A phase 1 trial of lipid-encapsulated mRNA encoding a monoclonal antibody with neutralizing activity against Chikungunya virus

Allison August, Husain Z Attarwala, Sunny Himansu, Shiva Kalidindi, Sophia Lu, Rolando Pajon, Shu Han, Jean-Michel Lecerf, Joanne E Tomassini, Marjie Hard, Leon M Ptaszek, James E Crowe, Tal Zaks, Allison August, Husain Z Attarwala, Sunny Himansu, Shiva Kalidindi, Sophia Lu, Rolando Pajon, Shu Han, Jean-Michel Lecerf, Joanne E Tomassini, Marjie Hard, Leon M Ptaszek, James E Crowe, Tal Zaks

Abstract

Chikungunya virus (CHIKV) infection causes acute disease characterized by fever, rash and arthralgia, which progresses to severe and chronic arthritis in up to 50% of patients. Moreover, CHIKV infection can be fatal in infants or immunocompromised individuals and has no approved therapy or prevention. This phase 1, first-in-human, randomized, placebo-controlled, proof-of-concept trial conducted from January 2019 to June 2020 evaluated the safety and pharmacology of mRNA-1944, a lipid nanoparticle-encapsulated messenger RNA encoding the heavy and light chains of a CHIKV-specific monoclonal neutralizing antibody, CHKV-24 ( NCT03829384 ). The primary outcome was to evaluate the safety and tolerability of escalating doses of mRNA-1944 administered via intravenous infusion in healthy participants aged 18-50 years. The secondary objectives included determination of the pharmacokinetics of mRNA encoding for CHKV-24 immunoglobulin heavy and light chains and ionizable amino lipid component and the pharmacodynamics of mRNA-1944 as assessed by serum concentrations of mRNA encoding for CHKV-24 immunoglobulin G (IgG), plasma concentrations of ionizable amino lipid and serum concentrations of CHKV-24 IgG. Here we report the results of a prespecified interim analysis of 38 healthy participants who received intravenous single doses of mRNA-1944 or placebo at 0.1, 0.3 and 0.6 mg kg-1, or two weekly doses at 0.3 mg kg-1. At 12, 24 and 48 h after single infusions, dose-dependent levels of CHKV-24 IgG with neutralizing activity were observed at titers predicted to be therapeutically relevant concentrations (≥1 µg ml-1) across doses that persisted for ≥16 weeks at 0.3 and 0.6 mg kg-1 (mean t1/2 approximately 69 d). A second 0.3 mg kg-1 dose 1 week after the first increased CHKV-24 IgG levels 1.8-fold. Adverse effects were mild to moderate in severity, did not worsen with a second mRNA-1944 dose and none were serious. To our knowledge, mRNA-1944 is the first mRNA-encoded monoclonal antibody showing in vivo expression and detectable ex vivo neutralizing activity in a clinical trial and may offer a treatment option for CHIKV infection. Further evaluation of the potential therapeutic use of mRNA-1944 in clinical trials for the treatment of CHIKV infection is warranted.

Conflict of interest statement

J.E.C. has served as a consultant for Luna Biologics, is a member of the Scientific Advisory Boards of Meissa Vaccines and is founder of IDBiologics. The Crowe laboratory at Vanderbilt University Medical Center has received sponsored research agreements from IDBiologics and AstraZeneca. L.M.P. is a consultant for Abbott, Broadview Ventures, Bristol Myers Squibb, Moderna, Pfizer and WorldCare Clinical. A.A., H.Z.A., S.Himansu, S.K., S.L., R.P., S.Han, J.-M.L. and T.Z. are employees of, and M.H. is a former employee of, Moderna and may hold stock/stock options in the company. J.E.T. is a consultant of Moderna.

© 2021. The Author(s).

Figures

Fig. 1. Trial flow.
Fig. 1. Trial flow.
The study protocol planned to enroll 8 participants per treatment group. Six of 8 participants were allocated in the 0.6 mg kg−1 dose group due to the decision to augment the premedication regimen with steroid to assess its potential mitigation of infusion-related reactions (0.6 mg kg−1 + steroid group). aParticipants received loratadine and ranitidine 90 min before infusion. bParticipants received loratadine, ranitidine (sentinel, expansion) and acetaminophen (expansion) 90 min before infusion. cParticipants received steroid (dexamethasone) and diphenhydramine and famotidine 90 min before infusion. dParticipants received diphenhydramine and famotidine 90 min before infusion. eParticipants were administered two 0.3 mg kg−1 doses on days 1 and 8.
Fig. 2. Serum concentration of CHKV-24 IgG,…
Fig. 2. Serum concentration of CHKV-24 IgG, mRNA encoding heavy and light chains of CHKV-24 IgG and IAL.
a, Mean serum concentration time profiles of CHKV-24 IgG after the administration of single doses of 0.1, 0.3 and 0.6 mg kg−1 and 2 doses of 0.3 mg kg−1 mRNA-1944 over the course of 366 d and during 28 d (inset). b, Mean serum concentration time profiles for mRNA (heavy and light chains) and IAL during 28 d. The error bars represent the s.e.m. The dotted lines represent the serum target concentration of 1 µg ml−1 antibody anticipated to provide neutralizing antibody protection against CHIKV infection,,,. n = 6 participants at each time point for the single-dose 0.1, 0.3 and 0.6 mg kg−1 + steroid and 2-dose 0.3 mg kg−1 groups; n = 4 participants at each time point for the single-dose 0.6 mg kg−1 group examined over 366 d for CHKV-24 IgG and over 28 d for mRNA (heavy and light chains) and IAL.
Fig. 3. Neutralizing antibody titers for single-dose…
Fig. 3. Neutralizing antibody titers for single-dose groups.
Serum neutralizing titers of CHKV-24 IgG against CHIKV were assessed using the PRNT at 12, 24 and 48 h after administration of 0.1, 0.3 and 0.6 mg kg−1 (without steroid) doses of mRNA-1944. The percentage of participants achieving PRNT50 GMTs >100 and the GMTs for each dose group are provided. PRNT50 GMT >100 represents a level of CHIKV neutralizing antibodies previously associated with protection from both symptomatic CHIKV infection and subclinical seroconversion in humans,.

References

    1. Gao S, Song S, Zhang L. Recent progress in vaccine development against chikungunya virus. Front. Microbiol. 2019;10:2881.
    1. Goyal M, et al. Recent development in the strategies projected for chikungunya vaccine in humans. Drug Des. Devel. Ther. 2018;12:4195–4206.
    1. Schwameis M, Buchtele N, Wadowski PP, Schoergenhofer C, Jilma B. Chikungunya vaccines in development. Hum. Vaccin. Immunother. 2016;12:716–731.
    1. Zaid A, et al. Chikungunya arthritis: implications of acute and chronic inflammation mechanisms on disease management. Arthritis Rheumatol. 2018;70:484–495.
    1. Edington F, Varjão D, Melo P. Incidence of articular pain and arthritis after chikungunya fever in the Americas: a systematic review of the literature and meta-analysis. Joint Bone Spine. 2018;85:669–678.
    1. Powers AM. Vaccine and therapeutic options to control chikungunya virus. Clin. Microbiol. Rev. 2017;31:e00104-16.
    1. Pan American Health Organization, Regional Office for the Americas of the World Health Organization. Chikungunya: Data, Maps and Statistics (2017).
    1. World Health Organization. Chikungunya (2020).
    1. Abeyratne E, et al. Liposomal delivery of the RNA genome of a live-attenuated chikungunya virus vaccine candidate provides local, but not systemic protection after one dose. Front. Immunol. 2020;11:304.
    1. Chen GL, et al. Effect of a chikungunya virus-like particle vaccine on safety and tolerability outcomes: a randomized clinical trial. JAMA. 2020;323:1369–1377.
    1. Garg H, Mehmetoglu-Gurbuz T, Joshi A. Virus like particles (VLP) as multivalent vaccine candidate against Chikungunya, Japanese encephalitis, yellow fever and Zika virus. Sci. Rep. 2020;10:4017.
    1. Reisinger EC, et al. Immunogenicity, safety, and tolerability of the measles-vectored chikungunya virus vaccine MV-CHIK: a double-blind, randomised, placebo-controlled and active-controlled phase 2 trial. Lancet. 2019;392:2718–2727.
    1. Couderc T, et al. Prophylaxis and therapy for Chikungunya virus infection. J. Infect. Dis. 2009;200:516–523.
    1. Kam Y-W, et al. Early neutralizing IgG response to Chikungunya virus in infected patients targets a dominant linear epitope on the E2 glycoprotein. EMBO Mol. Med. 2012;4:330–343.
    1. Kam Y-W, et al. Early appearance of neutralizing immunoglobulin G3 antibodies is associated with chikungunya virus clearance and long-term clinical protection. J. Infect. Dis. 2012;205:1147–1154.
    1. Milligan GN, Schnierle BS, McAuley AJ, Beasley DWC. Defining a correlate of protection for chikungunya virus vaccines. Vaccine. 2019;37:7427–7436.
    1. Galatas B, et al. Long-lasting immune protection and other epidemiological findings after Chikungunya emergence in a Cambodian rural community, April 2012. PLoS Negl. Trop. Dis. 2016;10:e0004281.
    1. Nitatpattana N, et al. Long-term persistence of Chikungunya virus neutralizing antibodies in human populations of North Eastern Thailand. Virol. J. 2014;11:183.
    1. Pierro A, et al. Persistence of anti-chikungunya virus-specific antibodies in a cohort of patients followed from the acute phase of infection after the 2007 outbreak in Italy. New Microbes New Infect. 2015;7:23–25.
    1. Smith SA, et al. Isolation and characterization of broad and ultrapotent human monoclonal antibodies with therapeutic activity against Chikungunya virus. Cell Host Microbe. 2015;18:86–95.
    1. Clayton AM. Monoclonal antibodies as prophylactic and therapeutic agents against chikungunya virus. J. Infect. Dis. 2016;214:S506–S509.
    1. Masrinoul P, et al. Monoclonal antibody targeting chikungunya virus envelope 1 protein inhibits virus release. Virology. 2014;464–465:111–117.
    1. Pal P, et al. Development of a highly protective combination monoclonal antibody therapy against Chikungunya virus. PLoS Pathog. 2013;9:e1003312.
    1. Broeckel R, et al. Therapeutic administration of a recombinant human monoclonal antibody reduces the severity of chikungunya virus disease in rhesus macaques. PLoS Negl. Trop. Dis. 2017;11:e0005637.
    1. Yoon I-K, et al. High rate of subclinical chikungunya virus infection and association of neutralizing antibody with protection in a prospective cohort in the Philippines. PLoS Negl. Trop. Dis. 2015;9:e0003764.
    1. Yoon I-K, et al. Pre-existing chikungunya virus neutralizing antibodies correlate with risk of symptomatic infection and subclinical seroconversion in a Philippine cohort. Int. J. Infect. Dis. 2020;95:167–173.
    1. Claydon J, et al. Respiratory syncytial virus-neutralizing serum antibody titers in infants following palivizumab prophylaxis with an abbreviated dosing regimen. PLoS ONE. 2017;12:e0176152.
    1. Rubison L, Corey A, Hanfling D. Estimation of time period for effective human inhalational anthrax treatment including antitoxin therapy. PLoS Curr. 2017;9:ecurrents.outbreaks.7896c43f69838f17ce1c2c372e79d55d.
    1. Emu B, et al. Phase 3 study of ibalizumab for multidrug-resistant HIV-1. N. Engl. J. Med. 2018;379:645–654.
    1. Mulangu S, et al. A randomized, controlled trial of Ebola virus disease therapeutics. N. Engl. J. Med. 2019;381:2293–2303.
    1. Eli Lilly and Company. Lilly Announces Proof of Concept Data for Neutralizing Antibody LY-CoV555 in the COVID-19 Outpatient Setting (2020).
    1. Regeneron. Regeneron’s Regn-Cov2 Antibody Cocktail Reduced Viral Levels and Improved Symptoms in Non-Hospitalized Covid-19 Patients (2020).
    1. Van Hoecke L, Roose K. How mRNA therapeutics are entering the monoclonal antibody field. J. Transl. Med. 2019;17:54.
    1. Marston HD, Paules CI, Fauci AS. Monoclonal antibodies for emerging infectious diseases––borrowing from history. N. Engl. J. Med. 2018;378:1469–1472.
    1. Sparrow E, Friede M, Sheikh M, Torvaldsen S. Therapeutic antibodies for infectious diseases. Bull. World Health Organ. 2017;95:235–237.
    1. Stadler CR, et al. Elimination of large tumors in mice by mRNA-encoded bispecific antibodies. Nat. Med. 2017;23:815–817.
    1. Pardi N, et al. Administration of nucleoside-modified mRNA encoding broadly neutralizing antibody protects humanized mice from HIV-1 challenge. Nat. Commun. 2017;8:14630.
    1. Thran M, et al. mRNA mediates passive vaccination against infectious agents, toxins, and tumors. EMBO Mol. Med. 2017;9:1434–1447.
    1. Feldman RA, et al. mRNA vaccines against H10N8 and H7N9 influenza viruses of pandemic potential are immunogenic and well tolerated in healthy adults in phase 1 randomized clinical trials. Vaccine. 2019;37:3326–3334.
    1. Alberer M, et al. Safety and immunogenicity of a mRNA rabies vaccine in healthy adults: an open-label, non-randomised, prospective, first-in-human phase 1 clinical trial. Lancet. 2017;390:1511–1520.
    1. Richner JM, et al. Modified mRNA vaccines protect against Zika virus infection. Cell. 2017;169:176.
    1. Baden LR, et al. Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N. Engl. J. Med. 2021;384:403–416.
    1. Polack FP, et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N. Engl. J. Med. 2020;383:2603–2615.
    1. Moderna. Moderna Announces Additional Positive Phase 1 Data from Cytomegalovirus (CMV) Vaccine (mRNA-1647) and First Participant Dosed in Phase 2 Study (2020).
    1. Aliprantis AO, et al. A phase 1, randomized, placebo-controlled study to evaluate the safety and immunogenicity of an mRNA-based RSV prefusion F protein vaccine in healthy younger and older adults. Hum. Vaccin. Immunother. 2021;17:1248–1261.
    1. Anderson EJ, et al. Safety and immunogenicity of SARS-CoV-2 mRNA-1273 vaccine in older adults. NEJM. 2020;383:2427–2438.
    1. Chu L, et al. A preliminary report of a randomized controlled phase 2 trial of the safety and immunogenicity of mRNA-1273 SARS-CoV-2 vaccine. Vaccine. 2021;39:2791–2799.
    1. Ugur SA, et al. BNT162b2 vaccine induces neutralizing antibodies and poly-specific T cells in humans. Nature. 2021;595:572–577.
    1. Gaudinski MR, et al. Safety and pharmacokinetics of the Fc-modified HIV-1 human monoclonal antibody VRC01LS: a phase 1 open-label clinical trial in healthy adults. PLoS Med. 2018;15:e1002493.
    1. Cohen YZ, et al. Safety, pharmacokinetics, and immunogenicity of the combination of the broadly neutralizing anti-HIV-1 antibodies 3BNC117 and 10-1074 in healthy adults: a randomized, phase 1 study. PLoS ONE. 2019;14:e0219142.
    1. Sievers SA, Scharf L, West AP, Jr., Bjorkman PJ. Antibody engineering for increased potency, breadth and half-life. Curr. Opin. HIV AIDS. 2015;10:151–159.
    1. Nelson J, et al. Impact of mRNA chemistry and manufacturing process on innate immune activation. Sci. Adv. 2020;6:eaaz6893.
    1. Bournazos S, Gupta A, Ravetch JV. The role of IgG Fc receptors in antibody-dependent enhancement. Nat. Rev. Immunol. 2020;20:633–643.
    1. Kose N, et al. A lipid-encapsulated mRNA encoding a potently neutralizing human monoclonal antibody protects against chikungunya infection. Sci. Immunol. 2019;4:eaaw6647.
    1. U.S. Department of Health and Human Services. Common Terminology Criteria for Adverse Events (CTCAE) Version 5.0 (2017).
    1. Hassett KJ, et al. Optimization of lipid nanoparticles for intramuscular administration of mRNA vaccines. Mol. Ther. Nucleic Acids. 2019;15:1–11.
    1. Kowalski PS, Rudra A, Miao L, Anderson DG. Delivering the messenger: advances in technologies for therapeutic mRNA delivery. Mol. Ther. 2019;27:710–728.
    1. Ko S-Y, et al. Enhanced neonatal Fc receptor function improves protection against primate SHIV infection. Nature. 2014;514:642–645.
    1. Sampson HA, et al. Second symposium on the definition and management of anaphylaxis: summary report––Second National Institute of Allergy and Infectious Disease/Food Allergy and Anaphylaxis Network symposium. J. Allergy Clin. Immunol. 2006;117:391–397.
    1. Center for Biologics Evaluation and Research. Toxicity Grading Scale for Healthy Adult and Adolescent Volunteers Enrolled in Preventive Vaccine Clinical Trials: Guidance for Industry (2007).

Source: PubMed

3
Se inscrever