Immunogenicity and safety of an adjuvanted inactivated polio vaccine, IPV-Al, following vaccination in children at 2, 4, 6 and at 15-18 months

Xavier Sáez-Llorens, Birgit Thierry-Carstensen, Lina Saem Stoey, Charlotte Sørensen, Henrik Wachmann, Ananda S Bandyopadhyay, Pernille Ingemann Nielsen, Mie Vestergaard Kusk, Xavier Sáez-Llorens, Birgit Thierry-Carstensen, Lina Saem Stoey, Charlotte Sørensen, Henrik Wachmann, Ananda S Bandyopadhyay, Pernille Ingemann Nielsen, Mie Vestergaard Kusk

Abstract

Background: Availability of affordable inactivated polio vaccines (IPV) is of major importance to meet the increasing global supply needs. The results presented here demonstrate non-inferiority of a reduced-dose, aluminium hydroxide-adjuvanted IPV (IPV-Al) to standard IPV.

Methods: A phase 3, observer-blinded, randomised, clinical trial was conducted in Panama in infants who received either IPV-Al (n = 400) or standard IPV (n = 400) at age 2, 4 and 6 months. In the booster trial, subjects received a single dose of IPV-Al at age 15-18 months. The primary endpoint was type-specific seroconversion, defined as an antibody titre ≥4-fold higher than the estimated maternal antibody titre and a titre ≥8, one month after the primary vaccination series. In the booster trial, the primary endpoint was the type-specific booster effects (geometric mean titre (GMT) post-booster (Day 28)/GMT pre-booster (Day 0).

Results: Seroconversion rates following primary vaccination with IPV-Al vs IPV were: 96.1% vs 100% (type 1); 100% vs 100% (type 2); and 99.2% vs 100% (type 3) respectively. IPV-Al was non-inferior to IPV, as the lower 95% confidence limits of the treatment differences were above the pre-defined -10%-point limit: 3.94% (-6.51; -2.01) for type 1; 0.0% (-1.30; -1.37) for type 2; -0.85 (-2.46; 0.40) for type 3. The booster effects for the group primed with IPV-Al versus the group primed with IPV were 25.3 vs 9.2 (type 1), 19.1 vs 6.5 (type 2) and 50.4 vs 12.5 (type 3). IPV-Al had a comparable safety profile to that of IPV.

Conclusions: Non-inferiority of IPV-Al to standard IPV with respect to seroconversion after vaccination at 2, 4 and 6 months was confirmed for all three poliovirus serotypes. A robust booster response was demonstrated following vaccination with IPV-Al, regardless of the primary vaccine received. Both vaccines were well tolerated. ClinicalTrials.gov identifiers: NCT03025750 and NCT03671616.

Funding: Bill & Melinda Gates Foundation.

Keywords: Affordable IPV; Aluminium hydroxide adjuvant; Booster vaccination; Immunogenicity; Polio; Primary vaccination; Standalone adjuvanted IPV.

Conflict of interest statement

Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. Disclosures XSL and HW have no conflicts of interest to disclose. BTC and LSS are employees of Statens Serum Institut, a governmental non-profit research organisation. CS, PIN and MVK are employees of AJ Vaccines A/S. Both Statens Serum Institut and AJ Vaccines A/S were involved in developing the vaccine at the time the trial was conducted. ASB is a full-time employee at the Bill & Melinda Gates Foundation.

Copyright © 2020 The Authors. Published by Elsevier Ltd.. All rights reserved.

Figures

Fig. 1
Fig. 1
Disposition of subjects in the primary (A) and booster (B) trial. Fig. 1A. The full analysis set (FAS) defined as all randomised and vaccinated infants with at least one post-baseline measurement. The per-protocol (PP) analysis set was defined as all infants in the FAS who had no major deviations from the protocol. The safety analysis set (SAF) was defined as all randomised infants who received at least one treatment dose. N = number of infants in the group. *One subject completed but did not have a sample post-primary vaccination series and was excluded from FAS (hence FAS = 371). Fig. 1B. The full analysis set (FAS-booster) was defined as all enrolled subjects with the primary endpoint, i.e. the booster effect, for at least one of the three poliovirus types. The per-protocol analysis set (PP) was defined as all FAS subjects who did not have major deviations from the protocol. The safety analysis set (SAF) was defined as all enrolled subjects who received at least one trial vaccination. *Two subjects primed with IPV-Al did not have post-booster samples due to discontinuation and difficult venous access. ** Two subjects primed with IPV did not have post-booster samples due to discontinuation and difficult venous access.
Fig. 1
Fig. 1
Disposition of subjects in the primary (A) and booster (B) trial. Fig. 1A. The full analysis set (FAS) defined as all randomised and vaccinated infants with at least one post-baseline measurement. The per-protocol (PP) analysis set was defined as all infants in the FAS who had no major deviations from the protocol. The safety analysis set (SAF) was defined as all randomised infants who received at least one treatment dose. N = number of infants in the group. *One subject completed but did not have a sample post-primary vaccination series and was excluded from FAS (hence FAS = 371). Fig. 1B. The full analysis set (FAS-booster) was defined as all enrolled subjects with the primary endpoint, i.e. the booster effect, for at least one of the three poliovirus types. The per-protocol analysis set (PP) was defined as all FAS subjects who did not have major deviations from the protocol. The safety analysis set (SAF) was defined as all enrolled subjects who received at least one trial vaccination. *Two subjects primed with IPV-Al did not have post-booster samples due to discontinuation and difficult venous access. ** Two subjects primed with IPV did not have post-booster samples due to discontinuation and difficult venous access.
Fig. 2
Fig. 2
Geometric mean titres (GMTs) in infants primed with IPV-Al and IPV at baseline (2 months), post-primary vaccination (7 months), pre-booster (mean 16.5 months, range 15–18 months) and post-booster (mean 17.5 months, range 16–19 months) for poliovirus type 1 (A), type 2 (B) and type 3 (C). GMTs are shown as log2 values.
Fig. 3
Fig. 3
Reverse cumulative titre distribution at baseline (2 months), post-primary vaccination (7 months), pre-booster (15–18 months) and post-booster (16–19 months) for poliovirus type 1 (A), type 2 (B) and type 3 (C). Subjects received either IPV-Al or IPV as primary vaccination; all subjects received IPV-Al as booster vaccination.

References

    1. Global Polio Eradication Initiative. Wild poliovirus list. 2019. Accessed 31Jan2020. .
    1. Global Polio Eradication Initiative. Global eradication of wild poliovirus type 2 declared.. 2015. Accessed 31Jan2020. .
    1. Bandyopadhyay A.S., Garon J., Seib K., Orenstein W.A. Polio vaccination: past, present and future. Future Microbiol. 2015;10(5):791–808.
    1. World Health Organization. Weekly epidemiological record. Meeting of the SAGE group on immunization, April 2019- conclusions and recommendations.. 2019. Accessed 16Oct2019. .
    1. Rivera L., Pedersen R.S., Pena L., Olsen K.J., Andreasen L.V., Kromann I. Immunogenicity and safety of three aluminium hydroxide adjuvanted vaccines with reduced doses of inactivated polio vaccine (IPV-Al) compared with standard IPV in young infants in the Dominican Republic: a phase 2, non-inferiority, observer-blinded, randomised, and controlled dose investigation trial. Lancet Infect Dis. 2017;17:745–753.
    1. Bravo L., Carlos J.C., Gatchalian S.R., Montellano M.E.B., Tabora C.F.C.B., Thierry-Carstensen B. Immunogenicity and safety of an adjuvanted inactivated polio vaccine, IPV-Al, compared to standard IPV: A phase 3 observer-blinded, randomised, controlled trial in infants vaccinated at 6, 10, 14 weeks and 9 months of age. Vaccine. 2020;38(3):503–538.
    1. Lindgren L.M., Tingskov P.N., Justesen A.H., Nedergaard B.S., Olsen K.J., Andreasen L.V. First-in-human safety and immunogenicity investigations of three adjuvanted reduced dose inactivated poliovirus vaccines (IPV-Al SSI) compared to full dose IPV Vaccine SSI when given as a booster vaccination to adolescents with a history of IPV vaccination at 3, 5, 12 months and 5 years of age. Vaccine. 2017;35:596–604.
    1. World Medical Association Declaration of Helsinki ethical principles for medical research involving human subjects. JAMA. 2000;284:3043–3045.
    1. International Conference on Harmonisation. ICH Harmonised guideline. Integrated addendum to ICH E6(1): guideline for good clinical practice E6(R2). 2016. Accessed 16Oct2019. .
    1. Robertson S. Module 6: Poliomyelitis. In: The Immunological Basis for Immunization Series, WHO. 1993. Accessed 16Oct2019. .
    1. Brown G.C., Rabson A.S., Schieble J.H. The effect of gamma globulin on subclinical infection in familial associates of poliomyelitis cases. II Serological Studies and Virus Isolations from Pharyngeal Secretions. J Immunol. 1955;74:71–80.
    1. Plotkin S.A. Correlates of protection induced by vaccination. Clin Vaccine Immunol. 2010;17:1055–1065.
    1. Melnick JL. Enteroviruses. In: AS E, editor. Viral infections of humans: Epidemiology and control. Switzerland: Springer Nature; 1984. p. 187-251.
    1. Aristegui J., Dal-Re R., Diez-Delgado J., Mares J., Casanovas J.M., Garcia-Corbeira P. Comparison of the reactogenicity of a combined diphtheria, tetanus, acellular pertussis, hepatitis B, inactivated polio (DTaPa-HBV-IPV) vaccine, mixed with the Haemophilus influenzae type b (Hib) conjugate vaccine and administered as a single injection, with the DTPa-IPV/Hib and hepatitis B vaccines administered in two simultaneous injections to infants at 2,4 and 6 months of age. Vaccine. 2003;21:3593–3600.
    1. Tejedor T.C., Omeñaca F., García-Sicilia J., Verdaguer J., Van Esso D., Esporrín C. Immunogenicity and reactogenicity of a three-dose primary vaccination course with a combined diphtheria-tetanus-acellular pertussis-hepatitis B-inactivated polio-haemophilus influenzae type B vaccine. Pediatr Infect Dis J. 2004;23:1109–1115.
    1. Tejedor J.C., Moro M., Ruiz-Contreras J., Castro J., Gómez-Campderá J.A., Navarro M.L. Immunogenicity and reactogenicity of primary immunization with a hexavalent diphtheria-tetanus-acellular pertussis-hepatitis B-inactivated polio-haemophilus influenzae type B vaccine coadministered with two doses of a meningococcal C-tetanus toxoid conjugate vaccine. Pediatr Infect Dis J. 2006;25:713–720.
    1. Yeh S.H., Ward J.I., Partridge S., Marcy S.M., Lee H., Jing J. Safety and immunogenicity of a pentavalent diphtheria, tetanus, pertussis, hepatitis B and polio combination vaccine in infants. Pediatr Infect Dis J. 2001;20:973–980.
    1. Li R., Li C.G., Li Y., Liu Y., Zhao H., Chen X. Primary and booster vaccination with an inactivated poliovirus vaccine (IPV) is immunogenic and well-tolerated in infants and toddlers in China. Vaccine. 2016;34:1436–1443.
    1. Marlow R., Kuriyakose S., Mesaros N., Han H.H., Tomlinson R., Faust S.N. A phase III, open-label, randomised multicentre study to evaluate the immunogenicity and safety of a booster dose of two different reduced antigen diphtheria-tetanus-acellular pertussis-polio vaccines, when co-administered with measles-mumps-rubella vacci. Vaccine. 2018;36:2300–2306.
    1. Glaxo Smith Kline. Infanrix Hexa - Product Monopraph. 2018. Accessed 16Oct2019.
    1. Sanofi Pasteur. Hexaxim - Summary of Product Characteristics. EMA. 2018. Accessed 16Oct2019. .
    1. Siegrist C-A. Vaccine Immunology. In: Plotkin, Stanley. Orensterin, W. Offit P et al, editor. Plotkin’s Vaccines. 7th ed. Philadelphia: Elsevier; 2018. p. 16–34.
    1. Faden H., Duffy L., Sun M., Shuff C. Long-term immunity to poliovirus in children immunized with live attenuated and enhanced-potency inactivated trivalent poliovirus vaccines. J Infect Dis. 1993;168:452–454.
    1. Böttiger M. Polio immunity to killed vaccine: an 18-year follow-up. Vaccine. 1990;8:443–445.
    1. Pietrzyk J.J., Wysocki J., Pejcz J., Galaj A., Majda-Stanislawska E., Käyhty H. Safety and immunogenicity of a DTaP-IPVVero (serum-free) combination vaccine in comparison to DTaP-IPVMkc when administered simultaneously with Haemophilus influenzae type b conjugate vaccine (PRP-T) in children at 2, 3.5, 5 and 16 months of age. Vaccine. 2008;26:5296–5303.
    1. Gyhrs A., Lyngholm E., Larsen S.O., Aggerbeck H., Heron I. Immunogenicity and safety of a tetravalent diphtheria-tetanus-acellular pertussis-inactivated poliovirus vaccine. Scand J Infect Dis. 1999;31:579–585.
    1. Haastrup E., Thierry-Carstensen B., Jensen A.M., Stellfeld M., Heilmann C. Safety and immunogenicity of a booster dose of inactivated poliovirus vaccine produced in vero-cells. Vaccine. 2004;22:958–962.
    1. Thierry-Carstensen B., Dalby T., Stevner M.A., Robbins J.B., Schneerson R., Trollfors B. Experience with monocomponent acellular pertussis combination vaccines for infants, children, adolescents and adults–a review of safety, immunogenicity, efficacy and effectiveness trials and 15 years of field experience. Vaccine. 2013;31:5178–5191.
    1. Thierry-Carstensen B., Jordan K., Uhlving H.H., Dalby T., Sorensen C., Jensen A.M. A randomised, double-blind, non-inferiority clinical trial on the safety and immunogenicity of a tetanus, diphtheria and monocomponent acellular pertussis (TdaP) vaccine in comparison to a tetanus and diphtheria (Td) vaccine when given as booster vaccinations to healthy adults. Vaccine. 2012;30:5464–5471.
    1. Hawken J., Troy S.B. Adjuvants and inactivated polio vaccine: a systematic review. Vaccine. 2012;30:6971–6979.
    1. Vidor E. Poliovirus Vaccine-Inactivated. In: Plotkin S.A., Orenstein W.A., Offit P.A., Edwards K.M., editors. Plotkin’s Vaccines. 7th ed. Elsevier; Philadelphia: 2018. pp. 841–865.
    1. Voysey M., Kelly D.F., Fanshawe T.R., Sadarangani M., O'Brien K.L., Perera R. The influence of maternally derived antibody and infant age at vaccination on infant vaccine responses: an individual participant meta-analysis. JAMA Pediatrics. 2017;171:637–646.
    1. Gaensbauer J.T., Gast C., Bandyopadhyay A.S., O’Ryan M., Saez-Llorens X., Rivera L. Impact of maternal antibody on the immunogenicity of inactivated polio vaccine in infants immunized with bivalent oral polio vaccine: implications for the Polio Eradication Endgame. Clin Infect Dis. 2018;67(suppl_1):S57–S65.
    1. Jafari H., Deshpande J.M., Sutter R.W., Bahl S., Verma H., Ahmad M. Polio eradication. Efficacy of inactivated poliovirus in India. Science. 2014;345(6199):922–925.
    1. John J., Giri S., Karthikeyan A.S., Iturriza-Gomara M., Muliyil J., Abraham A. Effect of a single inactivated poliovirus vaccine dose on intestinal immunity against poliovirus in children previously given oral vaccine: an open-label, randomised controlled trial. Lancet. 2014;384(9953):1505–1512.

Source: PubMed

3
구독하다