Safety and Immunogenicity of the Respiratory Syncytial Virus Vaccine RSV/ΔNS2/Δ1313/I1314L in RSV-Seronegative Children

Ruth A Karron, Cindy Luongo, Jocelyn San Mateo, Kimberli Wanionek, Peter L Collins, Ursula J Buchholz, Ruth A Karron, Cindy Luongo, Jocelyn San Mateo, Kimberli Wanionek, Peter L Collins, Ursula J Buchholz

Abstract

Background: Respiratory syncytial virus (RSV) is the leading global cause of severe pediatric acute respiratory tract illness, and a vaccine is needed. RSV/ΔNS2/Δ1313/I1314L contains 2 attenuating elements: (1) deletion of the interferon antagonist NS2 gene and (2) deletion of codon 1313 of the RSV polymerase gene and the stabilizing missense mutation I1314L. This live vaccine candidate was temperature-sensitive, genetically stable, replication restricted, and immunogenic in nonhuman primates.

Methods: A single intranasal dose of RSV/ΔNS2/Δ1313/I1314L was evaluated in a double-blind, placebo-controlled trial (vaccine-placebo ratio, 2:1) at 106 plaque-forming units (PFU) in 15 RSV-seropositive children and at 105 and 106 PFU in 21 and 30 RSV-seronegative children, respectively.

Results: In RSV-seronegative children, the 105 PFU dose was overattenuated, but the 106 PFU dose was well tolerated, infectious (RSV/ΔNS2/Δ1313/I1314L replication detected in 90% of vaccinees), and immunogenic (geometric mean serum RSV plaque-reduction neutralizing antibody titer, 1:64). After the RSV season, 9 of 20 vaccinees had increases in the RSV titer that were significantly greater than those in 8 of 10 placebo recipients (1:955 vs 1:69, respectively), indicating that the vaccine primed for anamnestic responses after natural RSV exposure.

Conclusion: Rational design yielded a genetically stable candidate RSV vaccine that is attenuated yet immunogenic in RSV-seronegative children, warranting further evaluation.

Clinical trials registration: NCT01893554.

Keywords: RSV; live-attenuated RSV vaccine; pediatric; vaccine.

© The Author(s) 2019. Published by Oxford University Press for the Infectious Diseases Society of America. All rights reserved. For permissions, e-mail: journals.permissions@oup.com.

Figures

Figure 1.
Figure 1.
Screening, enrollment, and follow-up of RSV-seropositive children (panel A) and RSV-seronegative children (panels B and C) in the phase I clinical trial of the RSV/ΔNS2/Δ1313/I1314L vaccine. As described in Methods, enrollment in each of the cohorts depicted in A, B, and C occurred sequentially following a satisfactory safety review of data from the previous cohort.
Figure 2.
Figure 2.
Proportions of RSV-seronegative vaccinees and placebo recipients with indicated illnesses in the 105 PFU cohort (panel A) or the 106 PFU cohort (panel B). Vaccinees are shown in black; placebo recipients are shown in gray.
Figure 3.
Figure 3.
Peak titers of vaccine virus shed as detected by culture (A) and quantitative reverse-transcription polymerase chain reaction (RT-qPCR) (B) among RSV-seronegative recipients of 105 or 106 plaque-forming units (PFU) of vaccine. C, D, Individual daily titers by culture (C) and RT-qPCR (D) for RSV-seronegative recipients of the 106 PFU dose. Triangles represent peak titers from individual vaccine recipients; continuous lines, mean values; and dotted lines, titers assigned for culture-negative (0.5 log10 PFU/mL) (C) and RT-qPCR–negative (D) (1.7 log10 copies/mL) nasal wash (NW) samples.
Figure 4.
Figure 4.
Respiratory syncytial virus (RSV) plaque reduction neutralization antibody titers (PRNT) in RSV-seronegative recipients of 106 plaque-forming units of vaccine (A) or placebo (B), measured before immunization (Pre), 56 days after immunization (Post), before the first RSV surveillance season (Pre–1st SS) and after the first RSV surveillance season (Post–1st SS). Triangles represent children with RSV-associated medically attended acute respiratory illness (RSV-MAARI); solid lines, means for the postinoculation, presurveillance, and postsurveillance time points; symbols enclosed by dotted line, titers from vaccinees with evidence of wild-type RSV infection during surveillance (≥4-fold increases in PRNT, RSV-MAARI, or both), with means from these subsets provided next to enclosed symbols.
Figure 5.
Figure 5.
Respiratory syncytial virus (RSV) plaque reduction neutralization antibody titers (PRNT) in 20 RSV-seronegative recipients of 106 plaque-forming units of vaccine (A) and 10 placebo recipients (B), by individual child. All 30 children were followed during the first surveillance season (SS); 16 of 20 vaccinees and 6 of 10 placebo recipients were followed through 2 RSV surveillance seasons. Dotted lines represent data from the 4 vaccinees and 4 placebo recipients who participated only in the first season; dashed lines, participants who experienced an episode of RSV-associated medically attended acute respiratory illness or lower respiratory illness during a surveillance season; triangles, occurrence of such illness. A, Vaccinees with a ≥4-fold increase in the first season (left), the second season (middle), or neither season (right). B, Placebo recipients with a ≥4-fold increase in 1 or both seasons. Only 1 placebo recipient did not have a detectable increase in RSV PRNT, and this child participated in only the first surveillance season. Abbreviations: Post, after; Pre, before.

References

    1. Hall CB, Weinberg GA, Iwane MK, et al. . The burden of respiratory syncytial virus infection in young children. N Engl J Med 2009; 360:588–98.
    1. Shi T, McAllister DA, O’Brien KL, et al. ; RSV Global Epidemiology Network. Global, regional, and national disease burden estimates of acute lower respiratory infections due to respiratory syncytial virus in young children in 2015: a systematic review and modelling study. Lancet 2017; 390:946–58.
    1. Wahl B, O’Brien KL, Greenbaum A, et al. . Burden of Streptococcus pneumoniae and Haemophilus influenzae type b disease in children in the era of conjugate vaccines: global, regional, and national estimates for 2000-15. Lancet Glob Health 2018; 6:e744–57.
    1. Karron RA, Black RE. Determining the burden of respiratory syncytial virus disease: the known and the unknown. Lancet 2017; 390:917–8.
    1. Wright PF, Karron RA, Belshe RB, et al. . Evaluation of a live, cold-passaged, temperature-sensitive, respiratory syncytial virus vaccine candidate in infancy. J Infect Dis 2000; 182:1331–42.
    1. Karron RA, Wright PF, Belshe RB, et al. . Identification of a recombinant live attenuated respiratory syncytial virus vaccine candidate that is highly attenuated in infants. J Infect Dis 2005; 191:1093–104.
    1. Wright PF, Karron RA, Madhi SA, et al. . The interferon antagonist NS2 protein of respiratory syncytial virus is an important virulence determinant for humans. J Infect Dis 2006; 193:573–81.
    1. Wright PF, Karron RA, Belshe RB, et al. . The absence of enhanced disease with wild type respiratory syncytial virus infection occurring after receipt of live, attenuated, respiratory syncytial virus vaccines. Vaccine 2007; 25:7372–8.
    1. Malkin E, Yogev R, Abughali N, et al. . Safety and immunogenicity of a live attenuated RSV vaccine in healthy RSV-seronegative children 5 to 24 months of age. PLoS One 2013; 8:e77104.
    1. Karron RA, Luongo C, Thumar B, et al. . A gene deletion that up-regulates viral gene expression yields an attenuated RSV vaccine with improved antibody responses in children. Sci Transl Med 2015; 7:312ra175.
    1. Buchholz UJ, Cunningham CK, Muresan P, et al. ; International Maternal Pediatric Adolescent AIDS Clinical Trials (IMPAACT) P1114 Study Team. Live respiratory syncytial virus (RSV) vaccine candidate containing stabilized temperature-sensitivity mutations is highly attenuated in RSV-seronegative infants and children. J Infect Dis 2018; 217:1338–46.
    1. McFarland EJ, Karron RA, Muresan P, et al. ; International Maternal Pediatric Adolescent AIDS Clinical Trials (IMPAACT) 2000 Study Team. Live-attenuated respiratory syncytial virus vaccine candidate with deletion of RNA synthesis regulatory protein M2-2 is highly immunogenic in children. J Infect Dis 2018; 217:1347–55.
    1. Kim HW, Canchola JG, Brandt CD, et al. . Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine. Am J Epidemiol 1969; 89:422–34.
    1. Murphy BR, Sotnikov AV, Lawrence LA, Banks SM, Prince GA. Enhanced pulmonary histopathology is observed in cotton rats immunized with formalin-inactivated respiratory syncytial virus (RSV) or purified F glycoprotein and challenged with RSV 3-6 months after immunization. Vaccine 1990; 8:497–502.
    1. Connors M, Collins PL, Firestone CY, et al. . Cotton rats previously immunized with a chimeric RSV FG glycoprotein develop enhanced pulmonary pathology when infected with RSV, a phenomenon not encountered following immunization with vaccinia–RSV recombinants or RSV. Vaccine 1992; 10:475–84.
    1. Schneider-Ohrum K, Cayatte C, Bennett AS, et al. . Immunization with low doses of recombinant postfusion or prefusion respiratory syncytial virus F primes for vaccine-enhanced disease in the cotton rat model independently of the presence of a Th1-biasing (GLA-SE) or Th2-biasing (alum) adjuvant. J Virol 2017; 91:e02180–16.
    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. Collins PL, Hill MG, Camargo E, Grosfeld H, Chanock RM, Murphy BR. Production of infectious human respiratory syncytial virus from cloned cDNA confirms an essential role for the transcription elongation factor from the 5’ proximal open reading frame of the M2 mRNA in gene expression and provides a capability for vaccine development. Proc Natl Acad Sci U S A 1995; 92:11563–7.
    1. Ramaswamy M, Shi L, Varga SM, Barik S, Behlke MA, Look DC. Respiratory syncytial virus nonstructural protein 2 specifically inhibits type I interferon signal transduction. Virology 2006; 344:328–39.
    1. Lo MS, Brazas RM, Holtzman MJ. Respiratory syncytial virus nonstructural proteins NS1 and NS2 mediate inhibition of Stat2 expression and alpha/beta interferon responsiveness. J Virol 2005; 79:9315–9.
    1. Spann KM, Tran KC, Chi B, Rabin RL, Collins PL. Suppression of the induction of alpha, beta, and lambda interferons by the NS1 and NS2 proteins of human respiratory syncytial virus in human epithelial cells and macrophages [published correction appears in J Virol 2005; 78:6705]. J Virol 2004; 78:4363–9.
    1. Whitehead SS, Bukreyev A, Teng MN, et al. . Recombinant respiratory syncytial virus bearing a deletion of either the NS2 or SH gene is attenuated in chimpanzees. J Virol 1999; 73:3438–42.
    1. Valarcher JF, Furze J, Wyld S, Cook R, Conzelmann KK, Taylor G. Role of alpha/beta interferons in the attenuation and immunogenicity of recombinant bovine respiratory syncytial viruses lacking NS proteins. J Virol 2003; 77:8426–39.
    1. Liesman RM, Buchholz UJ, Luongo CL, et al. . RSV-encoded NS2 promotes epithelial cell shedding and distal airway obstruction. J Clin Invest 2014; 124:2219–33.
    1. Luongo C, Winter CC, Collins PL, Buchholz UJ. Respiratory syncytial virus modified by deletions of the NS2 gene and amino acid S1313 of the L polymerase protein is a temperature-sensitive, live-attenuated vaccine candidate that is phenotypically stable at physiological temperature. J Virol 2013; 87:1985–96.
    1. Bukreyev A, Belyakov IM, Berzofsky JA, Murphy BR, Collins PL. Granulocyte-macrophage colony-stimulating factor expressed by recombinant respiratory syncytial virus attenuates viral replication and increases the level of pulmonary antigen-presenting cells. J Virol 2001; 75:12128–40.
    1. Karron RA, Wright PF, Crowe JE Jr, et al. . Evaluation of two live, cold-passaged, temperature-sensitive respiratory syncytial virus vaccines in chimpanzees and in human adults, infants, and children. J Infect Dis 1997; 176:1428–36.
    1. Beeler JA, van Wyke Coelingh K. Neutralization epitopes of the F glycoprotein of respiratory syncytial virus: effect of mutation upon fusion function. J Virol 1989; 63:2941–50.
    1. Coates HV, Alling DW, Chanock RM. An antigenic analysis of respiratory syncytial virus isolates by a plaque reduction neutralization test. Am J Epidemiol 1966; 83:299–313.
    1. Englund JA, Karron RA, Cunningham CK, et al. ; International Maternal Pediatric Adolescent AIDS Clinical Trials (IMPAACT) P1096 Study Group. Safety and infectivity of two doses of live-attenuated recombinant cold-passaged human parainfluenza type 3 virus vaccine rHPIV3cp45 in HPIV3-seronegative young children. Vaccine 2013; 31:5706–12.
    1. Liang B, Ngwuta JO, Herbert R, et al. . Packaging and prefusion stabilization separately and additively increase the quantity and quality of respiratory syncytial virus (RSV)-neutralizing antibodies induced by an RSV fusion protein expressed by a parainfluenza virus vector. J Virol 2016; 90:10022–38.

Source: PubMed

3
Iratkozz fel