Daria Podlekareva, Usman Hadi, Kathryn M Lloyd, Birgit Thorup Røge, Sho Saito, Kelly Sweerus, Jakob J Malin, Christoph Lübbert, Jose Muñoz, Matthew J Cummings, Marcelo H Losso, Dan Turner, Kathryn Shaw-Saliba, Robin Dewar, Helene Highbarger, Perrine Lallemand, Tauseef Rehman, Norman Gerry, Dona Arlinda, Christina C Chang, Birgit Grund, Michael R Holbrook, Horace P Holley, Fleur Hudson, Laura A McNay, Daniel D Murray, Sarah L Pett, Megan Shaughnessy, Mary C Smolskis, Giota Touloumi, Mary E Wright, Mittie K Doyle, Sharon Popik, Christine Hall, Roshan Ramanathan, Huyen Cao, Elsa Mondou, Todd Willis, Joseph V Thakuria, Leman Yel, Elizabeth Higgs, Virginia L Kan, Jens D Lundgren, James D Neaton, H Clifford Lane, ITAC (INSIGHT 013) Study Group, Mark N Polizzotto, Jacqueline Nordwall, Abdel G Babiker, Andrew Phillips, David M Vock, Nnakelu Eriobu, Vivian Kwaghe, Roger Paredes, Lourdes Mateu, Srikanth Ramachandruni, Rajeev Narang, Mamta K Jain, Susana M Lazarte, Jason V Baker, Anne E P Frosch, Garyfallia Poulakou, Konstantinos N Syrigos, Gretchen S Arnoczy, Natalie A McBride, Philip A Robinson, Farjad Sarafian, Sanjay Bhagani, Hassan S Taha, Thomas Benfield, Sean T H Liu, Anastasia Antoniadou, Jens Ulrik Stæhr Jensen, Ioannis Kalomenidis, Adityo Susilo, Prasetyo Hariadi, Tomas O Jensen Md, Jose Luis Morales-Rull, Marie Helleberg, Sreenath Meegada, Isik S Johansen, Daniel Canario, Eduardo Fernández-Cruz, Simeon Metallidis, Amish Shah, Aki Sakurai, Nikolaos G Koulouris, Robin Trotman, Amy C Weintrob, Daria Podlekareva, Usman Hadi, Kathryn M Lloyd, Birgit Thorup Røge, Sho Saito, Kelly Sweerus, Jakob J Malin, Christoph Lübbert, Jose Muñoz, Matthew J Cummings, Marcelo H Losso, Dan Turner, Kathryn Shaw-Saliba, Robin Dewar, Helene Highbarger, Perrine Lallemand, Tauseef Rehman, Norman Gerry, Dona Arlinda, Christina C Chang, Birgit Grund, Michael R Holbrook, Horace P Holley, Fleur Hudson, Laura A McNay, Daniel D Murray, Sarah L Pett, Megan Shaughnessy, Mary C Smolskis, Giota Touloumi, Mary E Wright, Mittie K Doyle, Sharon Popik, Christine Hall, Roshan Ramanathan, Huyen Cao, Elsa Mondou, Todd Willis, Joseph V Thakuria, Leman Yel, Elizabeth Higgs, Virginia L Kan, Jens D Lundgren, James D Neaton, H Clifford Lane

Abstract

Background: Passive immunotherapy using hyperimmune intravenous immunoglobulin (hIVIG) to SARS-CoV-2, derived from recovered donors, is a potential rapidly available, specific therapy for an outbreak infection such as SARS-CoV-2. Findings from randomised clinical trials of hIVIG for the treatment of COVID-19 are limited.

Methods: In this international randomised, double-blind, placebo-controlled trial, hospitalised patients with COVID-19 who had been symptomatic for up to 12 days and did not have acute end-organ failure were randomly assigned (1:1) to receive either hIVIG or an equivalent volume of saline as placebo, in addition to remdesivir, when not contraindicated, and other standard clinical care. Randomisation was stratified by site pharmacy; schedules were prepared using a mass-weighted urn design. Infusions were prepared and masked by trial pharmacists; all other investigators, research staff, and trial participants were masked to group allocation. Follow-up was for 28 days. The primary outcome was measured at day 7 by a seven-category ordinal endpoint that considered pulmonary status and extrapulmonary complications and ranged from no limiting symptoms to death. Deaths and adverse events, including organ failure and serious infections, were used to define composite safety outcomes at days 7 and 28. Prespecified subgroup analyses were carried out for efficacy and safety outcomes by duration of symptoms, the presence of anti-spike neutralising antibodies, and other baseline factors. Analyses were done on a modified intention-to-treat (mITT) population, which included all randomly assigned participants who met eligibility criteria and received all or part of the assigned study product infusion. This study is registered with ClinicalTrials.gov, NCT04546581.

Findings: From Oct 8, 2020, to Feb 10, 2021, 593 participants (n=301 hIVIG, n=292 placebo) were enrolled at 63 sites in 11 countries; 579 patients were included in the mITT analysis. Compared with placebo, the hIVIG group did not have significantly greater odds of a more favourable outcome at day 7; the adjusted OR was 1·06 (95% CI 0·77-1·45; p=0·72). Infusions were well tolerated, although infusion reactions were more common in the hIVIG group (18·6% vs 9·5% for placebo; p=0·002). The percentage with the composite safety outcome at day 7 was similar for the hIVIG (24%) and placebo groups (25%; OR 0·98, 95% CI 0·66-1·46; p=0·91). The ORs for the day 7 ordinal outcome did not vary for subgroups considered, but there was evidence of heterogeneity of the treatment effect for the day 7 composite safety outcome: risk was greater for hIVIG compared with placebo for patients who were antibody positive (OR 2·21, 95% CI 1·14-4·29); for patients who were antibody negative, the OR was 0·51 (0·29-0·90; pinteraction=0·001).

Interpretation: When administered with standard of care including remdesivir, SARS-CoV-2 hIVIG did not demonstrate efficacy among patients hospitalised with COVID-19 without end-organ failure. The safety of hIVIG might vary by the presence of endogenous neutralising antibodies at entry.

Funding: US National Institutes of Health.

Conflict of interest statement

Declaration of interests MNP reports grants from University of Minnesota (Minneapolis, MN, USA) during the conduct of the study, grants from National Institutes of Health (NIH) during the conduct of the study, Gilead Sciences, ViiV, Celgene, and Janssen Pharmaceuticals outside the submitted work. AGB reports grants from University of Minnesota during the conduct of the study, grants from UK Research and Innovation (UKRI) outside of the submitted work. MKJ reports donation of trial medications from Regeneron Pharmaceuticals, Janssen Pharmaceuticals, and from Merck; and grants, personal fees, and donation of trial medications from Gilead Sciences, outside the submitted work. SLP reports grants from University of Minnesota during the conduct of the study, European and Developing Countries Clinical Trials Partnership, UKRI, Academy of Medical Sciences, ViiV Healthcare, Medical Research Council, and Gilead Sciences outside the submitted work. MKD reports being an employee of CSL Behring. SP reports being an employee of CSL Behring. CH reports being an employee of Emergent; Ramanathan reports being an employee of Emergent. HC reports being an employee of Gilead Sciences. EM reports being an employee of Grifols. TW reports being an employee of Grifols. JVT reports being an employee of Takeda. LY reports being an employee of Takeda. JDN reports grants from NIH during the conduct of the study. All other members of the writing group declare no competing interests.

Copyright © 2022 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
Trial profile ITT=intention to treat.
Figure 2
Figure 2
Clinical efficacy of hIVIG (A) Clinical status at day 7. (B) Change in clinical status from baseline to day 7. (C) Time to discharge or most favourable ordinal category. hIVIG=hyperimmune intravenous immunoglobulin. NIHSS=National Institute of Health Stroke Scale. ECMO=Extra-corporeal membrane oxygenation. OR=odds ratio.
Figure 3
Figure 3
Subgroup analyses for primary endpoint Data are odds ratio (95% CI), unless stated otherwise. Score=mean score, 7=death, and 1=return to normal activities with minimal or no symptoms. hIVIV=hyperimmune intravenous immunoglobulin.*p value for interaction between subgroup and treatment group. In addition to heterogeneity across the 4 days since symptom onset categories, a test for linear trend was conducted (p=0·99). †Antibody data available for 272 hIVIG and 260 placebo participants.

References

    1. Beigel JH, Tomashek KM, Dodd LE, et al. Remdesivir for the treatment of COVID-19 - final report. N Engl J Med. 2020;383:1813–1826.
    1. Horby P, Lim WS, Emberson JR, et al. Dexamethasone in hospitalized patients with COVID-19. N Engl J Med. 2021;384:693–704.
    1. Horby PW, Pessoa-Amorim G, Peto L, et al. Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): preliminary results of a randomised, controlled, open-label, platform trial. Lancet. 2021;97:637–645.
    1. Gordon AC, Mouncey PR, Al-Beidh F, et al. Interleukin-6 receptor antagonists in critically ill patients with COVID-19. N Engl J Med. 2021;384:1491–1502.
    1. Long QX, Liu BZ, Deng HJ, et al. Antibody responses to SARS-CoV-2 in patients with COVID-19. Nat Med. 2020;26:845–848.
    1. Gharbharan A, Jordans CCE, GeurtsvanKessel C, et al. Effects of potent neutralizing antibodies from convalescent plasma in patients hospitalized for severe SARS-CoV-2 infection. Nat Commun. 2021;12
    1. Garcia-Beltran WF, Lam EC, Astudillo MG, et al. COVID-19-neutralizing antibodies predict disease severity and survival. Cell. 2021;184:476–488.
    1. Gottlieb RL, Nirula A, Chen P, et al. Effect of bamlanivimab as monotherapy or in combination with etesevimab on viral load in patients with mild to moderate COVID-19: a randomized clinical trial. JAMA. 2021;325:632–644.
    1. Weinreich DM, Sivapalasingam S, Norton T, et al. REGN-COV2, a neutralizing antibody cocktail, in outpatients with COVID-19. N Engl J Med. 2021;384:238–251.
    1. Lundgren JD, Grund B, Barkauskas CE, et al. A neutralizing monoclonal antibody for hospitalized patients with COVID-19. N Engl J Med. 2021;384:905–914.
    1. RECOVERY Collaborative Group. Horby PW, Mafham M, et al. Casirivimab and imdevimab in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial. medRxiv. 2021 published online June 16. (preprint).
    1. ACTIV-3/TICO Bamlanivimab Study Group Responses to a neutralizing monoclonal antibody for hospitalized patients with COVID-19 according to baseline antibody and antigen levels: a randomized controlled trial. Ann Intern Med. 2021 doi: 10.7326/M21-3507. published online Dec 21.
    1. Enria DA, Briggiler AM, Sánchez Z. Treatment of Argentine hemorrhagic fever. Antiviral Res. 2008;78:132–139.
    1. Libster R, Pérez Marc G, Wappner D, et al. Early high-titer plasma therapy to prevent severe COVID-19 in older adults. N Engl J Med. 2021;384:610–618.
    1. Korley FK, Durkalski-Mauldin V, Yeatts SD, et al. Early convalescent plasma for high-risk outpatients with COVID-19. N Engl J Med. 2021;385:1951–1960.
    1. Joyner MJ, Carter RE, Senefeld JW, et al. Convalescent plasma antibody levels and the risk of death from COVID-19. N Engl J Med. 2021;384:1015–1027.
    1. Simonovich VA, Burgos Pratx LD, Scibona P, et al. A randomized trial of convalescent plasma in COVID-19 severe pneumonia. N Engl J Med. 2021;384:619–629.
    1. Abani O, Abbas A, Abbas F, et al. Convalescent plasma in patients admitted to hospital with COVID-19 (RECOVERY): a randomised controlled, open-label, platform trial. Lancet. 2021;397:2049–2059.
    1. Bégin P, Callum J, Jamula E, et al. Convalescent plasma for hospitalized patients with COVID-19: an open-label, randomized controlled trial. Nat Med. 2021;27:2012–2024.
    1. Avendaño-Solá C, Ramos-Martínez A, Muñez-Rubio E, et al. A multicenter randomized open-label clinical trial for convalescent plasma in patients hospitalized with COVID-19 pneumonia. J Clin Invest. 2021;131
    1. Janiaud P, Axfors C, Schmitt AM, et al. Association of convalescent plasma treatment with clinical outcomes in patients with COVID-19: a systematic review and meta-analysis. JAMA. 2021;325:1185–1195.
    1. Vandeberg P, Cruz M, Diez JM, et al. Production of anti-SARS-CoV-2 hyperimmune globulin from convalescent plasma. Transfusion. 2021;61:1705–1709.
    1. Ali S, Uddin SM, Shalim E, et al. Hyperimmune anti-COVID-19 IVIG (C-IVIG) treatment in severe and critical COVID-19 patients: a phase I/II randomized control trial. EClinicalMedicine. 2021;36
    1. Lopardo G, Belloso WH, Nannini E, et al. RBD-specific polyclonal F(ab')2 fragments of equine antibodies in patients with moderate to severe COVID-19 disease: a randomized, multicenter, double-blind, placebo-controlled, adaptive phase 2/3 clinical trial. EClinicalMedicine. 2021;34
    1. Zhao W. Mass weighted urn design—a new randomization algorithm for unequal allocations. Contemp Clin Trials. 2015;43:209–216.
    1. Davey RT, Jr, Fernández-Cruz E, Markowitz N, et al. Anti-influenza hyperimmune intravenous immunoglobulin for adults with influenza A or B infection (FLU-IVIG): a double-blind, randomised, placebo-controlled trial. Lancet Respir Med. 2019;7:951–963.
    1. Marshall JC, Murthy S, Diaz J, et al. A minimal common outcome measure set for COVID-19 clinical research. Lancet Infect Dis. 2020;20:e192–e197.
    1. Fine JP, Gray RJ. A proportional hazards model for the subdistribution of a competing risk. J Am Stat Assoc. 1999;94:496–509.
    1. Zhou B, Latouche A, Rocha V, Fine J. Competing risks regression for stratified data. Biometrics. 2011;67:661–670.
    1. Aalen OO, Johansen S. An empirical transition matrix for non-homogeneous Markov chains based on censored observations. Scand J Stat. 1978;5:141–150.
    1. Griffin DO, Brennan-Rieder D, Ngo B, et al. The importance of understanding the stages of COVID-19 in treatment and trials. AIDS Rev. 2021;23:40–47.
    1. Bastard P, Rosen LB, Zhang Q, et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science. 2020;370
    1. Arvin AM, Fink K, Schmid MA, et al. A perspective on potential antibody-dependent enhancement of SARS-CoV-2. Nature. 2020;584:353–363.
    1. Lee WS, Wheatley AK, Kent SJ, DeKosky BJ. Antibody-dependent enhancement and SARS-CoV-2 vaccines and therapies. Nat Microbiol. 2020;5:1185–1191.
    1. Wang P, Nair MS, Liu L, et al. Antibody resistance of SARS-CoV-2 variants B.1.351 and B.1.1.7. Nature. 2021;593:130–135.
    1. Vanderven HA, Wragg K, Ana-Sosa-Batiz F, et al. Anti-influenza hyperimmune immunoglobulin enhances Fc-functional antibody immunity during human influenza infection. J Infect Dis. 2018;218:1383–1393.

Source: PubMed

3
S'abonner