Randomized, double-blind, controlled trial of human anti-LIGHT monoclonal antibody in COVID-19 acute respiratory distress syndrome

David S Perlin, Garry A Neil, Colleen Anderson, Inbal Zafir-Lavie, Shane Raines, Carl F Ware, H Jeffrey Wilkins, David S Perlin, Garry A Neil, Colleen Anderson, Inbal Zafir-Lavie, Shane Raines, Carl F Ware, H Jeffrey Wilkins

Abstract

BACKGROUNDSevere coronavirus disease 2019 (COVID-19) is associated with a dysregulated immune response, which can result in cytokine-release syndrome and acute respiratory distress syndrome (ARDS). Patients with COVID-19-associated ARDS have elevated free serum levels of the cytokine lymphotoxin-like inducible protein that competes with glycoprotein D for herpesvirus entry on T cells (LIGHT; also known as TNFSF14). Such patients may benefit from LIGHT-neutralization therapy.METHODSThis randomized, double-blind, multicenter, proof-of-concept trial enrolled adults hospitalized with COVID-19-associated pneumonia and mild to moderate ARDS. Patients received standard of care plus a single dose of a human LIGHT-neutralizing antibody (CERC-002) or placebo. The primary endpoint was the proportion of patients receiving CERC-002 who remained alive and free of respiratory failure through day 28. Safety was assessed via adverse event monitoring.RESULTSFor most of the 83 enrolled patients, standard of care included systemic corticosteroids (88.0%) or remdesivir (57.8%). A higher proportion of patients remained alive and free of respiratory failure through day 28 after receiving CERC-002 (83.9%) versus placebo (64.5%; P = 0.044), including in patients 60 years of age or older (76.5% vs. 47.1%, respectively; P = 0.042). Mortality rates were 7.7% (CERC-002) and 14.3% (placebo) on day 28 and 10.8% and 22.5%, respectively, on day 60. Treatment-emergent adverse events were less frequent with CERC-002 than placebo.CONCLUSIONFor patients with COVID-19-associated ARDS, adding CERC-002 to standard-of-care treatment reduces LIGHT levels and might reduce the risk of respiratory failure and death.TRIAL REGISTRATIONClinicalTrials.gov NCT04412057.FUNDINGAvalo Therapeutics.

Keywords: Adaptive immunity; COVID-19; Clinical Trials; Cytokines; Respiration.

Conflict of interest statement

Conflict of interest: GAN, CA, and HJW are employees of Avalo Therapeutics, which funded the study reported herein. IZL and SR are paid consultants to Avalo Therapeutics. CFW has received royalty payments from the La Jolla Institute and consulting fees and stock options from Avalo Therapeutics.

Figures

Figure 1. Enrollment and randomization.
Figure 1. Enrollment and randomization.
Figure 2. Efficacy of CERC-002 in COVID-19…
Figure 2. Efficacy of CERC-002 in COVID-19 patients.
Percentage of patients alive and free of respiratory failure through 28 days after treatment is presented. Analysis was performed for overall (n = 62) patients, and separately for subgroups of patients under the age of 60 (n = 34) and age 60 or above (n = 28). One-sided P values were calculated using the Wald χ2 test.
Figure 3. Serum free-LIGHT levels (pg/mL) over…
Figure 3. Serum free-LIGHT levels (pg/mL) over treatment period.
Mean free-LIGHT levels were comparable at baseline across treatment groups. Data represent mean + SD.

References

    1. Parr J. Pneumonia in China: lack of information raises concerns among Hong Kong health workers. BMJ. 2020;368:m56.
    1. Hui DS, et al. The continuing 2019-nCoV epidemic threat of novel coronaviruses to global health — The latest 2019 novel coronavirus outbreak in Wuhan, China. Int J Infect Dis. 2020;91:264–266. doi: 10.1016/j.ijid.2020.01.009.
    1. World Health Organization. WHO Coronavirus (COVID-19) Dashboard. Updated December 7, 2021. Accessed December 7, 2021.
    1. Fajgenbaum DC, June CH. Cytokine storm. N Engl J Med. 2020;383(23):2255–2273. doi: 10.1056/NEJMra2026131.
    1. Wang W, et al. Definition and risks of cytokine release syndrome in 11 critically ill COVID-19 patients with pneumonia: analysis of disease characteristics. J Infect Dis. 2020;222(9):1444–1451. doi: 10.1093/infdis/jiaa387.
    1. Huang C, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395(10223):497–506. doi: 10.1016/S0140-6736(20)30183-5.
    1. Chen N, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020;395(10223):507–513. doi: 10.1016/S0140-6736(20)30211-7.
    1. Challen R, et al. Risk of mortality in patients infected with SARS-CoV-2 variant of concern 202012/1: matched cohort study. BMJ. 2021;372:n579.
    1. Hojyo S, et al. How COVID-19 induces cytokine storm with high mortality. Inflamm Regen. 2020;40:37.
    1. Sanders JM, et al. Pharmacologic treatments for coronavirus disease 2019 (COVID-19): a review. JAMA. 2020;323(18):1824–1836.
    1. Izda V, et al. COVID-19: a review of therapeutic strategies and vaccine candidates. Clin Immunol. 2021;222:108634.
    1. World Health Organization. COVID-19: Landscape of novel coronavirus candidate vaccine development worldwide. Updated December 7, 2021. Accessed December 7, 2021.
    1. Wise J. Covid-19: New coronavirus variant is identified in UK. BMJ. 2020;371:m4857.
    1. Frampton D, et al. Genomic characteristics and clinical effect of the emergent SARS-CoV-2 B.1.1.7 lineage in London, UK: a whole-genome sequencing and hospital-based cohort study. Lancet Infect Dis. 2021;21(9):1246–1256. doi: 10.1016/S1473-3099(21)00170-5.
    1. Wibmer CK, et al. SARS-CoV-2 501Y.V2 escapes neutralization by South African COVID-19 donor plasma. Nat Med. 2021;27(4):622–625. doi: 10.1038/s41591-021-01285-x.
    1. World Health Organization. Emergencies preparedness, response: SARS-CoV-2 variants. Updated December 7, 2021. Accessed December 7, 2021.
    1. Koyama T, et al. Emergence of drift variants that may affect COVID-19 vaccine development and antibody treatment. Pathogens. 2020;9(5):324. doi: 10.3390/pathogens9050324.
    1. Wang P, et al. Antibody resistance of SARS-CoV-2 variants B.1.351 and B.1.1.7. Nature. 2021;593(7857):130–135. doi: 10.1038/s41586-021-03398-2.
    1. Shimabukuro-Vornhagen A, et al. Cytokine release syndrome. J Immunother Cancer. 2018;6(1):56. doi: 10.1186/s40425-018-0343-9.
    1. Remap-Cap Investigators, et al. Interleukin-6 receptor antagonists in critically ill patients with Covid-19. N Engl J Med. 2021;384(16):1491–1502. doi: 10.1056/NEJMoa2100433.
    1. Salama C, et al. Tocilizumab in patients hospitalized with Covid-19 pneumonia. N Engl J Med. 2021;384(1):20–30. doi: 10.1056/NEJMoa2030340.
    1. Campochiaro C, et al. Efficacy and safety of tocilizumab in severe COVID-19 patients: a single-centre retrospective cohort study. Eur J Intern Med. 2020;76:43–49. doi: 10.1016/j.ejim.2020.05.021.
    1. Bischof E, et al. The potential of rapalogs to enhance resilience against SARS-CoV-2 infection and reduce the severity of COVID-19. Lancet Healthy Longev. 2021;2(2):e105–e111. doi: 10.1016/S2666-7568(20)30068-4.
    1. Magro G. COVID-19: Review on latest available drugs and therapies against SARS-CoV-2. Coagulation and inflammation cross-talking. Virus Res. 2020;286:198070.
    1. Ward-Kavanagh LK, et al. The TNF receptor superfamily in co-stimulating and co-inhibitory responses. Immunity. 2016;44(5):1005–1019. doi: 10.1016/j.immuni.2016.04.019.
    1. Herro R, Croft M. The control of tissue fibrosis by the inflammatory molecule LIGHT (TNF superfamily member 14) Pharmacol Res. 2016;104:151–155. doi: 10.1016/j.phrs.2015.12.018.
    1. Doherty TA, et al. The tumor necrosis factor family member LIGHT is a target for asthmatic airway remodeling. Nat Med. 2011;17(5):596–603. doi: 10.1038/nm.2356.
    1. Desai P, et al. The TNF superfamily molecule LIGHT promotes the generation of circulating and lung-resident memory CD8 T cells following an acute respiratory virus infection. J Immunol. 2018;200(8):2894–2904. doi: 10.4049/jimmunol.1701499.
    1. Wang J, et al. The critical role of LIGHT in promoting intestinal inflammation and Crohn’s disease. J Immunol. 2005;174(12):8173–8182. doi: 10.4049/jimmunol.174.12.8173.
    1. del Rio ML, et al. LIGHT/HVEM/LTbetaR interaction as a target for the modulation of the allogeneic immune response in transplantation. Am J Transplant. 2013;13(3):541–551. doi: 10.1111/ajt.12089.
    1. Perlin DS, et al. Levels of the TNF-related cytokine LIGHT increase in hospitalized COVID-19 patients with cytokine release syndrome and ARDS. mSphere. 2020;5(4):e00699–20.
    1. Arunachalam PS, et al. Systems biological assessment of immunity to mild versus severe COVID-19 infection in humans. Science. 2020;369(6508):1210–1220. doi: 10.1126/science.abc6261.
    1. Haljasmagi L, et al. Longitudinal proteomic profiling reveals increased early inflammation and sustained apoptosis proteins in severe COVID-19. Sci Rep. 2020;10(1):20533. doi: 10.1038/s41598-020-77525-w.
    1. Ramana CV. Profiling transcription factor sub-networks in type I interferon signaling and in response to SARS-CoV-2 infection [preprint]. Posted on bioRxiv March 5, 2021.
    1. Henderson LA, et al. On the alert for cytokine storm: immunopathology in COVID-19. Arthritis Rheumatol. 2020;72(7):1059–1063. doi: 10.1002/art.41285.
    1. Recovery Collaborative Group. et al. Dexamethasone in hospitalized patients with Covid-19. N Engl J Med. 2021;384(8):693–704. doi: 10.1056/NEJMoa2021436.
    1. WHO Rapid Evidence Appraisal for COVID-19 Therapies Working Group. et al. Association between administration of systemic corticosteroids and mortality among critically ill patients with COVID-19: A meta-analysis. JAMA. 2020;324(13):1330–1341. doi: 10.1001/jama.2020.17023.
    1. Angus DC, et al. Effect of hydrocortisone on mortality and organ support in patients with severe COVID-19: the REMAP-CAP COVID-19 Corticosteroid Domain Randomized Clinical Trial. JAMA. 2020;324(13):1317–1329. doi: 10.1001/jama.2020.17022.
    1. Dequin PF, et al. Effect of hydrocortisone on 21-day mortality or respiratory support among critically ill patients with COVID-19: a randomized clinical trial. JAMA. 2020;324(13):1298–1306. doi: 10.1001/jama.2020.16761.
    1. Tomazini BM, et al. Effect of dexamethasone on days alive and ventilator-free in patients with moderate or severe acute respiratory distress syndrome and COVID-19: the CoDEX randomized clinical trial. JAMA. 2020;324(13):1307–1316. doi: 10.1001/jama.2020.17021.
    1. Shuto H, et al. A systematic review of corticosteroid treatment for noncritically ill patients with COVID-19. Sci Rep. 2020;10(1):20935. doi: 10.1038/s41598-020-78054-2.
    1. Cavalli G, Dagna L. The right place for IL-1 inhibition in COVID-19. Lancet Respir Med. 2021;9(3):223–224. doi: 10.1016/S2213-2600(21)00035-7.
    1. Lang FM, et al. GM-CSF-based treatments in COVID-19: reconciling opposing therapeutic approaches. Nat Rev Immunol. 2020;20(8):507–514. doi: 10.1038/s41577-020-0357-7.
    1. Pang J, et al. Efficacy and tolerability of bevacizumab in patients with severe Covid-19. Nat Commun. 2021;12(1):814. doi: 10.1038/s41467-021-21085-8.
    1. Mortarini R, et al. Constitutive expression and costimulatory function of LIGHT/TNFSF14 on human melanoma cells and melanoma-derived microvesicles. Cancer Res. 2005;65(8):3428–3436. doi: 10.1158/0008-5472.CAN-04-3239.
    1. Sonar S, Lal G. Role of tumor necrosis factor superfamily in neuroinflammation and autoimmunity. Front Immunol. 2015;6(364):364.
    1. Schwarz BT, et al. LIGHT signals directly to intestinal epithelia to cause barrier dysfunction via cytoskeletal and endocytic mechanisms. Gastroenterology. 2007;132(7):2383–2394. doi: 10.1053/j.gastro.2007.02.052.
    1. Edwards JP, et al. Biochemical and functional characterization of three activated macrophage populations. J Leukoc Biol. 2006;80(6):1298–1307. doi: 10.1189/jlb.0406249.
    1. Channappanavar R, Perlman S. Pathogenic human coronavirus infections: causes and consequences of cytokine storm and immunopathology. Semin Immunopathol. 2017;39(5):529–539. doi: 10.1007/s00281-017-0629-x.
    1. Xu W, et al. Transcriptome sequencing identifies novel immune response genes highly related to the severity of human adenovirus type 55 infection. Front Microbiol. 2019;10:130.
    1. Rodrigues KB, et al. Innate immune stimulation of whole blood reveals IFN-1 hyper-responsiveness in type 1 diabetes. Diabetologia. 2020;63(8):1576–1587. doi: 10.1007/s00125-020-05179-4.

Source: PubMed

3
Suscribir