Suptavumab for the Prevention of Medically Attended Respiratory Syncytial Virus Infection in Preterm Infants

Eric A F Simões, Eduardo Forleo-Neto, Gregory P Geba, Mohamed Kamal, Feng Yang, Helen Cicirello, Matthew R Houghton, Ronald Rideman, Qiong Zhao, Sarah L Benvin, Alicia Hawes, Erin D Fuller, Elzbieta Wloga, Jose M Novoa Pizarro, Flor M Munoz, Scott A Rush, Jason S McLellan, Leah Lipsich, Neil Stahl, George D Yancopoulos, David M Weinreich, Christos A Kyratsous, Sumathi Sivapalasingam, Eric A F Simões, Eduardo Forleo-Neto, Gregory P Geba, Mohamed Kamal, Feng Yang, Helen Cicirello, Matthew R Houghton, Ronald Rideman, Qiong Zhao, Sarah L Benvin, Alicia Hawes, Erin D Fuller, Elzbieta Wloga, Jose M Novoa Pizarro, Flor M Munoz, Scott A Rush, Jason S McLellan, Leah Lipsich, Neil Stahl, George D Yancopoulos, David M Weinreich, Christos A Kyratsous, Sumathi Sivapalasingam

Abstract

Background: Respiratory syncytial virus (RSV) is a major cause of childhood medically attended respiratory infection (MARI).

Methods: We conducted a randomized, double-blind, placebo-controlled phase 3 trial in 1154 preterm infants of 1 or 2 doses of suptavumab, a human monoclonal antibody that can bind and block a conserved epitope on RSV A and B subtypes, for the prevention of RSV MARI. The primary endpoint was proportion of subjects with RSV-confirmed hospitalizations or outpatient lower respiratory tract infection (LRTI).

Results: There were no significant differences between primary endpoint rates (8.1%, placebo; 7.7%, 1-dose; 9.3%, 2-dose). Suptavumab prevented RSV A infections (relative risks, .38; 95% confidence interval [CI], .14-1.05 in the 1-dose group and .39 [95% CI, .14-1.07] in the 2-dose group; nominal significance of combined suptavumab group vs placebo; P = .0499), while increasing the rate of RSV B infections (relative risk 1.36 [95% CI, .73-2.56] in the 1-dose group and 1.69 [95% CI, .92-3.08] in the 2-dose group; nominal significance of combined suptavumab group vs placebo; P = .12). Sequenced RSV isolates demonstrated no suptavumab epitope changes in RSV A isolates, while all RSV B isolates had 2-amino acid substitution in the suptavumab epitope that led to loss of neutralization activity. Treatment emergent adverse events were balanced across treatment groups.

Conclusions: Suptavumab did not reduce overall RSV hospitalizations or outpatient LRTI because of a newly circulating mutant strain of RSV B. Genetic variation in circulating RSV strains will continue to challenge prevention efforts.

Clinical trials registration: NCT02325791.

Keywords: efficacy; infants; respiratory syncytial virus; safety.

© The Author(s) 2020. Published by Oxford University Press for the Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
Randomization, trial assignment, and follow-up. aOne subject eligible for the trial was not randomized but was dosed. Note: The full analysis set (all infants randomized and dosed with study drug) is used for analysis. Abbreviation: AE, adverse events.
Figure 2.
Figure 2.
Cumulative incidence of (A) overall RSV primary endpoint (RSV hospitalization or outpatient LRTI) over time, by treatment group, and (B) by RSV-A and RSV-B subtype primary endpoint, by treatment group. Subjects who had no event during the 150-day efficacy assessment period were censored at the last time point when their primary endpoint was assessed, ie, day 150 visit (day 150 ± 5 days) for completers of that visit, or the last visit (scheduled or unscheduled) completed by a subject up to day 150 for noncompleters of the day 150 visit. Abbreviations: LRTI, lower respiratory tract infection; RSV, respiratory syncytial virus.
Figure 3.
Figure 3.
Neutralization assay indicating that suptavumab is able to neutralize RSV-A clinical isolates from trial participants but not RSV-B isolates. A neutralization assay using the indicated RSV strain was performed using suptavumab or palivizumab. To determine neutralization ability, each antibody was incubated with clinical trial sample subtype A (MOI: .02) or subtype B (MOI: .05) for 2 hours (37°C, 5% CO2). Virus-free and antibody-free controls were included. Postincubation, the antibody–virus mixture was added to the HEp-2 cells and the infection was maintained for 3 days. The degree of infection was determined by enzyme-linked immunosorbent assay. Luminescence values were analyzed by a 3-parameter logistic equation over an 11-point response curve (GraphPad Prism). Abbreviations: MOI, multiplicity of infection; RSV, respiratory syncytial virus.

References

    1. Shi T, McAllister DA, O’Brien KL, et al. . 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. Mazur NI, Higgins D, Nunes MC, et al. ; Respiratory Syncytial Virus Network (ReSViNET) Foundation . The respiratory syncytial virus vaccine landscape: lessons from the graveyard and promising candidates. Lancet Infect Dis 2018; 18:e295–311.
    1. IMpact-RSV. Palivizumab, a humanized respiratory syncytial virus monoclonal antibody, reduces hospitalization from respiratory syncytial virus infection in high-risk infants. Pediatrics 1998; 102:531–7.
    1. American Academy of Pediatrics Committee on Infectious Diseases; American Academy of Pediatrics Bronchiolitis Guidelines Committee. Updated guidance for palivizumab prophylaxis among infants and young children at increased risk of hospitalization for respiratory syncytial virus infection. Pediatrics 2014; 134:e620–38.
    1. Adis Insight. Suptavumab. Available at: . Accessed 5 March 2019.
    1. Chintha P, Wloga E, Kamat V, Duan X, Perez-Caballero D. REGN222, a potent neutralizing antibody. In: 9th International Respiratory Syncytial Virus Symposium 2014. 9–13 November, 2014. Capetown, South Africa.
    1. Hause AM, Henke DM, Avadhanula V, Shaw CA, Tapia LI, Piedra PA. Sequence variability of the respiratory syncytial virus (RSV) fusion gene among contemporary and historical genotypes of RSV/A and RSV/B. PLoS One 2017; 12:e0175792.
    1. Sivapalasingam S, Caballero-Perez D, Houghton M, et al. . Phase 1 study evaluating safety, tolerability, pharmacokinetics and immunogenicity of REGN2222 in healthy adults: a new human monoclonal RSV-F antibody for RSV prevention [poster]. In: ID Week 2015. 7–11 October, 2015. San Diego, CA.
    1. Robbie GJ, Zhao L, Mondick J, Losonsky G, Roskos LK. Population pharmacokinetics of palivizumab, a humanized anti-respiratory syncytial virus monoclonal antibody, in adults and children. Antimicrob Agents Chemother 2012; 56:4927–36.
    1. Chen X, Xu B, Guo J, et al. . Genetic variations in the fusion protein of respiratory syncytial virus isolated from children hospitalized with community-acquired pneumonia in China. Sci Rep 2018; 8:4491.
    1. Lu B, Liu H, Tabor DE, et al. . Emergence of new antigenic epitopes in the glycoproteins of human respiratory syncytial virus collected from a US surveillance study, 2015-17. Sci Rep 2019; 9:3898.
    1. Simões EAF, Bont L, Manzoni P, et al. . Past, present and future approaches to the prevention and treatment of respiratory syncytial virus infection in children. Infect Dis Ther 2018; 7:87–120.
    1. Zhao X, Sullender WM. In vivo selection of respiratory syncytial viruses resistant to palivizumab. J Virol 2005; 79:3962–8.
    1. Zhu Q, McAuliffe JM, Patel NK, et al. . Analysis of respiratory syncytial virus preclinical and clinical variants resistant to neutralization by monoclonal antibodies palivizumab and/or motavizumab. J Infect Dis 2011; 203:674–82.
    1. Oliveira DB, Iwane MK, Prill MM, et al. . Molecular characterization of respiratory syncytial viruses infecting children reported to have received palivizumab immunoprophylaxis. J Clin Virol 2015; 65:26–31.
    1. Choi SH, Park KS, Kim YJ. Analysis of respiratory syncytial virus fusion protein from clinical isolates of Korean children in palivizumab era, 2009–2015. J Infect Chemother 2019; 25:514–9.
    1. Tapia LI, Shaw CA, Aideyan LO, et al. . Gene sequence variability of the three surface proteins of human respiratory syncytial virus (HRSV) in Texas. PLoS One 2014; 9:e90786.
    1. Zhu Q, McLellan JS, Kallewaard NL, et al. . A highly potent extended half-life antibody as a potential RSV vaccine surrogate for all infants. Sci Transl Med 2017; 9:eaaj1928.
    1. Sivapalasingam S, Kamal M, Slim R, et al. . Safety, pharmacokinetics, and immunogenicity of a co-formulated cocktail of three human monoclonal antibodies targeting Ebola virus glycoprotein in healthy adults: a randomised, first-in-human phase 1 study. Lancet Infect Dis 2018; 18:884–93.
    1. Pascal KE, Dudgeon D, Trefry JC, et al. . Development of clinical-stage human monoclonal antibodies that treat advanced Ebola virus disease in nonhuman primates. J Infect Dis 2018; 218:612–26.
    1. Piedra PA, Jewell AM, Cron SG, Atmar RL, Glezen WP. Correlates of immunity to respiratory syncytial virus (RSV) associated hospitalization: establishment of minimum protective threshold levels of serum neutralizing antibodies. Vaccine 2003; 21:3479–82.
    1. Kulkarni PS, Hurwitz JL, Simões EAF, Piedra PA. Establishing correlates of protection for vaccine development: considerations for the respiratory syncytial virus vaccine field. Viral Immunol 2018; 31:195–203.

Source: PubMed

3
Abonneren