A single dose of ChAdOx1 Chik vaccine induces neutralizing antibodies against four chikungunya virus lineages in a phase 1 clinical trial

Pedro M Folegatti, Kate Harrison, Lorena Preciado-Llanes, Fernando Ramos Lopez, Mustapha Bittaye, Young Chan Kim, Amy Flaxman, Duncan Bellamy, Rebecca Makinson, Jonathan Sheridan, Sasha R Azar, Rafael Kroon Campos, Mark Tilley, Nguyen Tran, Daniel Jenkin, Ian Poulton, Alison Lawrie, Rachel Roberts, Eleanor Berrie, Shannan L Rossi, Adrian Hill, Katie J Ewer, Arturo Reyes-Sandoval, Pedro M Folegatti, Kate Harrison, Lorena Preciado-Llanes, Fernando Ramos Lopez, Mustapha Bittaye, Young Chan Kim, Amy Flaxman, Duncan Bellamy, Rebecca Makinson, Jonathan Sheridan, Sasha R Azar, Rafael Kroon Campos, Mark Tilley, Nguyen Tran, Daniel Jenkin, Ian Poulton, Alison Lawrie, Rachel Roberts, Eleanor Berrie, Shannan L Rossi, Adrian Hill, Katie J Ewer, Arturo Reyes-Sandoval

Abstract

Chikungunya virus (CHIKV) is a reemerging mosquito-borne virus that causes swift outbreaks. Major concerns are the persistent and disabling polyarthralgia in infected individuals. Here we present the results from a first-in-human trial of the candidate simian adenovirus vectored vaccine ChAdOx1 Chik, expressing the CHIKV full-length structural polyprotein (Capsid, E3, E2, 6k and E1). 24 adult healthy volunteers aged 18-50 years, were recruited in a dose escalation, open-label, nonrandomized and uncontrolled phase 1 trial (registry NCT03590392). Participants received a single intramuscular injection of ChAdOx1 Chik at one of the three preestablished dosages and were followed-up for 6 months. The primary objective was to assess safety and tolerability of ChAdOx1 Chik. The secondary objective was to assess the humoral and cellular immunogenicity. ChAdOx1 Chik was safe at all doses tested with no serious adverse reactions reported. The vast majority of solicited adverse events were mild or moderate, and self-limiting in nature. A single dose induced IgG and T-cell responses against the CHIKV structural antigens. Broadly neutralizing antibodies against the four CHIKV lineages were found in all participants and as early as 2 weeks after vaccination. In summary, ChAdOx1 Chik showed excellent safety, tolerability and 100% PRNT50 seroconversion after a single dose.

Conflict of interest statement

Pedro M. Folegatti is a consultant to Vaccitech, which is developing adenoviral vectored vaccines. Adrian Hill is a cofounder of and consultant to Vaccitech Ltd and is named as an inventor on a patent covering the design and use of Ch.

© 2021. The Author(s).

Figures

Fig. 1. Trial profile.
Fig. 1. Trial profile.
Study profile showing the allocation of participants to three dosage groups: Group 1, low dose at 5 × 109 vp, Group 2 intermediate dose at 2.5 × 1010 vp, and Group 3 high dose at 5 × 1010 vp. None of the 24 recruited participants were lost in follow-up.
Fig. 2. PRNT 50 values over time.
Fig. 2. PRNT50 values over time.
PRNT50 reciprocal titers are shown for each participant. Arrows indicate when ChAdOx1 Chik was administered. Lower limit of detection (LLOD) is 5. a Overall responses by timepoint in all trial volunteers (n = 24). Geometric means and 95% CI; Kruskal–Wallis test with Dunn’s correction. b Same data as (a) but analyzed by dosage groups: low dose was 5 × 109 vp (n = 6), intermediate (mid) dose was 2.5 × 1010 vp (n = 9), and high dose was 5 × 1010 vp (n = 9). Geometric means and 95% CI; Kruskal–Wallis test with Dunn’s correction. One day 56, the sample from the low-dose group was excluded from analysis due to QC failure.
Fig. 3. ELISA titers over time.
Fig. 3. ELISA titers over time.
CHIKV IgG response by standardized ELISA to E2 protein in 120 serum samples of trial participants. a Individual IgG titers over time (n = 24). The dashed line represents the cut-off value for seropositivity. Geometric means and 95% CI; Friedman test with Dunn’s correction. b Same data as (a) but represented as fold change from baseline (day 0) and analyzed by dosage group: low dose was 5 × 109 vp (n = 6), intermediate (mid) dose was 2.5 × 1010 vp (n = 9), and high dose was 5 × 1010 vp (n = 9). Geometric means and 95% CI; Friedman test with Dunn’s correction. c Correlation of PRNT50 and IgG ELISA by dosage group at 5 time points. Low dose, six participants, n = 29 (on day 56, samples were excluded due to QC failure); intermediate (mid) dose, nine participants (n = 45); high dose, nine participants (n = 45). Spearman correlation, two-tailed. d Same data as (c) but correlation is only shown for the nine participants vaccinated at the intermediate (mid) dose (n = 9 per timepoint). Spearman correlation, two-tailed.
Fig. 4. T-cell responses over time.
Fig. 4. T-cell responses over time.
a Ex vivo enzyme-linked immunospot (ELISpot) for IFN-γ to CHIKV structural antigens measured as total responses to CHIKV peptides (sum of 13 pools spanning C, E3, E2, 6 K, and E1). SFC per million PBMC during a 6-month follow-up period (n = 24). Median and IQR; Kruskal–Wallis test with Dunn’s correction. b Proportion of spots contributed by C, E3, E2, 6 K and E1 over time. c Intracellular cytokine staining (ICS) by flow cytometry to assess CD4+ and CD8+ T-cell functionality. Percentage of cytokine-producing CD4+ and CD8+ T cells (n = 24). Median and IQR; Mann–Whitney test, two-tailed.

References

    1. Robinson MC. An epidemic of virus disease in Southern Province, Tanganyika territory, in 1952–1953. Trans. R. Soc. Trop. Med. Hyg. 1955;49:28–32. doi: 10.1016/0035-9203(55)90080-8.
    1. Kariuki Njenga M, et al. Tracking epidemic chikungunya virus into the Indian Ocean from East Africa. J. Gen. Virol. 2008;89:2754–2760. doi: 10.1099/vir.0.2008/005413-0.
    1. Yactayo S, Staples JE, Millot V, Cibrelus L, Ramon-Pardo P. Epidemiology of chikungunya in the Americas. J. Infect. Dis. 2016;214:S441–S445. doi: 10.1093/infdis/jiw390.
    1. Suhrbier A. Rheumatic manifestations of chikungunya: emerging concepts and interventions. Nat. Rev. Rheumatol. 2019;15:597–611. doi: 10.1038/s41584-019-0276-9.
    1. Kraemer MU, et al. The global distribution of the arbovirus vectors Aedes aegypti and Ae. albopictus. eLife. 2015;4:e08347. doi: 10.7554/eLife.08347.
    1. Amraoui F, Failloux A-B. Chikungunya: an unexpected emergence in Europe. Curr. Opin. Virol. 2016;21:146–150. doi: 10.1016/j.coviro.2016.09.014.
    1. Sergon K, et al. Seroprevalence of chikungunya virus infection on Grande Comore Island, union of the Comoros, 2005. Am. J. Trop. Med. Hyg. 2007;76:1189–1193. doi: 10.4269/ajtmh.2007.76.1189.
    1. Josseran L, et al. Chikungunya disease outbreak, Reunion Island. Emerg. Infect. Dis. 2006;12:1994–1995. doi: 10.3201/eid1212.060710.
    1. Rezza G, et al. Infection with chikungunya virus in Italy: an outbreak in a temperate region. Lancet. 2007;370:1840–1846. doi: 10.1016/S0140-6736(07)61779-6.
    1. Calba C, et al. Preliminary report of an autochthonous chikungunya outbreak in France, July to September 2017. Eurosurveillance. 2017;22:17-00647. doi: 10.2807/1560-7917.ES.2017.22.39.17-00647.
    1. Delisle E, et al. Chikungunya outbreak in Montpellier, France, September to October 2014. Eurosurveillance. 2015;20:21108. doi: 10.2807/1560-7917.ES2015.20.17.21108.
    1. Grandadam M, et al. Chikungunya virus, Southeastern France. Emerg. Infect. Dis. 2011;17:910–913. doi: 10.3201/eid1705.101873.
    1. da Cunha RV, Trinta KS. Chikungunya virus: clinical aspects and treatment - a review. Mem. Inst. Oswaldo Cruz. 2017;112:523–531. doi: 10.1590/0074-02760170044.
    1. Cardona-Ospina JA, Diaz-Quijano FA, Rodríguez-Morales AJ. Burden of chikungunya in Latin American countries: estimates of disability-adjusted life-years (DALY) lost in the 2014 epidemic. Int. J. Infect. Dis. 2015;38:60–61. doi: 10.1016/j.ijid.2015.07.015.
    1. López-Camacho C, et al. Assessment of immunogenicity and neutralisation efficacy of viral-vectored vaccines against chikungunya virus. Viruses. 2019;11:322. doi: 10.3390/v11040322.
    1. Kroon Campos R, et al. A single and un-adjuvanted dose of a chimpanzee adenovirus-vectored vaccine against chikungunya virus fully protects mice from lethal disease. Pathogens. 2019;8:231. doi: 10.3390/pathogens8040231.
    1. Kam Y, 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. doi: 10.1002/emmm.201200213.
    1. Lum F-M, et al. An essential role of antibodies in the control of chikungunya virus infection. J. Immunol. 2013;190:6295–6302. doi: 10.4049/jimmunol.1300304.
    1. Quiroz JA, et al. Human monoclonal antibodies against chikungunya virus target multiple distinct epitopes in the E1 and E2 glycoproteins. PLoS Pathog. 2019;15:e1008061. doi: 10.1371/journal.ppat.1008061.
    1. Kam Y-W, et al. Unique epitopes recognized by antibodies induced in chikungunya virus-infected non-human primates: implications for the study of immunopathology and vaccine development. PLoS ONE. 2014;9:e95647. doi: 10.1371/journal.pone.0095647.
    1. Fox JM, et al. Broadly neutralizing alphavirus antibodies bind an epitope on E2 and inhibit entry and egress. Cell. 2015;163:1095–1107. doi: 10.1016/j.cell.2015.10.050.
    1. Rodríguez-Morales AJ, Cardona-Ospina JA, Fernanda Urbano-Garzón S, Sebastian Hurtado-Zapata J. Prevalence of post-chikungunya infection chronic inflammatory arthritis: a systematic review and meta-analysis. Arthritis Care Res. 2016;68:1849–1858. doi: 10.1002/acr.22900.
    1. Handler MZ, Handler NS, Stephany MP, Handler GA, Schwartz RA. Chikungunya fever: an emerging viral infection threatening North America and Europe. Int. J. Dermatol. 2017;56:e19–e25. doi: 10.1111/ijd.13439.
    1. Chu H, et al. Deciphering the protective role of adaptive immunity to CHIKV/IRES a novel candidate vaccine against chikungunya in the A129 mouse model. Vaccine. 2013;31:3353–3360. doi: 10.1016/j.vaccine.2013.05.059.
    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. doi: 10.1371/journal.pntd.0003764.
    1. Plante K, et al. Novel chikungunya vaccine candidate with an IRES-based attenuation and host range alteration mechanism. PLoS Pathog. 2011;7:e1002142. doi: 10.1371/journal.ppat.1002142.
    1. Chen GL, et al. Effect of a chikungunya virus–like particle vaccine on safety and tolerability outcomes. JAMA. 2020;323:1369. doi: 10.1001/jama.2020.2477.
    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. 2018;392:2718–2727. doi: 10.1016/S0140-6736(18)32488-7.
    1. Wressnigg N, et al. Single-shot live-attenuated chikungunya vaccine in healthy adults: a phase 1, randomised controlled trial. Lancet Infect. Dis. 2020;20:1193–1203. doi: 10.1016/S1473-3099(20)30238-3.
    1. Goo L, et al. A virus-like particle vaccine elicits broad neutralizing antibody responses in humans to all chikungunya virus genotypes. J. Infect. Dis. 2016;214:1487–1491. doi: 10.1093/infdis/jiw431.
    1. Hoarau J-J, et al. Identical strength of the T cell responses against E2, nsP1 and capsid CHIKV proteins in recovered and chronic patients after the epidemics of 2005–2006 in La Reunion Island. PLoS ONE. 2013;8:e84695. doi: 10.1371/journal.pone.0084695.
    1. Messaoudi I, et al. Chikungunya virus infection results in higher and persistent viral replication in aged rhesus macaques due to defects in anti-viral immunity. PLoS Negl. Trop. Dis. 2013;7:e2343. doi: 10.1371/journal.pntd.0002343.
    1. Wauquier N, et al. The acute phase of chikungunya virus infection in humans is associated with strong innate immunity and T CD8 cell activation. J. Infect. Dis. 2011;204:115–123. doi: 10.1093/infdis/jiq006.
    1. Roques P, et al. Attenuated and vectored vaccines protect nonhuman primates against Chikungunya virus. JCI Insight. 2017;2:e83527. doi: 10.1172/jci.insight.83527.
    1. Dias CN, et al. Human CD8 T-cell activation in acute and chronic chikungunya infection. Immunology. 2018;155:499–504. doi: 10.1111/imm.12992.
    1. Hoarau J-J, et al. Persistent chronic inflammation and infection by chikungunya arthritogenic alphavirus in spite of a robust host immune response. J. Immunol. 2010;184:5914–5927. doi: 10.4049/jimmunol.0900255.
    1. Teo T-H, et al. A pathogenic role for CD4 + T cells during chikungunya virus infection in mice. J. Immunol. 2013;190:259–269. doi: 10.4049/jimmunol.1202177.
    1. Miner JJ, et al. Therapy with CTLA4-Ig and an antiviral monoclonal antibody controls chikungunya virus arthritis. Sci. Transl. Med. 2017;9:eaah3438. doi: 10.1126/scitranslmed.aah3438.
    1. Venugopalan A, Ghorpade RP, Chopra A. Cytokines in acute chikungunya. PLoS ONE. 2014;9:e111305. doi: 10.1371/journal.pone.0111305.
    1. Carissimo G, et al. Viperin controls chikungunya virus–specific pathogenic T cell IFNγ Th1 stimulation in mice. Life Sci. Alliance. 2019;2:e201900298. doi: 10.26508/lsa.201900298.
    1. Burrack KS, Montgomery SA, Homann D, Morrison TE. CD8 + T Cells Control Ross River Virus Infection in Musculoskeletal Tissues of Infected Mice. J. Immunol. 2015;194:678–689. doi: 10.4049/jimmunol.1401833.
    1. Dicks MDJ, et al. A novel chimpanzee adenovirus vector with low human seroprevalence: improved systems for vector derivation and comparative immunogenicity. PLoS ONE. 2012;7:e40385. doi: 10.1371/journal.pone.0040385.
    1. Folegatti PM, et al. Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: a preliminary report of a phase 1/2, single-blind, randomised controlled trial. Lancet. 2020;396:467–478. doi: 10.1016/S0140-6736(20)31604-4.
    1. Folegatti PM, et al. Safety and immunogenicity of a candidate Middle East respiratory syndrome coronavirus viral-vectored vaccine: a dose-escalation, open-label, non-randomised, uncontrolled, phase 1 trial. Lancet Infect. Dis. 2020;20:816–826. doi: 10.1016/S1473-3099(20)30160-2.
    1. Kim YC, et al. Development of an E2 ELISA methodology to assess chikungunya seroprevalence in patients from an endemic region of Mexico. Viruses. 2019;11:407. doi: 10.3390/v11050407.
    1. Rossi SL, et al. Immunogenicity and efficacy of a measles virus-vectored chikungunya vaccine in nonhuman primates. J. Infect. Dis. 2019;220:735–742. doi: 10.1093/infdis/jiz202.
    1. Langsjoen RM, et al. Chikungunya virus strains show lineage-specific variations in virulence and cross-protective ability in murine and nonhuman primate models. MBio. 2018;9:e02449–17. doi: 10.1128/mBio.02449-17.
    1. Ramsauer K, et al. Immunogenicity, safety, and tolerability of a recombinant measles-virus-based chikungunya vaccine: a randomised, double-blind, placebo-controlled, active-comparator, first-in-man trial. Lancet Infect. Dis. 2015;15:519–527. doi: 10.1016/S1473-3099(15)70043-5.
    1. Ewer K, et al. A monovalent chimpanzee adenovirus ebola vaccine boosted with MVA. N. Engl. J. Med. 2016;374:1635–1646. doi: 10.1056/NEJMoa1411627.

Source: PubMed

3
Sottoscrivi