Safety and immunogenicity evaluation of recombinant BCG vaccine against respiratory syncytial virus in a randomized, double-blind, placebo-controlled phase I clinical trial

Katia Abarca, Emma Rey-Jurado, Natalia Muñoz-Durango, Yaneisi Vázquez, Jorge A Soto, Nicolás M S Gálvez, Javier Valdés-Ferrada, Carolina Iturriaga, Marcela Urzúa, Arturo Borzutzky, Jaime Cerda, Luis Villarroel, Victoria Madrid, Pablo A González, José V González-Aramundiz, Susan M Bueno, Alexis M Kalergis, Katia Abarca, Emma Rey-Jurado, Natalia Muñoz-Durango, Yaneisi Vázquez, Jorge A Soto, Nicolás M S Gálvez, Javier Valdés-Ferrada, Carolina Iturriaga, Marcela Urzúa, Arturo Borzutzky, Jaime Cerda, Luis Villarroel, Victoria Madrid, Pablo A González, José V González-Aramundiz, Susan M Bueno, Alexis M Kalergis

Abstract

Background: Respiratory syncytial virus (RSV) is responsible for most respiratory tract infections and hospitalizations in infants and represents a significant economic burden for public health. The development of a safe, effective, and affordable vaccine is a priority for the WHO.

Methods: We conducted a double-blinded, escalating-dose phase 1 clinical trial in healthy males aged 18-50 years to evaluate safety, tolerability, and immunogenicity of a recombinant Mycobacterium bovis BCG vaccine expressing the nucleoprotein of RSV (rBCG-N-hRSV). Once inclusion criteria were met, volunteers were enrolled in three cohorts in an open and successive design. Each cohort included six volunteers vaccinated with 5 × 103, 5 × 104, or 1 × 105 CFU, as well as two volunteers vaccinated with the full dose of the standard BCG vaccine. This clinical trial (clinicaltrials.gov NCT03213405) was conducted in Santiago, Chile.

Findings: The rBCG-N-RSV vaccine was safe, well-tolerated, and no serious adverse events related to the vaccine were recorded. Serum IgG-antibodies directed against Mycobacterium and the N-protein of RSV increased after vaccination, which were capable of neutralizing RSV in vitro. Additionally, all volunteers displayed increased cellular response consisting of IFN-γ and IL-2 production against PPD and the N-protein, starting at day 14 and 30 post-vaccination respectively.

Interpretation: The rBCG-N-hRSV vaccine had a good safety profile and induced specific cellular and humoral responses.

Funding: This work was supported by Millennium Institute on Immunology and Immunotherapy from Chile (P09/016), FONDECYT 1190830, and FONDEF D11E1098.

Keywords: BCG vaccine; Human respiratory syncytial virus; Immunogenicity; Phase I clinical trial; Safety; Transmissibility.

Conflict of interest statement

Authors KA, ERJ, NMD, YV, JS, NG, JV, CI, MU, AB, JC, VM, PG, JG, SB, and AMK report grants from Millennium Institute on Immunology and Immunotherapy from Chile, grants from Fondo Nacional de Desarrollo Científico y Tecnológico, grants from Fondo de Fomento al Desarrollo Tecnológico, personal fees from Pontificia Universidad Católica de Chile, during the conduct of the study. LV reports personal fees from Pontificia Universidad Católica de Chile, during the conduct of the study .

© 2020 The Authors.

Figures

Fig. 1
Fig. 1
Clinical study flow diagram and timeline. This phase 1 clinical study was double-blinded and dose-escalated. (A) Each cohort included 6 volunteers vaccinated with escalating doses of rBCG-N-hRSV (Cohort A: 5 × 103 CFU (blue); Cohort B: 5 × 104 CFU (red); Cohort C: 1 × 105 CFU (green)) and 2 volunteers vaccinated with the standard BCG at full dose (BCG-WT 2 × 105 CFU (purple)). A DSMB evaluated the safety data of each cohort after the first 30 days of follow-up and decided if escalation could continue. (B) A timeline indicating the periods of immunization and end of visits is shown. RSV peaks reported during 2017 and 2018 are also indicated.
Fig. 2
Fig. 2
PPD- and N-RSV-specific humoral immune response in rBCG-N-hRSV-immunized volunteers. Anti-PPD and anti-N-RSV IgG levels were measured in the sera of all the volunteers at 0, 14, 30, 60, 120, and 180 dpv with rBCG-N-hRSV doses 5 × 103 (blue), 5 × 104 (red), and 1 × 105 (green) CFU and the full dose of BCG-WT (Purple). IgG levels are expressed as the geometric mean (and geometric standard deviation) of the concentrations of IgG (ng/mL) on a logarithmic scale. The responses against (A) PPD and (B) N-RSV antigens are shown. Statistical differences were evaluated by a two-way ANOVA with a posterior Tukey test. If different letters are shown above a specific time point (with their respective color-bar representing the corresponding group), then statistical differences (p<0.05) were found among those groups. If no letters are indicated above a group, then no statistical differences were found among them. n.s. = no statistical differences were found among any of the groups.
Fig. 3
Fig. 3
Evaluation of the neutralizing capacity of antibodies obtained from vaccinated subjects. The neutralizing capacities of the sera obtained from the immunized subjects were tested and are shown as fold change (Normalized to Day 0 for each subject). The sera samples were tested at 0, 14, 30, 60, 120, and 180 dpv with rBCG-N-hRSV doses 5 × 103 (A - Blue), 5 × 104 (B - Red), and 1 × 105 (C - Green) CFU and the full dose of BCG-WT (D - Purple). Neutralization was tested with amounts of IgG ranging from 10 µg to 0.1 µg, which are equivalent to dilutions 1/160 and 1/20.480, respectively. Highest amounts resulted in 100% neutralization, while lowest amounts resulted in 0% neutralization. Therefore, the level of neutralization for 0.75 µg of total antibodies -dilution 1/2.560- is shown. Statistical differences were evaluated by a one-way ANOVA with a posterior Tukey test. n.s.= No statistical differences; *=P<0.05.
Fig. 4
Fig. 4
PPD- and N-RSV-specific cellular immune response in the rBCG-N-hRSV-immunized subjects evaluated by ELISPOT. Cellular responses to PPD (A and B) and N-RSV (C and D) antigens in all PBMCs samples were measured at 0, 14, 30, 60, 120, and 180 dpv with rBCG-N-hRSV doses 5 × 103 (blue), 5 × 104 (red), and 1 × 105 (green) CFU and the full dose of BCG-WT (Purple). The IFN-γ- (A and C) and IL-2- (B and D) secreting cells were detected by ELISPOT and plotted as spot-forming cells (SFC) per million cells. Statistical differences were evaluated by a two-way ANOVA with a posterior Tukey test. If different letters are shown above a specific time point (with their respective color-bar representing the corresponding group), then statistical differences (p<0.05) were found among those groups. If no letters are indicated above a group, then no statistical differences were found among them.

References

    1. Nair H, Nokes DJ, Gessner BD. Global burden of acute lower respiratory infections due to respiratory syncytial virus in young children: a systematic review and meta-analysis. Lancet. 2010;375:1545–1555.
    1. Collins PL, Melero JA. Progress in understanding and controlling respiratory syncytial virus: Still crazy after all these years. Virus Res. 2011;162:80–99.
    1. Breese Hall C. The burgeoning burden of respiratory syncytial virus among children. Infect Disord – Drug Targets. 2012;12:92–97.
    1. Bohmwald K, Gálvez NMS, Ríos M, Kalergis AM. Neurologic alterations due to respiratory virus infections. Front Cell Neurosci. 2018;12:386.
    1. Andrade CA, Pacheco GA, Gálvez NMS, Soto JA, Bueno SM, Kalergis AM. Innate immune components that regulate the pathogenesis and resolution of hRSV and hMPV Infections. Viruses. 2020;12:637.
    1. Huang K, Incognito L, Cheng X, Ulbrandt ND, Wu H. Respiratory syncytial virus-neutralizing monoclonal antibodies motavizumab and palivizumab inhibit fusion. J Virol. 2010;84:8132–8140.
    1. Vekemans J, Moorthy V, Giersing B. Respiratory syncytial virus vaccine research and development: World Health Organization technological roadmap and preferred product characteristics. Vaccine. 2019;37:7394–7395.
    1. Kruijsen D, Schijf MA, Lukens M V. et al. Local innate and adaptive immune responses regulate inflammatory cell influx into the lungs after vaccination with formalin inactivated RSV. Vaccine. 2011;29:2730–2741.
    1. Knudson CJ, Hartwig SM, Meyerholz DK, Varga SM. RSV Vaccine-enhanced disease is orchestrated by the combined actions of distinct CD4 T cell subsets. PLoS Pathog. 2015;11:1–23.
    1. Kim HW, Canchola JG, Brandt CD. Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine. Am J Epidemiol. 1969;89:422–434.
    1. Bendelja K, Gagro A, Baće A. Predominant type-2 response in infants with respiratory syncytial virus (RSV) infection demonstrated by cytokine flow cytometry. Clin Exp Immunol. 2000;121:332–338.
    1. Acosta PL, Caballero MT, Polack FP. Brief history and characterization of enhanced respiratory syncytial virus disease. Clin Vaccine Immunol. 2016;23:189–195.
    1. PATH . Program for Appropriate Technology in Health; 2020. RSV vaccine and mAb Snapshot; p. 1.
    1. Bueno SM, González PA, Cautivo KM. Protective T cell immunity against respiratory syncytial virus is efficiently induced by recombinant BCG. Proc Natl Acad Sci. 2008;105:20822–20827.
    1. Zwerling A, Behr MA, Verma A, Brewer TF, Menzies D, Pai M. The BCG World atlas: a database of global BCG vaccination policies and practices. PLoS Med. 2011;8
    1. Abubakar I, Pimpin L, Ariti C. Systematic review and meta-analysis of the current evidence on the duration of protection by bacillus Calmette-Guérin vaccination against tuberculosis. Health Technol Assess. 2013;17:1–4.
    1. Aggarwal A, Dutta AK. Timing and dose of BCG vaccination in infants as assessed by postvaccination tuberculin sensitivity. Indian Pediatr. 1995;32:635–639.
    1. Marchant A, Goetghebuer T, Ota MO. Newborns develop a Th1-type immune response to Mycobacterium bovis bacillus Calmette-Guerin vaccination. J Immunol. 1999;163:2249–2255.
    1. Cautivo KM, Bueno SM, Cortes CM, Wozniak A, Riedel CA, Kalergis AM. Efficient lung recruitment of respiratory syncytial virus-specific Th1 cells induced by recombinant bacillus calmette-guerin promotes virus clearance and protects from infection. J Immunol. 2010;185:7633–7645.
    1. Céspedes PF, Rey-Jurado E, Espinoza JA. A single, low dose of a cGMP recombinant BCG vaccine elicits protective T cell immunity against the human respiratory syncytial virus infection and prevents lung pathology in mice. Vaccine. 2017;35:757–766.
    1. Soto JA, Gálvez NMS, Rivera CA. Recombinant BCG vaccines reduce pneumovirus-caused airway pathology by inducing protective humoral immunity. Front Immunol. 2018;9:2875.
    1. Valdespino Gómez JL, García García MDL. Declaración de Helsinki. Gac Med Mex. 2001;137:391.
    1. U. S. Department of Health and Human Services Toxicity Grading Scale for Healthy Adult and Adolescent Volunteers Enrolled in Preventive Vaccine Clinical Trials. Guid Ind. 2007;1:1–10.
    1. van Remmerden Y, Xu F, van Eldik M, Heldens JGM, Huisman W, Widjojoatmodjo MN. An improved respiratory syncytial virus neutralization assay based on the detection of green fluorescent protein expression and automated plaque counting. Virol J. 2012;9:253.
    1. Raj A, Singh N, Gupta KB. Comparative evaluation of several gene targets for designing a multiplex-PCR for an early diagnosis of extrapulmonary tuberculosis. Yonsei Med J. 2016;57:88.
    1. van Pinxteren LAH, Ravn P, Agger EM, Pollock J, Andersen P. Diagnosis of tuberculosis based on the two specific antigens ESAT-6 and CFP10. Clin Vaccine Immunol. 2000;7:155–160.
    1. Nissen TN, Birk NM, Kjærgaard J. Adverse reactions to the Bacillus Calmette-Guérin (BCG) vaccine in new-born infants-an evaluation of the Danish strain 1331 SSI in a randomized clinical trial. Vaccine. 2016;34:2477–2482.
    1. Bont L, Versteegh J, Swelsen WTN. Natural reinfection with respiratory syncytial virus does not boost virus-specific T-cell immunity. Pediatr Res. 2002;52:363–367.
    1. Hall CB, Walsh EE, Long CE, Schnabel KC. Immunity to and frequency of reinfection with respiratory syncytial virus. J Infect Dis. 1991;163:693–698.
    1. Whelan KT, Pathan AA, Sander CR. Safety and immunogenicity of boosting BCG vaccinated subjects with BCG: comparison with boosting with a new TB vaccine, MVA85A. PLoS One. 2009;4:e5934.

Source: PubMed

3
Abonnieren