Anti-C5a antibody IFX-1 (vilobelimab) treatment versus best supportive care for patients with severe COVID-19 (PANAMO): an exploratory, open-label, phase 2 randomised controlled trial

Alexander P J Vlaar, Sanne de Bruin, Matthias Busch, Sjoerd A M E G Timmermans, Ingeborg E van Zeggeren, Rutger Koning, Liora Ter Horst, Esther B Bulle, Frank E H P van Baarle, Marcel C G van de Poll, E Marleen Kemper, Iwan C C van der Horst, Marcus J Schultz, Janneke Horn, Frederique Paulus, Lieuwe D Bos, W Joost Wiersinga, Martin Witzenrath, Simon Rueckinger, Korinna Pilz, Matthijs C Brouwer, Ren-Feng Guo, Leo Heunks, Pieter van Paassen, Niels C Riedemann, Diederik van de Beek, Alexander P J Vlaar, Sanne de Bruin, Matthias Busch, Sjoerd A M E G Timmermans, Ingeborg E van Zeggeren, Rutger Koning, Liora Ter Horst, Esther B Bulle, Frank E H P van Baarle, Marcel C G van de Poll, E Marleen Kemper, Iwan C C van der Horst, Marcus J Schultz, Janneke Horn, Frederique Paulus, Lieuwe D Bos, W Joost Wiersinga, Martin Witzenrath, Simon Rueckinger, Korinna Pilz, Matthijs C Brouwer, Ren-Feng Guo, Leo Heunks, Pieter van Paassen, Niels C Riedemann, Diederik van de Beek

Abstract

Background: Severe COVID-19 is characterised by inflammation and coagulation in the presence of complement system activation. We aimed to explore the potential benefit and safety of selectively blocking the anaphylatoxin and complement protein C5a with the monoclonal antibody IFX-1 (vilobelimab), in patients with severe COVID-19.

Methods: We did an exploratory, open-label, randomised phase 2 trial (part of the adaptive phase 2/3 PANAMO trial) of intravenous IFX-1 in adults with severe COVID-19 at three academic hospitals in the Netherlands. Eligibility criteria were age 18 years or older; severe pneumonia with pulmonary infiltrates consistent with pneumonia, a clinical history of severe shortness of breath within the past 14 days, or a need for non-invasive or invasive ventilation; severe disease defined as a ratio of partial pressure of arterial oxygen to fractional concentration of oxygen in inspired air (PaO2/FiO2) between 100 mm Hg and 250 mm Hg in the supine position; and severe acute respiratory syndrome coronavirus 2 infection confirmed by RT-PCR. Patients were randomly assigned 1:1 to receive IFX-1 (up to seven doses of 800 mg intravenously) plus best supportive care (IFX-1 group) or best supportive care only (control group). The primary outcome was the percentage change in PaO2/FiO2 in the supine position between baseline and day 5. Mortality at 28 days and treatment-emergent and serious adverse events were key secondary outcomes. The primary analysis was done in the intention-to-treat population and safety analyses were done in all patients according to treatment received. This trial is registered at ClinicalTrials.gov (NCT04333420).

Findings: Between March 31 and April 24, 2020, 30 patients were enrolled and randomly assigned to the IFX-1 group (n=15) or the control group (n=15). During the study it became clear that several patients could not be assessed regularly in the supine position because of severe hypoxaemia. It was therefore decided to focus on all PaO2/FiO2 assessments (irrespective of position). At day 5 after randomisation, the mean PaO2/FiO2 (irrespective of position) was 158 mm Hg (SD 63; range 84-265) in the IFX-1 group and 189 mm Hg (89; 71-329) in the control group. Analyses of the least squares mean relative change in PaO2/FiO2 at day 5 showed no differences between treatment groups (17% change in the IFX-1 group vs 41% in the control group; difference -24% [95% CI -58 to 9], p=0·15. Kaplan-Meier estimates of mortality by 28 days were 13% (95% CI 0-31) for the IFX-1 group and 27% (4-49) for the control group (adjusted hazard ratio for death 0·65 [95% CI 0·10-4·14]). The frequency of serious adverse events were similar between groups (nine [60%] in the IFX-1 group vs seven [47%] in the control group) and no deaths were considered related to treatment assignment. However, a smaller proportion of patients had pulmonary embolisms classed as serious in the IFX-1 group (two [13%]) than in the control group (six [40%]). Infections classed as serious were reported in three (20%) patients in the IFX-1 group versus five (33%) patients in the control group.

Interpretation: In this small exploratory phase 2 part of the PANAMO trial, C5a inhibition with IFX-1 appears to be safe in patients with severe COVID-19. The secondary outcome results in favour of IFX-1 are preliminary because the study was not powered on these endpoints, but they support the investigation of C5a inhibition with IFX-1 in a phase 3 trial using 28-day mortality as the primary endpoint.

Funding: InflaRx.

© 2020 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
Trial profile
Figure 2
Figure 2
Shift plots for eGFR and lymphocyte concentrations and least squares mean plots for relative changes in selected outcome parameters Relative change in mean PaO2/FiO2 (A) and eGFR (B). eGFR in the IFX-1 group (C) and control group (D); lymphocyte counts in the IFX-1 group (E) and control group (F). Relative change in mean lactate dehydrogenase (G) and D-dimers (H). Error bars show 95% CI. Units for eGFR are mL/min per 1·73 m2. eGFR=estimated glomerular filtration rate. PaO2/FiO2=ratio of partial pressure of arterial oxygen to fractional concentration of oxygen in inspired air.

References

    1. Zhou F, Yu T, Du R. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054–1062.
    1. Guan WJ, Ni ZY, Hu Y. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382:1708–1720.
    1. Grasselli G, Zangrillo A, Zanella A. Baseline characteristics and outcomes of 1591 patients infected with SARS-CoV-2 admitted to ICUs of the Lombardy region, Italy. JAMA. 2020;323:1574–1581.
    1. Tay MZ, Poh CM, Rénia L, MacAry PA, Ng LFP. The trinity of COVID-19: immunity, inflammation and intervention. Nat Rev Immunol. 2020;20:363–374.
    1. Li H, Liu L, Zhang D. SARS-CoV-2 and viral sepsis: observations and hypotheses. Lancet. 2020;395:1517–1520.
    1. Gao T, Hu M, Zhang X. Highly pathogenic coronavirus N protein aggravates lung injury by MASP-2-mediated complement over-activation. MedRxiv. 2020 doi: 10.1101/2020.03.29.20041962. published online June 18. (preprint)
    1. Diao B, Feng Z, Wang C. Human kidney is a target for novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. MedRxiv. 2020 doi: 10.1101/2020.03.04.20031120. published online March 6. (preprint)
    1. Cugno M, Meroni PL, Gualtierotti R. Complement activation in patients with COVID-19: a novel therapeutic target. J Allergy Clin Immunol. 2020;146:215–217.
    1. Carvelli J, Demaria O, Vély F. Association of COVID-19 inflammation with activation of the C5a-C5aR1 axis. Nature. 2020 doi: 10.1038/s41586-020-2600-6. published online July 29.
    1. Seshan SV, Franzke CW, Redecha P, Monestier M, Mackman N, Girardi G. Role of tissue factor in a mouse model of thrombotic microangiopathy induced by antiphospholipid antibodies. Blood. 2009;114:1675–1683.
    1. Kambas K, Markiewski MM, Pneumatikos IA. C5a and TNF-alpha up-regulate the expression of tissue factor in intra-alveolar neutrophils of patients with the acute respiratory distress syndrome. J Immunol. 2008;180:7368–7375.
    1. Ritis K, Doumas M, Mastellos D. A novel C5a receptor-tissue factor cross-talk in neutrophils links innate immunity to coagulation pathways. J Immunol. 2006;177:4794–4802.
    1. Wang R, Xiao H, Guo R, Li Y, Shen B. The role of C5a in acute lung injury induced by highly pathogenic viral infections. Emerg Microbes Infect. 2015;4:e28.
    1. Jiang Y, Zhao G, Song N. Blockade of the C5a-C5aR axis alleviates lung damage in hDPP4-transgenic mice infected with MERS-CoV. Emerg Microbes Infect. 2018;7:77.
    1. Campbell CM, Kahwash R. Will complement inhibition be the new target in treating COVID-19-related systemic thrombosis? Circulation. 2020;141:1739–1741.
    1. Risitano AM, Mastellos DC, Huber-Lang M. Complement as a target in COVID-19? Nat Rev Immunol. 2020;20:343–344.
    1. Sun S, Zhao G, Liu C. Treatment with anti-C5a antibody improves the outcome of H7N9 virus infection in African green monkeys. Clin Infect Dis. 2015;60:586–595.
    1. Mastaglio S, Ruggeri A, Risitano AM. The first case of COVID-19 treated with the complement C3 inhibitor AMY-101. Clin Immunol. 2020;215
    1. Riedemann NC, Habel M, Ziereisen J. Controlling the anaphylatoxin C5a in diseases requires a specifically targeted inhibition. Clin Immunol. 2017;180:25–32.
    1. Mukhopadhyay S, Johnson TA, Duru N. Fibrinolysis and inflammation in venous thrombus resolution. Front Immunol. 2019;10
    1. Helms J, Tacquard C, Severac F. High risk of thrombosis in patients with severe SARS-CoV-2 infection: a multicenter prospective cohort study. Intensive Care Med. 2020;46:1089–1098.
    1. Magro C, Mulvey JJ, Berlin D. Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID-19 infection: a report of five cases. Transl Res. 2020;220:1–13.
    1. Ackermann M, Verleden SE, Kuehnel M. Pulmonary vascular endothelialitis, thrombosis, and angiogenesis in COVID-19. N Engl J Med. 2020;383:120–128.
    1. Noris M, Benigni A, Remuzzi G. The case of complement activation in COVID-19 multiorgan impact. Kidney Int. 2020;98:314–322.
    1. Ikeda K, Nagasawa K, Horiuchi T, Tsuru T, Nishizaka H, Niho Y. C5a induces tissue factor activity on endothelial cells. Thromb Haemost. 1997;77:394–398.
    1. Wojta J, Huber K, Valent P. New aspects in thrombotic research: complement induced switch in mast cells from a profibrinolytic to a prothrombotic phenotype. Pathophysiol Haemost Thromb. 2003;33:438–441.
    1. Yan L, Zhang H-T, Goncalves J. An interpretable mortality prediction model for COVID-19 patients. Nat Mach Int. 2020;2:283–288.
    1. Wu C, Chen X, Cai Y. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern Med. 2020;180:1–11.

Source: PubMed

3
購読する