Summary
Respiratory syncytial virus (RSV) is the second most common cause of infant mortality and an important cause of morbidity and mortality in older adults. Efforts to develop an RSV vaccine or immunoprophylaxis remain highly active. Thirty-three RSV prevention candidates are in clinical development using six different approaches: recombinant vector, subunit, particle-based, live-attenuated, chimeric, and nucleic acid in addition to monoclonal antibodies (mAbs). Eight candidates are in phase III clinical trials. Understanding the epitopes targeted by highly neutralizing antibodies has resulted in a shift from empirical to rational and structure-based vaccine and mAb design. An extended half-life mAb for all infants is likely within one year of regulatory approval for high income countries. Live-attenuated vaccines are in development for older infants. Subunit vaccines are in late-stage trials for pregnant women to protect infants, while vector, subunit and nucleic acid approaches are being developed for older adults. Urgent next steps include ensuring access and affordability of an RSV vaccine globally. This review gives an overview of RSV vaccines and mAbs in clinical development highlighting different target populations, antigens, and latest trial results.
Keywords: respiratory syncytial virus, vaccines
Introduction
In the past decade, the substantial burden of RSV disease has received increasing recognition globally. RSV is the second leading cause of infant mortality after the neonatal period1 with more than 99% of childhood deaths occurring in low and middle income countries (LMICs)2. Nevertheless, the RSV burden in children is likely underestimated, and major gaps in knowledge regarding RSV disease burden have only been recently addressed. More than 50% of pediatric RSV mortality occurs in the community in LMICs3 with poverty as an important risk factor [Figure 1]. Infants at highest risk of RSV disease in HICs include the very young infants born prematurely and those with underlying congenital heart or chronic lung disease4, Down’s Syndrome5 and neuromuscular disorders6. Maternal vaccination is insufficient to protect infants with extreme prematurity as transplacental antibody transfer only reaches mature levels towards the end of the third trimester7.
In older adults (>60 years of age), the burden of morbidity and mortality due to RSV was also under recognized until recently. Modelling studies now estimate that the RSV burden is similar to the burden of seasonal influenza in adults >65 years of age8–10. Preliminary economic evaluations have highlighted the potential value of a vaccine for older adults, especially in high income countries (HICs). Key economic drivers of cost-effectiveness include RSV incidence, risk of death, and level and duration of protection11,12.
Natural immunity to RSV is incomplete, and reinfection occurs throughout life13. A concern in the development of RSV vaccines is the potential for enhanced respiratory disease (ERD) in which more severe illness occurs upon natural infection after vaccination of RSV-naïve infants as was observed with formalin-inactivated RSV (FI-RSV) in the 1960s14. ERD was associated with induction of poorly neutralizing antibodies in vaccine recipients15 and animal models of ERD suggest a Th-2 biased T cell response16. For this reason, an RSV vaccine for RSV-naïve recipients ideally elicits potent neutralizing antibodies without a Th2 bias. While a definitive correlate of protection against RSV infection remains elusive, cell-mediated immunity17, mucosal IgA18, and neutralizing antibodies19–22 have been associated with protection from RSV infection.
Stabilization of the prefusion (pre-F) conformation of the RSV fusion (F) protein has led to the determination of viral epitopes that elicit highly neutralizing antibodies. Antibodies that recognize pre-F provide the majority of the neutralizing activity in human RSV-immune sera23 supporting development of vaccine candidates and mAbs based on stabilized pre-F antigens. There are three different target populations for RSV prevention: (1) pediatric, (2) maternal, and (3) older adult population [Figure 3]24. Leading strategies for the pediatric population include passive immunoprophylaxis with mAbs for young infants (<6 months) and live-attenuated vaccines for active immunization of older infants (> 6 months). Young infants may also be protected by passively transferred antibodies in immunized pregnant women. Stabilized pre-F subunit vaccines are in late-phase development for maternal vaccination. Finally, for older adults three vaccination approaches (nucleic acid, subunit and vector-based) that employ pre-F antigen are in late phase trials.
In 2018, we conducted a comprehensive review of the RSV vaccine landscape in which we distilled lessons learned from late-phase vaccine failures and identified 19 vaccine candidates and monoclonal antibodies in clinical trials25. The review may have provided vaccine developers with guidance for future vaccine development by endorsing pre-F as a new target for RSV preventive interventions. The pre-F antigen is now the basis of six vaccine candidates and two mAbs in phase III trials26–29. Furthermore, we endorsed controlled human infection models (CHIM) as a unique tool to generate rapid proof of concept of protection and extensive immunological characterization. This approach has been adopted into clinical development for six current RSV vaccine candidates (MV-012–968. RSVPreF, MVA-BN-RSV, palivizumab biosimilar, clesrovimab, Ad26.RSV.PreF). This updated review shows that the majority, 58% (11/19), of candidates from 2018 (and 30%, 3/10 candidates from our 2015 review) have continued development, with simultaneous expansion of the field with 19 additional candidates having entered clinical trials [Figure 2]. Finally, following the success of mRNA SAR-CoV-2 vaccine development, vaccines delivered as mRNA is a novel preventative approach that has rapidly accelerated to late-phase trials.
Methods
Vaccine and mAb candidates in clinical phases of development were identified using the PATH RSV Vaccine and mAb snapshot (last updated September 28, 202124). The data collection template from previous reviews25 was updated[Supplementary Table 1] and filled out by searching PubMed, clinical trial registries, WHO, European Medicines Agency (EMA) and pharmaceutical websites for each vaccine candidate, with no date or language restrictions (NM, JT, YL, TB, MW, FV). We did not intend to conduct a systematic review of the peer-reviewed literature but instead provide an update on the current development by capturing all recent publicly available information. No inclusion or exclusion criteria were used. Instead, for each vaccine candidate or mAb in clinical development information was selected by date (with preference for more recent literature) and by relevance (with preference for trial data). When available, peer-reviewed publications were preferred to information from trial registries or pharmaceutical websites. To supplement the data collected and the identified gaps in knowledge, data for this review were systematically collected using the data collection template [Supplemental Table 1] at the virtual RSV Vaccines for the World conference organized by the Respiratory Syncytial Virus Network (ReSViNET) from November 10–12, 2021. The goal of this meeting was to share scientific data and expertise on RSV vaccine development, and to connect stakeholders involved in RSV research. During the meeting, information was collected (NM, JT, YL, TB, MW, FV) from scientific presentations, posters, and discussions. Any publicly available data from this meeting has been included in this manuscript. Vaccines were divided into six major groups: recombinant vector, subunit, particle-based, live-attenuated, chimeric, and nucleic acid. Immunoprophylaxis with monoclonal antibodies included as a seventh category. Vaccine characteristics such as mechanism of action, adjuvants, route of administration and summary of trial results have been compiled in Table 1.
Table 1:
Vaccine | Company/sponsor | Manufacturin g process | Antigen | Adjuvant | Mechanism of action | Target populatio n | Route of administratio n | Clinical Phase | Animal models | Phase I | Phase II | Phase III | Result summary |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|
SUBUNIT | |||||||||||||
VN-0200 | Daiichi Sankyo | N/A | VAGA-9001a | MABH-9002b | N/A | O | IM | I | N/A | Jun 2021-Jan 2022; NCT04914520 (n=48) | N/A | N/A | N/A |
DS-Cav1 | NIH/NIAID | Structure-based vaccine design | Stabiliz ed PreF DS-Cav1 | Non-adjuvante d /alum | RSV pre-F | M&O | IM | I | Cotton rats, Calves, Mice, Macaques | Feb 2017-Oct 2019; NCT03049488 (n=95) | N/A | N/A | PhI: safe & well-tolerated; vaccination elicited robust neutralizing Ab response sustained at 44 weeks |
DPX-RSV | Immunovaccine | DepovaxTM, a lipid-in-oil delivery system | SHe | DepoVax (DPX-RSV[A]), DPX, or aluminu m hydroxide | Exposure to SHe antigen to generate a nonneutralizing Ab & CD4+ T-cell response | O | IM | I | Cotton rats, Mice | May 2015-Jun 2017; NCT02472548 (n=40) | N/A | N/A | PhI: Safe & well-tolerated, no SAEs, antigen-specific Ab response durable >6 months |
BARS13 | Advaccine | N/A | G | Non-adjuvante d/CSA | RSV G; immunosuppressant | P/O | IM | II | N/A | Oct 2018 – Aug 2019; NCT04851977/ACTRN12618000948291 (n=60) | May 2021-Jun 2023; NCT04681833 (n=120) | N/A | PhI: Safe & well tolerated, significant Ab response (90% in low dose groups, 100% in high dose groups) |
RSVPre-F3/GSK3844766A | GlaxoSmithKline | N/A | PreF3 | Non-adjuvanted | Induce immune response with stabilized pre-F | O | IM | III | N/A | Jan 2019-Nov 2020; NCT04090658/NCT03814590/NCT04657198 (n=1055) | Jan 2019-Nov 2020; NCT04090658/NCT04657198 (n=1055) | Feb 2021May 2024; NCT04886596/NCT04732871/NCT05059301 (n=25000; n=1720; n=750) | PhI/II: humoral and cellular immune responses in all vaccines. Older adults: Higher humoral response (mostly neutralizing) with higher dosage & higher cellular response with adjuvant |
RSVPreF3/GSK3888550A | GlaxoSmithKline | N/A | PreF3 | Non-adjuvanted | Induce immune response with stabilized pre-F | M | IM | III | N/A | Oct 2018-Sep 2019; NCT03674177 (n=502) | Jul 2020-May 2021; NCT04126213 (n=534) | Nov 2020 – Feb 2024 NCT04605159 (GRACE, n=20,000) Sep 2021May 2022; NCT05045144 (n=1541) | PhI/II: robust increase in maternal RSV-specific Ab responses and RSVA/RSV-B nAb titres. Successful Ab transfer to fetus until 6 months after birth |
RSVPreF | Pfizer | Bivalent stabilized prefusion F, sequence based on contemporary RSV A and B strains | pre-F | Non-adjuvante d or Al(OH)3 or alum-adjuvanted | Induce immune response with stabilized pre-F | M | IM | III | Animal studies, specifics unknown | Aug 2020-Sep 2021 & Nov 2020-Aug 2021; NCT03529773/CHIM: NCT04785612/NCT04071158/NCT04032093 (n=1253; n=62; n=713;n=1154) | Jun 2020Nov 2023; NCT04424316 (n=10000) | PhI/II: 18 – 49-yearolds: Safe & well tolerated. Immunization elicited 10- to 20-fold increases in neutralizing Ab titers | |
RSVPreF | Pfizer | Bivalent stabilized prefusion F, sequence based on contemporary RSV A and B strains | pre-F | Non-adjuvanted | Induce immune response with stabilized pre-F | O | IM | III | Animal studies, specifics unknown | Aug 2019-Sep 2021 & Nov 2020-Aug 2021; NCT03529773/HCT NCT04785612/NCT04071158/NCT04032093 (n=1253; n=62; n=713;n=1154) | Aug 2021 – Jun 2024; NCT05035212 (n=30000) | PhI/II: 18–49-year-olds: Safe & well tolerated. Immunization elicited 10- to 20-fold increases in neutralizing Ab titers | |
PARTICLE-BASED | |||||||||||||
V306-SVLP | Virometix | The platform uses synthetic peptides conjugated to a synthetic nanoparticle made from Self-assembling lipopeptides | V-306 | Pam2Cys | Synthetic VLP displays a ‘universal’ T-helper epitope, lipid component (Pam2C) and mimetic of the Palivizumab epitope (FSIIm) | M | IM with epicutaneous skin patch boosters | I | Mice, Rabbits | Sep 2020-Mar 2022; NCT04519073 (n=60) | N/A | N/A | N/A |
IVX-121 | Icosavax | Self-assembling VLP platform technology to deliver stabilized trimeric pre-F proteins | Stabiliz ed PreF DS-Cav1 | Non-adjuvante d/alum | presentation of DS-Cav1 on computationally designed VLP generates a neutralizing Ab response against pre-F protein | O | IM | I | Mice | July 2021 – 2022; 2020–003633-38 (n=90) | N/A | N/A | N/A |
NUCLEIC ACID | |||||||||||||
mRNA-1345 | Moderna | Lipid nanoparticle containing optimized protein and codon sequences | pre-F | Non-adjuvanted | mRNA encodes for a stabilized pre-F glycoprotein eliciting neutralizing antibodies | O | IM | II/III | N/A | Sep 2020-Sep 2023; NCT04528719 (n=100 healthy adults, 300 older adults, 180 women, 40 children) | PhII/III Nov 2021-Nov 2024 (n=34000) | PhII/III Nov 2021-Nov 2024 (n=34000) | PhI: well tolerated at doses up to 200 μg. GMC-fold rise in nAbs at 1 month ≥9.8 for RSV-A and ≥5.3 for RSV-B; 3 doses helped maintain peak titers through month 5 in younger adults |
mRNA-1345 | Moderna | Lipid nanoparticle containing optimized protein and codon sequences | pre-F | Non-adjuvanted | mRNA encodes for a stabilized pre-F glycoprotein eliciting neutralizing antibodies | P | IM | I | N/A | Sept 2020 - Sept 2023 NCT04528719 (n=40 children 12-59 months) | N/A | N/A | N/A |
RECOMBINANT VECTORS | |||||||||||||
MVA-BN RSV | Bavarian Nordic | MVA-BN platform technology | F, G (A & B subtype), N and M2 | Non-adjuvanted | Simulate robust T cell response against 5 RSV antigens nand moderate humoral response against both RSV subtypes | O | IM | III | BALB/c mice, cotton rats | Aug 2015-May 2016; NCT02419391 (n=63) | Sep 2016-Dec 2018 & Feb 2021-Jun 2021; NCT02873286/NCT04752644 (n=420; n=73) | To be announced end 2021 | PhII: Broad and durable Ab & T cell response, significant booster response after 1 year PhII: HCT: Significant reduction in viral load & no vaccine-related SAE |
AD26.RSV.Pre-F | Johnson & Johnson | human cell line, PERC6 (Ad26) encoding RSV F from RSV-A2 strain | pre-F | Non-adjuvanted | Replicationin-competent Adenovirus 26 containing DNA for RSV F protein from A2 strain stabilized in pre-fusion conformation | O | IM | III | Neonatal & adult mice | Multiple trials between Nov 2016Jan 2021; NCT02926430/NCT03795441/NCT04354480 (n=73; n=24; n=36) | Multiple trials between Oct 2017-Jun 2022; NCT04453202/NCT03502707/NCT03303625/NCT03334695/NCT03339713/NCT03982199 (n=459; n=669; n=48; n=64; n=180; n=5815) | Jul 2021-Jan 2024; NCT04908683 (n=23000) | PhI: Safe in older adults and sustaine immune responses after 2 years PhII: 80% vaccine efficacy and robust cellular and humoral immune response. PhII CHIM: Lower viral load and lower infection rate and disease severity in intervention group. |
AD26.RSV.PreF | Johnson & Johnson | human cell line, PERC6 (Ad26) encoding RSV F from RSV-A2 strain | Pre-F | Non-adjuvanted | Replication-incompetent Adenovirus 26 containing DNA for RSV F protein from A2 strain stabilized in pre-fusion conformation | P | IM | I/II | Neonatal & adult mice | Nov 2017-Apr 2020 & Jan 2019-Nov 2021; NCT03303625/NCT03606512 (n=48; n=38) | Nov 2017-Apr 2020 & Jan 2019-Nov 2021; NCT03303625/NCT03606512 (n=48; n=38) | N/A | PhI/II: Well-tolerated and elicited both humoral and cellular immune responses |
IMMUNOPROPHYLAXIS | |||||||||||||
Narsyn (palivizumab) | UMC Utrecht | Intranasal formulation of humanized mouse mAb | N.A | Non-adjuvanted | mAb targeting site II of the F-protein of RSV; neutralization | P | IN | II | Balb/c mice | Oct 2018-Nov 2018; NTR7378 (n=19) | Nov 2018 – Apr 2020 NTR7403 (n=408) | N/A | PhI: Safe in healthy adults |
Clesrovimab (MK 1654) | Merck | In vitro optimized human mAb with 3 YTE mutations in Fc-domain | N/A | Non-adjuvanted | mAb targeting site IV of the F-protein of RSV with extended half-life; neutralization | P | IM/IV | I/II/III | Cotton rats | Jun 2017-Feb 2019 (n=152 adults); Sep 2018-Sep 2022; NCT03524118 (n=180 infants) | Sep 2018-Sep 2022 NCT03524118 (n=180 intants) Mar 2020-Aug 2020 NCT04086472 (CHIM, n=80) | Nov 2021-Aug 2025 NCT0493883 0 (n=1000 high-risk infants) Apr 2021-Sep 2024 NCT04767373 (n=3300 healthy infants) | PhI: Safe in adults CHIM: efficacy 0.62 (95%CI −0.05–0.86) for prevention of RSV-A LRTI |
Nirsevimab, MEDI8897 | Astra Zeneca, MedImmune LLC | In vitro optimized human mAb with YTE mutation in Fc | N/A | Non-adjuvanted | mAb targeting site 0 of the F-protein of RSV with an extended half-life; neutralization | P | IM | III | Cotton rats, cynomolgus, monkeys | Apr 2015-June 2015 & Jan 2015-Sep 2016; NCT02114268/NCT02290340 (n=342; n=151) | Nov 2016-Jul 2018; NCT02878330 (n=1453) Aug 2020 – Jan 2023 NTC04484935 (n=100 immunocompr omised children) | Jul 2019-May 2022; NCT0395948 8 (MEDLEY n=925 high-risk children) Jul 2019 – Mar 2023; NCT0397931 3 (MELODY; n=3000 healthy children) | PhIIb: safety and tolerability similar to palivizumab in 25 countries worldwide Ph III interim: 75% efficacy against medically attended RSV LRTI |
RSM01 | Gates MRI | In vitro optimized human mAb with YTE mutation in Fc | N/A | Non-adjuvanted | mAb targeting site 0 of the F-protein of RSV with an extended half-life; neutralization | P | IM/IV | I | N/A | Nov 2021 – Feb 2022 NCT05118386 (n=56) | N/A | N/A | N/A |
CHIMERIC | |||||||||||||
SeV/RSV | NIAID | modified mouse parainfluenza virus type 1 | F-Protein | Non-adjuvanted | RSV F-expressing SeV carrier | P | IN | I | AGM | May 2018-Feb 2019; NCT03473002 (n=21) | N/A | N/A | N/A |
rBCG-N-hRSV | Pontificia Universidad Católica de Chile | Live-attenuated recombinant Mycobacteriu m bovis BCG (rBCG) based on Danish strain 1331 that expresses N | N-Protein | Non-adjuvanted | rBCG used as a vector to deliver RSV N | P | ID | I | Mice; Holstein calves | Jun 2016-Jun 2018; NCT03213405 (n=24) | N/A | N/A | PhI: Safe & well-tolerated. No SAEs. Humoral and cellular response against N and PPD. |
LIVE-ATTENUATED | |||||||||||||
RSV-MinL4.0 | Codagenix | Vero Master Cell Bank derived from Vero WHO-Seed Lot 1087 | All viral proteins | Non-adjuvanted | Codon-pair deoptimization (CPD) of the L gene | O | IN | I | Non-human primates | Jul 2020-May 2021; NCT04295070 (n=36) | N/A | N/A | N/A |
RSV-MinL4.0 | Codagenix | Vero Master Cell Bank derived from Vero WHO-Seed Lot 1087 | All viral proteins | Non-adjuvanted | Codon-pair deoptimization (CPD) of the L gene | P | IN | I | Non-human primates | Mar 2022-Feb 2023; NCT04919109 n=36) | N/A | N/A | N/A |
IT-RSVΔG | Intravacc | Reverse genetics used to construct LAV from which G protein deleted from RSV genome (RSVΔG) | All viral proteins | Non-adjuvanted | severely impaired binding to host cells due to lacking G-protein and therefore reducing infectivity | P | IN | I | Cotton rats | May 2018-Mar 2019; NTR7173 (n=48) | N/A | N/A | PhI: Safe & well-tolerated. neutralizing antibody response limited in seropositive adults. |
MV-012–968 | Meissa | AttenuBlock synthetic biology platform used to construct LAV with codon deoptimizatio n of NS1/NS2/G, SH deletion, and ablation of secreted form of G | All viral proteins | Non-adjuvanted | Reduced NS1 and NS2 expression for enhanced immunogenicity, SH deletion and G deoptimization for attenuation | P | IN | I/II | Balb/c mice; Cotton rats | Jan 2020-Aug 2020 & Jun 2020-May 2021&Jun 2021-Oct 2022; NCT04227210/NCT04444284/NCT04909021 (n=20; n=34; n=45) | Dec 2020-May 2021; NCT04690335 (n=60) | N/A | PhI: well-tolerated, heavily attenuated, and induces an RSV-specific mucosal IgA response in healthy seropositive adults and pediatric participants |
RSV ΔNS2/Δ1313 /I1314L | NIAID (Sanofi) | NS2 gene deletion and deletion of L codon 1313; I1314L stabilizing mutation; via reverse genetics | All viral proteins | Non-adjuvanted | NS2 deletion bolsters innate response. Deletion at position 1313 of L protein, and I1314L stabilization confers moderate temperature sensitivity | P | IN | I/II | Mice and chimpanz ees | Multiple trials between Jun 2013Apr 2023; NCT03227029/NCT03422237/NCT03916185/NCT01893554 (n=65; n=80; n=160; n= 105) | May 2019-Apr 2023; NCT03916185 (n=160) | N/A | Genetically stable. Attenuated yet immunogenic in RSV-seronegative children, warranting further evaluation |
RSV LID/ΔM2-2/1030s | NIAID (Sanofi) | M2–2 deletion via reverse genetics and temperature sensitivity mutation 1030s in the L polymerase protein | All viral proteins | Non-adjuvanted | Deletion of regulator factor M2–2causes inefficient replication but high immunogenicity; temperature sensitive mutation at position 1030 of L gene | P | IN | I | Mice, African green monkeys | Sep 2020-Apr 2022; NCT04520659 (n=81) | N/A | N/A | 85% of vaccinees shed LID/ΔM2-2/1030s vaccine & ≥4-fold rise in serum-neutralizing Abs |
RSV 6120/ΔNS2/1030s | NIAID (Sanofi) | NS2 gene deletion and temperature sensitivity mutation (1030s) in the L polymerase protein | All viral proteins | Non-adjuvanted | NS2 deletion bolsters innate response, 1030s mutation confers moderate temperature sensitivity | P | IN | I/II | N/A | May 2019-Apr 2023 & Oct 2017-May 2021; NCT03916185/NCT03387137 (n=160; n=45) | May 2019-Apr 2023; NCT03916185 (n=160) | N/A | N/A |
RSV 6120/F1/G2/ΔNS1 | NIAID (Sanofi) | An optimized NS1 gene-deleted live attenuated vaccine | All viral proteins | Non-adjuvanted | NS1 gene-deletion; increased F and G expression by moving to first and second genome positions | P | IN | I | N/A | Jun 2018-dec 2023; NCT03596801 (n=75) | N/A | N/A | N/A |
RSV 6120//ΔNS1 | NIAID (Sanofi) | An optimized NS1 gene-deleted live attenuated vaccine | All viral proteins | Non-adjuvanted | NS1 genedeletion | P | IN | I | N/A | Jun 2018-dec 2023; NCT03596801 (n=75) | N/A | N/A | N/A |
VAD00001 (SP0125) | Sanofi (NIAID) | live, attenuated virus | N/A | Non-adjuvanted | N/A | P | IN | I/II | Mice | Sep 2020-apr 2023; NCT04491877 (n=300) | Sep 2020-apr 2023; NCT04491877 (n=300) | N/A | N/A |
Ab(s) = Antibody(-ies)
AE = Adverse events
AGM = African Green Monkey
CSA =
DNA = Deoxyribonucleic acid
HCT = human challenge trial
ID = Intradermal
IM = Intramuscular
IN = Intranasal
N= RSV nucleoprotein
N/A = not applicable or not available
M = Maternal mAb = Monoclonal Antibody
O = Adults /Older adults
P = Pediatric
pre-F = Pre-fusion protein
RSV = Respiratory Syncytial Virus
SAE = Serious adverse events
URT = Upper respiratory tract
VLP = Virus-like Particle
Lessons Learned
We examine lessons learned from three late phase clinical trial failures since our last review. The PREPARE trial was a milestone, the first phase III trial of an RSV maternal vaccine30. More than 4000 pregnant women received an RSV F nanoparticle vaccine or placebo (2:1 ratio) during the third trimester. RSV maternal vaccination was determined to be safe. Although the vaccine did not meet the primary endpoint, the candidate showed the first proof-of-concept for efficacy of RSV maternal immunization against severe RSV infection in infants. Efficacy was shown through day 90 in South Africa, where more than 50% of participants were enrolled: 56% (95% CI 33–71%) against medically significant RSV LRTI and 74% (95% CI 50–86%) against RSV LRTI with severe hypoxemia. Moreover, there was 49% efficacy against all-cause infant pneumonia through one year after vaccination30. The difference in efficacy may be explained by hospitalization for less severe disease and lower background rates of severe RSV infection in HICs compared to LMICs. Lessons learned include geographical heterogeneity of RSV disease burden and potential efficacy between different countries, the importance of timing of vaccination in relation to RSV season and gestational age30. Furthermore, it was shown that RSV-neutralizing antibodies and F surface glycoprotein binding antibodies were correlated with protection against RSV LRTI with severe hypoxemia, e.g. a vaccine-induced maternal anti-F IgG fold-rise of around 16-fold from the maternal enrollment to day 14 was associated with a baseline covariate-adjusted vaccine efficacy (VE) of 75%31. Proven efficacy poses an ethical dilemma that a potentially life-saving vaccine may not become available in these countries as drug development was discontinued because prespecified criteria for efficacy were not met32. A roll-over trial may be considered to confirm efficacy and develop this vaccine for LMICs.
At the time of our last review in 201825, analysis of the late-stage clinical trial failure of REGN2222 (suptavumab), an antigenic site V monoclonal antibody which did not meet its primary endpoint, had not yet been made public. In a phase III study in 18 countries, it was demonstrated that suptavumab did not reduce RSV hospitalization or outpatient RSV LRTI due to a natural mutation in the predominant circulation strain of RSV subgroup B that resulted in loss of antibody binding and neutralization. There were no changes in circulating RSV A strains and negligible anti-suptavumab antibody responses. Post-hoc analysis suggested the antibody was relatively efficacious against the subgroup A strains but not the new circulating B strain; the relative risk for RSV subgroup A hospitalization or outpatient LRTI versus placebo was 0.38 (95%CI 0.17–0.86)33. These findings highlight the importance of characterization of the viral fitness of monoclonal antibody resistant viral mutants (MARMs) in clinical development and the risk associated with targeting a single viral epitope as well as more potential variability of certain targeted antigenic sites.
Finally, ChAd155.RSV, a recombinant chimpanzee adenovirus vector vaccine expressing RSV F, N and M2–1 proteins, was in development for the pediatric population. Development was halted after preliminary analyses of a phase II trial in infants aged 3–7 months showed that the target efficacy profile was unlikely to be met34. The published first-in-human trial in healthy adults showed adequate safety as well as increased specific humoral and cellular immune responses35. The results of the phase II study have not yet been published so further lessons learned and analysis of this failure are pending. Potentially the choice of vaccine antigens was not optimal for an effective immune response.
Live-attenuated
Live-attenuated vaccines (LAV) are designed to generate a potent immune response (including a local mucosal antibody and cellular response) by mimicking natural infection while being attenuated for reduced virulence. Genetic stability is important to limit the chance of reversion to wildtype virus. A better understanding of the RSV genome and reverse genetics has allowed the rational design of LAV candidates by deleting (or reducing the expression of) proteins known to be important in RNA synthesis regulation or interference with host immune responses (M2–2, NS2, SH and G) leading to restricted viral replication36.
An analysis of the compiled results of seven phase I trials using intranasal LAV (n=239 children 6–24 months of age) provides information on vaccine safety, efficacy and duration of protection of RSV LAV candidates36. LAV are considered safe following first exposure, since vaccine-enhanced disease has not been detected following LAV immunization, although LAV have the potential to induce upper respiratory illness if attenuation is insufficient36. Estimated efficacy from compiled data of five vaccine candidates was 67% (95%CI 24–85) against medically-attended RSV acute-respiratory illness and 88% (95%CI −9–99) against medically-attended RSV LRTI. On an immunologic level, a 4-fold rise in RSV-plaque reduction neutralizing antibody titer (RSV-PRNT) was predictive of vaccine efficacy and responses was durable through one year post vaccination36.
Currently there are six phase I trials and four candidates that have progressed to phase II trials The NIH/NIAID and others are developing LAV candidates with an NS2 deletion and temperature-sensitivity mutation: RSV ΔNS2/Δ1313/I1314L (phase II)37; and RSV 6120/ΔNS2/1030s (phase II).38,39 MV-012–968 (altered NS1/NS2 and G proteins, SH deletion, and ablation of secreted G protein40) has been shown to be safe and to generate a mucosal IgA response in seropositive adults and children41. A safety trial in seronegative children and a human challenge trial in healthy adults to demonstrate efficacy are being conducted for this vaccine candidate42–44. IT-RSV-ΔG (lacking the G protein) was safe in seropositive healthy adults. However, the serum neutralizing antibody response was limited, and nasal IgA antibodies were below the level of detection; immunogenicity needs to be further studied in children and eventually in seronegative infants45. Other candidates include LIDΔ M2–2/1030s46, 6120/ΔNS1, and 6120/F1/G2/ΔNS1 in phase I trials, and VAD00001 in phase II trial. RSV-MinL4.0 (altered polymerase gene) showed a humoral and cellular immune response comparable to wildtype infection in non-human primates and is currently in phase I trials47,48. Overall, LAV provide an important needle-free tool for active intranasal immunization older infants who will not be sufficiently protected by a mAb or maternal vaccine. Moreover, a relatively small sample size (n=540) is needed for a phase III trial in this population36. Further clinical development using this vaccination approach may impact pediatric health directly by reducing pediatric infections and infections in older adults indirectly through herd immunity.
Chimeric
Chimeric live virus vaccine candidates express RSV proteins in related attenuated viruses with favorable safety profiles. In contrast to vectored vaccine candidates, chimeric show favourable antigen presentation which activates an adaptive immune response49,50. There are two chimeric RSV vaccine candidates in phase I trials. One of these candidates uses a replication-deficient Sendai virus modified to express RSV F protein (SeV/RSV)49 and the other uses a live-attenuated recombinant BCG vector expressing RSV N protein (rBCG-N-hRSV) administered via the intradermal route. The latter vaccine candidate was found to be safe in phase I trials51.
Subunit
Subunit vaccines are protein based; this approach has been avoided in RSV-naïve children due to the FI-RSV experience with ERD in which it became clear ERD is a concern for persons not primed with live virus infection52. In parallel to the phase III failure of a post-F subunit vaccine candidate, five vaccine candidates have adopted pre-F as vaccine antigen. Currently, eight subunit candidates are in development for two different target populations: pregnant women and older adults. We discuss vaccine candidates using F antigens first, followed by candidates employing non-F antigens.
The phase I results of DS-Cav1, a subunit vaccine using stabilized pre-F developed by the NIAID/NIH, provide proof-of-concept of structure-based vaccine design. Vaccination resulted in >10-fold increase in serum neutralizing activity53 and is sustainable for an entire RSV season54. Two other candidates use a stabilized pre-F protein as vaccine antigen. RSVpreF (PF-06928316) is a bivalent (subtype A and B) stabilized pre-F without adjuvants. A phase II trial was conducted in nonpregnant women with RSVpreF co-administered with Tdap which showed safety and noninferiority relative to RSVpreF alone55. The anti-pertussis response was inferior (GMC between 0.59 – 0.8 for pertussis antigens compared to Tdap alone) yet the clinical significance of these findings is still unclear and did not differ when adjusted for age56. The phase III MATISSE trial in pregnant women was started in 2020 and is expected to be unblinded Q4 of 202357. A human challenge trial58 showed 75% efficacy of RSVpreF against RSV infection and informed dose and formulation selection for the maternal vaccine candidate59. The phase III RENOIR trial using the same vaccine candidate has started in the fall of 2021 in 30,000 older healthy and high-risk adults60. Another pre-F subunit vaccine, RSVPreF3, is in phase III clinical trials without adjuvant (GSK3888550A) for RSV maternal immunization to protect infants “GRACE” trial61 and with AS01 adjuvant (GSK3844766A)62 to protect the older adult population. Development has been paused for the maternal vaccine candidate in February 2022 due to a safety signal. The older adult candidate was safe and induced approximately a tenfold increase in pre-F IgG and IgA antibodies (n=48 18–40 year old adults; n=1005 60–80 year old adults) in phase I/II clinical trials63–65. For the maternal candidate, phase I/II studies showed a 14-fold increase in RSV A and B neutralizing antibody titers one week after vaccination and maintaining a >6 fold increase after 91 days in healthy non-pregnant women (n=502)65. In the phase III study of the maternal vaccine candidate, the immune response was durable as antibody levels for vaccinees remained elevated against RSV A and RSV B for 6 months after birth66. Registration of RSVpreF maternal vaccine is expected in 2024 and RSVPreF older adult and both RSVPreF3 vaccine candidates in 2025 assuming registration is obtained within one year after phase III completion date according to the clinical trial registry. There are three protein-based vaccines in clinical development that use non-F viral antigens. First, BARS13 uses RSV G protein as an antigen and cyclosporine A (CSA) immunosuppressant to induce Treg cells. ADV110 was safe and immunogenic in phase I67 and is now in phase II68 trials. Second, DPX-RSV, uses the ectodomain of RSV A SH protein (SHe) as a vaccine antigen formulated in depot-based lipid-in-oil delivery platform to allow for prolonged antigen and adjuvant exposure. The proposed mechanism of action against this antigen is generation of SHe-specific antibodies which promote clearance of RSV-infected cells by alveolar macrophage phagocytosis. DPX-RSV showed safety and immunogenicity in a phase I first-in-human trial in older adults69,70. Finally, VN-0200, uses VAGA-9001a as antigen and an MABH-9002b adjuvant (phase I)71. We were not able to define the biological background of VAGA-9001a.
Particle-based
Particle-based vaccines harness the immunogenic potential of displaying multiple antigens via particle assembly. IVX-121 uses a self-assembling synthetic virus-like particle (SVLP) platform technology to deliver 20 copies of stabilized trimeric pre-F proteins (DsCav-1). The computationally designed nanoparticle allows for stabilization of the pre-F protein and in vitro adjustment of antigen density. IVX-121 showed 10 times higher neutralizing antibody responses than DSCav1 alone in preclinical studies72. A phase I trial73 started in 2021 with first results expected in 2022. Following completion of the monovalent RSV candidate trial, the company plans to shift to development of a bivalent VLP vaccine with both RSV and human metapneumovirus (hMPV) antigens.
A second particle-based vaccine candidate, V306-VLP, uses a synthetic VLP to display a site II F protein epitope. The vaccine platform uses conformationally constrained synthetic peptides conjugated to a synthetic nanoparticle made from self-assembling lipopeptides containing a T-helper epitope and toll-like receptor (TLR) ligand74. The vaccine candidate aims to boost preexisting immunity in pregnant women or older adults75. A phase I trial is being conducted in healthy women76. The needle-free intradermal delivery route via an epicutaneous patch is being explored for boosters and has shown similar antibody titers for pertussis as a commercial vaccine in a phase I trial75 but may require a delivery enhancement procedure to optimize vaccine delivery. Overall, particle-based vaccines are still in early development but have the potential to elicit a powerful immune response for pregnant women and the elderly.
Nucleic acid
mRNA vaccines have shown safety and high efficacy against SARS-CoV-2 infection and were developed based on previous work for RSV. Both mRNA COVID-19 vaccines express stabilized versions of SARS-CoV-2 prefusion spike protein patterned after the success of RSV pre-F as a vaccine antigen. The extensive work on RSV vaccine-associated enhanced respiratory disease was also important for the rapid development of COVID-19 vaccines and provided regulatory guidelines for vaccine safety. Because of the successful scale-up and establishment of a robust supply chain, mRNA will be a new vaccine modality available for other purposes including RSV vaccines. An mRNA vaccine (mRNA-1345) encodes stabilized RSV pre-F and uses the same lipid nanoparticle formulation as for the SARS-CoV-2 vaccine SpikeVax that is known to induce and boost antibody and T cell responses including CD8, Th1, and Tfh. The interim results of the phase I trial in younger and older adults showed favorable safety and potent boosting of neutralizing activity77. A phase II/III trial (ConquerRSV) started in November 2021 with 34,000 older adults >60 years of age78. The company intends to combine mRNA-1345 with mRNA-1653 (an mRNA vaccine against two other pediatric viruses, hMPV and parainfluenza virus type 3 (PIV-3)) intended for use in the pediatric population. A phase 1 trial is ongoing in women of childbearing age and seropositive children.
Recombinant vectors
Recombinant vector vaccines use a modified replication-defective virus to induce humoral and cellular immunity by delivering genes for RSV antigens; 3 such candidates are currently in clinical development for the pediatric and elderly population.
Firstly, MVA-BN-RSV uses a poxvirus vector, modified vaccinia Ankara virus (MVA), to express RSV surface antigens F, and G (A&B) and intracellular proteins M2 and N79. In a phase I trial cellular and humoral immune responses were similar in younger and older adults79. Results of a phase IIa human challenge study (n=61) showed 79% reduction in symptomatic RSV infection and a significant reduction in viral load80. The phase II trial in older adults showed elevated antibody responses for 6 months which can be safely boosted at 12 months81. Following dose selection from the phase II trial, preparations for a phase III trial are ongoing82. Ad26.RSV.preF is being developed for two different target populations: (1) pediatric (phase II) and (2) elderly (phase III). Ad26.RSV.preF vaccine candidate uses an adenoviral vector to express the RSV F protein in the pre-F conformation83. The vaccine candidate showed improved immunogenicity in comparison to the previous vaccine candidate with post-F RSV protein (Ad26.RSV.FA2). In neonatal mice, the vaccine candidate showed a Th1-biased response84. A durable humoral and cellular immune response was shown for at least 2 years postimmunization in the first-in-human study in older adults85. Proof-of-concept was obtained in the first RSV vaccine human challenge study86. In the primary efficacy results from the proof-of-concept CYPRESS study, Ad26.RSV.preF showed 80% (95%CI: 52–93) efficacy against RSV LRTI through the first RSV season in the older adult population87. Ad26.RSV.preF was found to be safe and well-tolerated88 and showed efficacy in adults ≥65 years with (68%, 95%CI: −27–95) or without risk factors (85%, 95%CI: 50–97)89. Furthermore, there was no interference when an RSV vaccine was co-administered with seasonal influenza vaccine in older adults in a phase II trial, and the vaccine candidate was determined to have an acceptable safety profile though showing increased reactogenicity compared to influenza vaccination90. In 2019, this candidate was granted FDA breakthrough therapy designation. Subsequently, the phase III EVERGREEN trial was started in Q3 2021 and will examine efficacy in across two RSV seasons in 23 000 adults aged 60 years and above91. For the pediatric vaccine candidate a phase I/II trial in seropositive infants aged 12–24 months showed Ad26.RSV.PreF was well-tolerated and elicited both humoral and cellular immune responses92. Of note, a Sars-CoV-2 adenovirus vector vaccine candidate uncovered new safety concerns with adenoviral vector vaccines including vaccine-induced immune thrombotic thrombocytopenia (VITT) which was observed for at least two of the COVID-19 adenoviral vector vaccines.93
mAbs
Monoclonal antibodies have been labelled as the “magic bullet” against infection given their high pathogen specificity94. For RSV, increased knowledge of the structure and immunogenicity of the RSV F protein has resulted in next generation antibodies targeting highly neutralization-sensitive epitopes located on the RSV pre-F protein. Furthermore, next generation RSV antibodies have been engineered with Fc mutations to extended half-life and enable protection of all infants against lower respiratory tract disease for an entire RSV season. The leading candidate is nirsevimab (formerly MEDI-8897), a human mAb targeting site Ø of the F protein with a YTE mutation in the Fc portion to allow for an extended half-life. In phase II trial results (n=1453) nirsevimab showed 70% (95%CI 52–81) efficacy against medically-attended RSV LRTI and 78% (95%CI 52–90) against RSV hospitalization in preterm infants95, which is similar to the phase III trial interim results: 75% (95%CI 50–87) against RSV LRTI and 62% (95%CI −9–87) against RSV hospitalization among healthy late preterm and full-term infants (n=1490)96. The safety profile of nirsevimab is similar to that of the current standard-of-care, monthly palivizumab, administered to infants with congenital heart or lung disease (n=310) and preterm infants 29 to <35 weeks gestational age (n=615)96. RSV MARMs were generated and were shown not to have an impact on viral replication and had a low natural frequency amongst circulating strains97. The major advantages of nirsevimab in contrast to the currently approved palivizumab are: (1) a single intramuscular injection protects infants for an entire season compared to monthly doses, and (2) reduced costs (vaccine-like pricing expected) allowing for administration to all infants compared to only high-risk children.
Clesrovimab (MK-1654), an extended half-life mAb with the same YTE mutation as nirsevimab, targets site IV (though preferentially binding pre-F due to partial targeting of site V) of the RSV F protein. This mAb is currently in phase IIb/III clinical trials in infants. This mAb has shown high potency against RSV clinical isolates in vitro and is equipotent against RSV subgroup A and B strains98. A human challenge trial (n=70) showed reduced viral load after viral challenge and reduced RSV symptomatic infection rates99. A meta-analysis was performed to assess the relationship between serum neutralizing antibodies and clinical endpoints; the study estimated a single 75 mg dose would have more than 75% efficacy lasting 5 months in term infants100. The company developing this candidate has committed to helping navigate uncertainty and improving issues of access through ongoing research and innovation to help address the burden of potentially preventable childhood diseases (personal communications, Andrew W Lee).
Affordability remains a key consideration for mAb development as it is a potential barrier to global access. There are three different clinical development efforts underway to circumvent this problem: (1) an affordable extended half-life site Ø mAb (2) local administration, and (3) a biosimilar. First, A phase I trial of RSM01101, a site Ø mAb, targeting the LMICs with a target price of less than $5 per dose102. Second, local needle-free administration of palivizumab, a market-approved site II mAb, via nose drops may significantly reduce costs by reducing the drug dose needed103. Results of a phase I and IIb trial will soon be published for intranasal palivizumab administration to prevent RSV infection. Finally, a biosimilar for palivizumab is being developed in a public-private partnership between the Utrecht Center for Affordable Biotherapeutics and MabXience for which a human challenge trial was performed in 2020 (n=56), but the results of this trial have not yet been made publicly available104.
Important considerations for the development of next-generation mAbs: (1) affordability by investing in higher efficiency production or developing biosimilars and potentially through local administration, and (2) viral resistance which needs to be monitored and may be prevented through administration of a cocktail of monoclonals targeting multiple epitopes. Monthly administration of intranasal mAbs or a palivizumab biosimilar may pose a programmatic limitation in most LMICs. Potentially, a combination of monoclonal antibodies targeted to different epitopes may provide a solution to loss of efficacy due to viral resistance. However practical barriers exist to this solution as the combination would have to consist of separately registered antibodies. Thus far, the epitopes for mAbs currently in development are highly conserved with minimal naturally occurring antibody-resistant strains which have shown similar/lower viral fitness compared to non-resistant viral strains in vitro.
Discussion
In the past decade, the RSV vaccine landscape has undergone a major transition from empirical to rational vaccine design. In two previous reviews25,105 we characterized the dynamics of the RSV vaccine landscape which included multiple late-phase failures. These failures have laid the foundation for future success by guiding development of vaccines: supporting pre-F as a vaccine antigen, highlighting the importance of conducting vaccine trials over more than one RSV season, providing knowledge of a protective immune response, emphasizing the importance of monitoring viral resistance to mAbs, and highlighting the value of CHIM to derisk RSV vaccine development. The number of candidates in late-phase development is expanding: only one mAb and one maternal vaccine candidate were in phase III development as of 2015 and 2018 respectively. Development was halted for both after failure to meet the primary endpoints of the trials, but important lessons learned have been incorporated into current trials. A better understanding of RSV neutralizing epitopes has resulted in rapid expansion to the current eight vaccine candidates in phase III trials. From 2015 to 2018, 60% of vaccine candidates did not continue development. Since 2018, 37% of vaccine candidates did not continue development. However, 30% (3/10) of vaccines are still in development from 2015.
RSV prevention appears to be on the horizon with market access expected for nirsevimab within the next 12 to 24 months. This approval may be followed shortly by approval of a maternal vaccine and a vaccine for older adults (subunit, vector-based and nucleic vaccines in late phase trials). In this case a situation will emerge in which multiple RSV vaccine candidates are approved. If all current phase III trials generate positive results, relative efficacy and safety trial data, delivery strategies, and costs may determine vaccine uptake for different maternal and older adult candidates. Despite the approval of next-generation antibodies, palivizumab may remain on the market to fill the gap in time to global market access of extended-half-life antibodies and because mAb supply may fail to meet global demand. RSV has shown negligible viral resistance against palivizumab after twenty years on the market106. For this reason, despite multidose schedule and costs, palivizumab may act as a back-up prophylaxis strategy while waiting for global real-time viral resistance data upon mAb implementation in HICs.
With both infant immunoprophylaxis and maternal vaccines on the market, it is important to consider how these two prevention strategies aiming to protect young infants will coexist. Maternal vaccines and infant immunoprophylaxis may play a complementary role in the prevention of severe RSV infection during infancy. There is a clear use-case for mAbs even if a maternal vaccine is approved. mAbs are expected to protect premature and full term infants, may have a longer duration of protection than maternal vaccines107,108, may be applied flexibly where RSV seasonality is variable, and may be implemented in cases where maternal immunization did not occur. There is also a use-case for a maternal vaccine in co-existence with an approved mAb. Active maternal vaccination provides broad protection; it is still unclear whether maternal vaccination persists until a subsequent pregnancy and whether booster vaccination provides even stronger protection. Maternal vaccination may also provide an alternative for parents preferring not to vaccinate their babies. Finally, maternal vaccines serve as a back-up in the case of viral resistance to mAbs or prohibitively high costs related to the production of biologicals that limit administration only to select patient populations.
In LMICs, there is a use-case for mAbs to protect young infants: (1) if there is a substantial time gap until a vaccine becomes available, (2) if their mothers are not immunized, and (3) if there is insufficient time for an immune response after vaccination (i.e., premature infants)102. LMICs may even prefer mAbs over maternal immunization due to potential higher coverage and ease of implementation into EPI programs. However, there are several challenges to LMIC implementation, including (1) a year-round RSV circulation (mAb administration at birth may be more cost-effective than seasonal), (2) scalability dependent upon use of multi-dose vials and stability at ambient temperature, (3) limited burden and RSV healthcare utilization data in LMICs to build a case for implementation, (4) trial design in the case nirsevimab reaches the market (requiring a large sample size), and (5) potential limitations of surveillance of viral resistance in LMICs (though potentially enhanced post-COVID). Thus, mAbs could have a high impact in LMICs, but implementation challenges remain.
Passively acquired antibodies from maternal immunization or mAb immunoprophylaxis will wane over time, at which point pediatric vaccines for active immunization may serve an important role. Live-attenuated or replicating vector vaccines do not prime for ERD; delivered via the intranasal route, they are immunogenic in presence of maternal antibodies. Thus, these active immunization approaches may be safe and effective for the older pediatric age group and will be complementary to infant immunoprophylaxis and maternal vaccines.
Although the approval of multiple RSV vaccines is within reach, several obstacles to worldwide access remain. Not only are globally representative trials needed, but vaccine trials need to be performed in countries with the highest disease burden as efficacy can differ between LMIC and HICs as observed for vaccines against both RSV30 and other pathogens109. Access in LMICs may be delayed due to (1) lack of trial data in populations with a high incidence of HIV and malaria as well as (2) regulatory drug lag between time of regulatory submission and approval in Sub-Saharan Africa110. The Vaccine Alliance (GAVI) vaccine investment strategy (VIS) includes RSV and will be important in making RSV prevention available in GAVI-eligible countries. Differential pricing may be an important consideration for non-GAVI eligible countries. Other challenges to global access include measuring protection in the case maternal vaccine boosters are indicated. A correlate of protection for RSV is lacking as well as a simple tool to measure protection following RSV vaccination. It will be important to assess the sustainability of RSV prevention via ongoing genetic surveillance. The development of viral resistance is most relevant for infant immunoprophylaxis with mAbs. A cocktail of mAbs targeting different epitopes may help prevent the emergence of viral resistance. However, there are currently no ongoing clinical trials with multiple mAbs which may hamper approval of a drug cocktail by regulatory bodies such as the approved mAb cocktail to prevent SARS-CoV-2. Finally, awareness of RSV is limited amongst patients, policymakers and healthcare providers and further cost-effectiveness studies of products are needed111. These knowledge gaps related to global vaccine implementation are important future research priorities [Box 1]. Overall, we have reached an exciting phase in which RSV prevention is within reach. It is likely that multiple immunization strategies with complementary value, unique advantages and use-case scenarios will share the RSV prevention landscape. To guarantee worldwide access, urgent steps are required to surmount challenges of measuring protection, monitoring viral resistance, and prioritizing LMIC access and affordability.
Box 1. Future RSV Vaccine and mAb Landscape Research Priorities.
Generating knowledge of RSV awareness
Understanding cost-effectiveness of RSV prevention in different parts of the world
Defining a correlate of protection for RSV
Defining efficacy of RSV prevention in LMICs
RSV genetic surveillance
Supplementary Material
Key Points.
Knowledge of neutralization-sensitive viral epitopes informed a shift from empirical to structure-based vaccine and monoclonal antibody (mAb) design
Market access for an extended half-life RSV mAb for prophylaxis in all infants is within reach in 2023 and may be followed by approval of a maternal vaccine to protect all infants
No vaccine or mAb is within reach for resource poor areas with the highest pediatric mortality burden
Subunit, vector-based, and nucleic acid vaccine approaches are in late-phase trials for older adults
Acknowledgements:
We would like to thank Femke Vernooij, Trisja Boom, and Michelle van Wijk for their excellent assistance in data collection.
UJB was supported by the Intramural Program of the NIAID, NIH.
Declaration of interest:
DMW has received consulting fees from Pfizer, Merck, Affinivax, and Matrivax, unrelated to this manuscript and is Principal Investigator on grants from Pfizer and Merck to Yale University, unrelated to this manuscript.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Lozano R, Naghavi M, Foreman K, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: A systematic analysis for the Global Burden of Disease Study 2010. Lancet 2012; 380: 2095–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Nair H, Brooks WA, Katz M, et al. Global burden of respiratory infections due to seasonal influenza in young children: a systematic review and meta-analysis. Lancet 2011; 378: 1917–30. [DOI] [PubMed] [Google Scholar]
- 3.Srikantiah P, Vora P, Klugman KP. Assessing the Full Burden of Respiratory Syncytial Virus in Young Infants in Low- and Middle-Income Countries: The Importance of Community Mortality Studies. Clin Infect Dis 2021; 73: S177–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Bulkow LR, Singleton RJ, Karron RA, Harrison LH. Risk factors for severe respiratory syncytial virus infection among Alaska Native children. Pediatrics 2002. DOI: 10.1542/peds.109.2.210. [DOI] [PubMed] [Google Scholar]
- 5.Beckhaus AA, Castro-Rodriguez JA. Down syndrome and the risk of severe RSV infection: A meta-Analysis. Pediatrics. 2018. DOI: 10.1542/peds.2018-0225. [DOI] [PubMed] [Google Scholar]
- 6.Resch B, Manzoni P, Lanari M. Severe respiratory syncytial virus (RSV) infection in infants with neuromuscular diseases and immune deficiency syndromes. Paediatr. Respir. Rev 2009; 10: 148–53. [DOI] [PubMed] [Google Scholar]
- 7.Omer SB. Maternal Immunization. N Engl J Med 2017; 376: 1256–67. [DOI] [PubMed] [Google Scholar]
- 8.Ackerson B, Tseng HF, Sy LS, et al. Severe Morbidity and Mortality Associated With Respiratory Syncytial Virus Versus Influenza Infection in Hospitalized Older Adults. Clin Infect Dis 2019; 69: 197–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Falsey AR, Hennessey PA, Formica MA, Cox C, Walsh EE. Respiratory Syncytial Virus Infection in Elderly and High-Risk Adults. N Engl J Med 2005; 352: 1749–59. [DOI] [PubMed] [Google Scholar]
- 10.Korsten K, Adriaenssens N, Coenen S, et al. Burden of respiratory syncytial virus infection in community-dwelling older adults in Europe (RESCEU): an international prospective cohort study. Eur Respir J 2021; 57: 2002688 Print 2021 Apr. [DOI] [PubMed] [Google Scholar]
- 11.Herring WL, Zhang Y, Shinde V, Stoddard J, Talbird SE, Rosen B. Clinical and economic outcomes associated with respiratory syncytial virus vaccination in older adults in the United States. Vaccine 2021; 40: 483–93. [DOI] [PubMed] [Google Scholar]
- 12.Zeevat F, Luttjeboer J, Paulissen JHJ, et al. Exploratory Analysis of the Economically Justifiable Price of a Hypothetical RSV Vaccine for Older Adults in the Netherlands and the United Kingdom. J Infect Dis 2021; jiab118: Online ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Johnson KM, Bloom HH, Mufson MA, Chanock RM. Natural reinfection of adults by respiratory syncytial virus. Possible relation to mild upper respiratory disease. N Engl J Med 1962; 267: 68–72. [DOI] [PubMed] [Google Scholar]
- 14.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. [DOI] [PubMed] [Google Scholar]
- 15.Killikelly AM, Kanekiyo M, Graham BS. Pre-fusion F is absent on the surface of formalin-inactivated respiratory syncytial virus. Sci Rep 2016; 6: 34108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.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: e1004757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Jozwik A, Habibi MS, Paras A, et al. RSV-specific Airway Resident Memory CD8+ T Cells and Differential Disease Severity After Experimental Human Infection. Nat Commun 2015; 6: 10224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Habibi MS, Jozwik A, Makris S, et al. Impaired antibody-mediated protection and defective iga b-cell memory in experimental infection of adults with respiratory syncytial virus. Am J Respir Crit Care Med 2015; 191: 1040–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Chu HY, Steinhoff MC, Magaret A, et al. Respiratory Syncytial Virus Transplacental Antibody Transfer and Kinetics in Mother-Infant Pairs in Bangladesh. J Infect Dis 2014; 210: 1582–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Glezen WP, Paredes A, Allison JE, Taber LH, Frank AL. Risk of respiratory syncytial virus infection for infants from low-income families in relationship to age, sex, ethnic group, and maternal antibody level. J Pediatr 1981; 98: 708–15. [DOI] [PubMed] [Google Scholar]
- 21.Stensballe LG, Ravn H, Kristensen K, et al. Respiratory syncytial virus neutralizing antibodies in cord blood, respiratory syncytial virus hospitalization, and recurrent wheeze. J Allergy Clin Immunol 2009; 123: 398–403. [DOI] [PubMed] [Google Scholar]
- 22.Piedra PA, Jewell AM, Cron SG, Atmar RL, Paul Glezen W. 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. [DOI] [PubMed] [Google Scholar]
- 23.Ngwuta JO, Chen M, Modjarrad K, et al. Prefusion F-specific antibodies determine the magnitude of RSV neutralizing activity in human sera. Sci Transl Med 2015; 7: 309ra162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.PATH. RSV Vaccine and mAb Snapshot | PATH. 2021. https://www.path.org/resources/rsv-vaccine-and-mab-snapshot/ (accessed April 12, 2022).
- 25.Mazur NI, Higgins D, Nunes MC, et al. The respiratory syncytial virus vaccine landscape: lessons from the graveyard and promising candidates. Lancet Infect Dis 2018; 18: e295–311. [DOI] [PubMed] [Google Scholar]
- 26.ClinicalTrials.gov. Identifier: NCT03959488, A Study to Evaluate the Safety of MEDI8897 for the Prevention of Medically Attended Respiratory Syncytial Virus(RSV) Lower Respiratory Track Infection (LRTI) in High-risk Children - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2019. May 22. https://clinicaltrials.gov/ct2/show/NCT03959488 (accessed April 12, 2022). [Google Scholar]
- 27.ClinicalTrials.gov. Identifier: NCT03979313, A Study to Evaluate the Safety and Efficacy of MEDI8897 for the Prevention of Medically Attended Lower Respiratory Tract Infection Due to Respiratory Syncytial Virus in Healthy Late Preterm and Term Infants (MELODY) - Full Text Vi. Bethesda Natl. Libr. Med. (US). 2019. June 7. https://clinicaltrials.gov/ct2/show/NCT03979313 (accessed April 12, 2022). [Google Scholar]
- 28.ClinicalTrials.gov. Identifier: NCT04938830, MK-1654 in Infants and Children at Increased Risk for Severe Respiratory Syncytial Virus (RSV) Disease (MK-1654–007) - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2021. June 24. https://clinicaltrials.gov/ct2/show/NCT04938830 (accessed April 12, 2022). [Google Scholar]
- 29.ClinicalTrials.gov. Identifier: NCT04767373, Efficacy and Safety of MK-1654 in Infants (MK-1654–004) - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2021. Feb 23. https://clinicaltrials.gov/ct2/show/NCT04767373 (accessed April 12, 2022). [Google Scholar]
- 30.Madhi SA, Polack FP, Piedra PA, et al. Respiratory Syncytial Virus Vaccination during Pregnancy and Effects in Infants. N Engl J Med 2020; 383: 426–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Fong Y, Borate B, Zhang W, van der Laan L, Huang Y, Gilbert P. Immunologic Biomarker Correlates of Severe RSV Disease in a Vaccine Efficacy Trial. In: Presentation at RSVVW’21. Virtual: 6th ReSViNet Conference: November 10–12, 2021. Avaiable from: https://www.resvinet.org/uploads/2/2/2/7/22271200/_virtual__rsvvw21_information_brochure_v3.0.pdf, 2021. [Google Scholar]
- 32.Mazur NI, Bont LJ, van Delden JJM, Omer SB. An ethics framework and practical guidance for post-trial access to an RSV maternal vaccine. Lancet Respir. Med 2019; 7: 474–6. [DOI] [PubMed] [Google Scholar]
- 33.Simões EAF, Forleo-Neto E, Geba GP, et al. Suptavumab for the Prevention of Medically Attended Respiratory Syncytial Virus Infection in Preterm Infants. Clin Infect Dis 2021; 73: e4400–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Taylor NP. Biotech GSK culls phase 2 RSV vaccine in kids, as feladilimab flops again in lung cancer. Fierce Biotech. 2021. https://www.fiercebiotech.com/biotech/gsk-drops-phase-2-viral-vector-rsv-vaccine-pediatric-ages#:~:text=Biotech-,GSK culls phase 2 RSV vaccine in kids%2C as,flops again in lung cancer&text=GlaxoSmithKline is shutting down development,become severe in some (accessed Dec 1, 2021). [Google Scholar]
- 35.Cicconi P, Jones C, Sarkar E, et al. First-in-Human Randomized Study to Assess the Safety and Immunogenicity of an Investigational Respiratory Syncytial Virus (RSV) Vaccine Based on Chimpanzee-Adenovirus-155 Viral Vector-Expressing RSV Fusion, Nucleocapsid, and Antitermination Viral Proteins. Clin Infect Dis 2020; 70: 2073–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Karron RA, Atwell JE, McFarland EJ, et al. Live-attenuated Vaccines Prevent Respiratory Syncytial Virus-associated Illness in Young Children. Am J Respir Crit Care Med 2021; 203: 594–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Karron RA, Luongo C, Mateo JS, Wanionek K, Collins PL, Buchholz UJ. Safety and Immunogenicity of the Respiratory Syncytial Virus Vaccine RSV/ΔNS2/Δ1313/I1314L in RSV-Seronegative Children. J Infect Dis 2020; 222: 82–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.ClinicalTrials.gov. Identifier: NCT03227029, Evaluating the Infectivity, Safety, and Immunogenicity of Recombinant Live-Attenuated RSV Vaccines RSV ΔNS2/Δ1313/I1314L or RSV 276 in RSV-Seronegative Infants 6 to 24 Months of Age - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2022. Jan 8. https://clinicaltrials.gov/ct2/show/NCT03227029 (accessed April 12, 2022). [Google Scholar]
- 39.ClinicalTrials.gov. Identifier: NCT03916185, Safety and Immunogenicity of a Single Dose of the Recombinant Live-Attenuated Respiratory Syncytial Virus (RSV) Vaccines RSV ΔNS2/Δ1313/I1314L, RSV 6120/ΔNS2/1030s, RSV 276 or Placebo, Delivered as Nose Drops to RSV-Seronegative C. Bethesda Natl. Libr. Med. (US). 2019. Apr 16. https://clinicaltrials.gov/ct2/show/NCT03916185 (accessed April 12, 2022). [Google Scholar]
- 40.Stobart CC, Rostad CA, Ke Z, et al. A live RSV vaccine with engineered thermostability is immunogenic in cotton rats despite high attenuation. Nat Commun 2016; 7: 13916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Meissa Vaccines I. Meissa Vaccines Provides a Pipeline Update on Vaccine Candidates for COVID-19 and RSV. 2020; published online June 30. https://www.meissavaccines.com/post/meissa-vaccines-provides-a-pipeline-update-on-vaccine-candidates-for-covid-19-and-rsv (accessed April 12, 2022). [Google Scholar]
- 42.ClinicalTrials.gov. Identifier: NCT04227210, Safety and Immune Response to an Investigational Vaccine Against Respiratory Syncytial Virus in Healthy Adults - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2020. Jan 13. https://clinicaltrials.gov/ct2/show/NCT04227210 (accessed April 12, 2022). [Google Scholar]
- 43.ClinicalTrials.gov. Identifier: NCT04909021, Safety and Immunogenicity of an Intranasal Vaccine for Respiratory Syncytial Virus in Seronegative Children 6–36 Months - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2021. Jun 1. https://clinicaltrials.gov/ct2/show/NCT04909021 (accessed April 12, 2022). [Google Scholar]
- 44.Vaccines M. Meissa Announces 1st Dosing in Phase 2 Study of Intranasal Live Attenuated Vaccine Candidate for RSV. 2021; published online Jan 21. https://www.meissavaccines.com/post/meissa-announces-1st-dosing-in-phase-2-study-of-intranasal-live-attenuated-vaccine-candidate-for-rsv (accessed April 12, 2022). [Google Scholar]
- 45.Verdijk P, van der Plas JL, van Brummelen EMJ, et al. First-in-human administration of a live-attenuated RSV vaccine lacking the G-protein assessing safety, tolerability, shedding and immunogenicity: a randomized controlled trial. Vaccine 2020; 38: 6088–95. [DOI] [PubMed] [Google Scholar]
- 46.McFarland EJ, Karron RA, Muresan P, et al. Live-Attenuated Respiratory Syncytial Virus Vaccine With M2–2 Deletion and With Small Hydrophobic Noncoding Region Is Highly Immunogenic in Children. J Infect Dis 2020; 221: 2050–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Mueller S, Stauft CB, Kalkeri R, et al. A codon-pair deoptimized live-attenuated vaccine against respiratory syncytial virus is immunogenic and efficacious in non-human primates. Vaccine 2020; 38: 2943–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Le Nouën C, McCarty T, Brown M, et al. Genetic stability of genome-scale deoptimized RNA virus vaccine candidates under selective pressure. Proc Natl Acad Sci 2017; 114: E386–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Wiegand MA, Gori-Savellini G, Gandolfo C, et al. A Respiratory Syncytial Virus Vaccine Vectored by a Stable Chimeric and Replication-Deficient Sendai Virus Protects Mice without Inducing Enhanced Disease. J Virol 2017; 91. DOI: 10.1128/JVI.02298-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Scaggs Huang F, Bernstein DI, Slobod KS, et al. Safety and immunogenicity of an intranasal sendai virus-based vaccine for human parainfluenza virus type I and respiratory syncytial virus (SeVRSV) in adults. Hum Vaccin Immunother 2021; 17: 554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Abarca K, Rey-Jurado E, Muñoz-Durango N, et al. Safety and immunogenicity evaluation of recombinant BCG vaccine against respiratory syncytial virus in a randomized, double-blind, placebo-controlled phase I clinical trial. EClinicalMedicine 2020; 27: 100517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Anderson LJ, Dormitzer PR, Nokes DJ, Rappuoli R, Roca A, Graham BS. Strategic priorities for respiratory syncytial virus (RSV) vaccine development. Vaccine 2013; 31: B209–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Crank MC, Ruckwardt TJ, Chen M, et al. A proof of concept for structure-based vaccine design targeting RSV in humans. Science 2019; 365: 505–9. [DOI] [PubMed] [Google Scholar]
- 54.Ruckwardt TJ, Morabito KM, Phung E, et al. Safety, tolerability, and immunogenicity of the respiratory syncytial virus prefusion F subunit vaccine DS-Cav1: a phase 1, randomised, open-label, dose-escalation clinical trial. Lancet Respir Med 2021; 9: 1111–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.ClinicalTrials.gov. Identifier: NCT04071158. A study of a RSV vaccine when given together with TDap in healthy nonpregnant women aged between 18 to 49 years - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2019. Aug 28. https://clinicaltrials.gov/ct2/show/NCT04071158 (accessed April 12, 2022). [Google Scholar]
- 56.Peterson JT, Zareba AM, Fitz-Patrick D, et al. Safety and Immunogenicity of a Respiratory Syncytial Virus Prefusion F Vaccine When Coadministered With a Tetanus, Diphtheria, and Acellular Pertussis Vaccine. J Infect Dis 2021; jiab505: 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.ClinicalTrials.gov. Identifier: NCT04424316, A Trial to Evaluate the Efficacy and Safety of RSVpreF in Infants Born to Women Vaccinated During Pregnancy. - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2020. Jun 9. https://clinicaltrials.gov/ct2/show/NCT04424316 (accessed April 12, 2022). [Google Scholar]
- 58.ClinicalTrials.gov. Identifier: NCT04785612, Study of RSVpreF Vaccination and RSV Challenge in Healthy Adults - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2021. Mar 8. https://clinicaltrials.gov/ct2/show/NCT04785612 (accessed April 12, 2022). [Google Scholar]
- 59.ClinicalTrials.gov. Identifier: NCT04032093, A phase 2b placebo-controlled, randomized study of a respiratory syncytial virus (RSV) vaccine in pregnant women-Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2019. Jul 25. https://clinicaltrials.gov/ct2/show/NCT04032093 (accessed April 12, 2022). [Google Scholar]
- 60.ClinicalTrials.gov. Identifier: NCT05035212, Study to Evaluate the Efficacy, Immunogenicity, and Safety of RSVpreF in Adults. - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2021. Sep 5. https://www.clinicaltrials.gov/ct2/show/NCT05035212 (accessed April 12, 2022). [Google Scholar]
- 61.ClinicalTrials.gov. Identifier: NCT04605159, A Phase III Double-blind Study to Assess Safety and Efficacy of an RSV Maternal Unadjuvanted Vaccine, in Pregnant Women and Infants Born to Vaccinated Mothers - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2020, Oct 27. https://clinicaltrials.gov/ct2/show/NCT04605159 (accessed April 12, 2022). [Google Scholar]
- 62.ClinicalTrials.gov. Identifier: NCT04886596, Efficacy Study of GSK’s Investigational Respiratory Syncytial Virus (RSV) Vaccine in Adults Aged 60 Years and Above - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2021. May 14. https://clinicaltrials.gov/ct2/show/NCT04886596 (accessed April 12, 2022). [Google Scholar]
- 63.Tica J, Guiñazú JR, Andrews CP, et al. 119. A Respiratory Syncytial Virus Prefusion F Protein (RSVPreF3) Candidate Vaccine Administered in Older Adults in a Phase I/II Randomized Clinical Trial Is Well Tolerated. Open Forum Infect Dis 2020; 7: S187–S188. [Google Scholar]
- 64.Guiñazú JR, Tica J, Andrews CP, et al. 121. A Respiratory Syncytial Virus Prefusion F Protein (RSVPreF3) Candidate Vaccine Administered in Older Adults in a Phase I/II Randomized Clinical Trial Is Immunogenic. Open Forum Infect Dis 2020; 7: S188–9. [Google Scholar]
- 65.GSK. GSK presents positive clinical data on maternal and older adults RSV candidate vaccines | GSK. 2020; published online Oct 21. https://www.gsk.com/engb/media/press-releases/gsk-presents-positive-clinical-data-on-maternal-and-olderadults-rsv-candidate-vaccines/ (accessed April 12, 2022). [Google Scholar]
- 66.Kantele A. CS-7: Investigational maternal respiratory syncytial virus vaccine (rsvpref3) boosts maternal immunity against RSV-A and RSV-B, with transplacentally acquired antibodies persisting in infants until six months post-birth. In: RSVVW’21 Abstract Booklet. Virtual: RSVVW’21: 6th ReSViNet Conference: November 10–12, 2021. Available from: https://www.resvinet.org/uploads/2/2/2/7/22271200/abstract_booklet_rsvvw21.pdf, 2021: 13. [Google Scholar]
- 67.ClinicalTrials.gov. Identifier: NCT04851977, A First in Human Study to Evaluate the Safety and Immune Response to a Vaccine for the Treatment of a Respiratory Virus, When Administered Into the Arm in Healthy Adult Participants - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2021. Apr 21. https://clinicaltrials.gov/ct2/show/NCT04851977 (accessed April 12, 2022). [Google Scholar]
- 68.ClinicalTrials.gov. Identifier: NCT04681833, Safety and Efficacy of BARS13 in the Elderly - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2020. Dec 23. https://clinicaltrials.gov/ct2/show/NCT04681833 (accessed April 12, 2022). [Google Scholar]
- 69.Langley JM, MacDonald LD, Weir GM, et al. A Respiratory Syncytial Virus Vaccine Based on the Small Hydrophobic Protein Ectodomain Presented With a Novel Lipid-Based Formulation Is Highly Immunogenic and Safe in Adults: A First-in-Humans Study. J Infect Dis 2018; 3: 378–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Torrey HL, Kaliaperumal V, Bramhecha Y, et al. Evaluation of the protective potential of antibody and T cell responses elicited by a novel preventative vaccine towards respiratory syncytial virus small hydrophobic protein. Hum Vaccin Immunother 2020; 16: 2007–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.ClinicalTrials.gov. Identifier: NCT04914520, Study of VN-0200 in Japanese Healthy Adults and Elderly Subjects - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2021. Jun 4. https://clinicaltrials.gov/ct2/show/NCT04914520 (accessed April 12, 2022). [Google Scholar]
- 72.Marcandalli J, Fiala B, Ols S, et al. Induction of Potent Neutralizing Antibody Responses by a Designed Protein Nanoparticle Vaccine for Respiratory Syncytial Virus. Cell 2019; 176: 1420–1431.e17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Icosavax Inc. Icosavax Provides Corporate Update and Anticipated. Globe Newswire. 2022; published online Jan 7. https://www.globenewswire.com/news-release/2022/01/07/2363089/0/en/Icosavax-Provides-Corporate-Update-and-Anticipated-Milestones-for-2022.html (accessed April 12, 2022). [Google Scholar]
- 74.Zuniga A, Rassek O, Vrohlings M, et al. An epitope-specific chemically defined nanoparticle vaccine for respiratory syncytial virus. NPJ vaccines 2021; 6: 85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Hervé PL, Dhelft V, Zuniga A, et al. Epicutaneous immunization using synthetic virus-like particles efficiently boosts protective immunity to respiratory syncytial virus. Vaccine 2021; 39: 4555–63. [DOI] [PubMed] [Google Scholar]
- 76.ClinicalTrials.gov. Identifier: NCT04519073, Phase 1 Study to Evaluate the Safety and Immunogenicity of a Candidate Vaccine Against Respiratory Syncytial Virus - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2020. Aug 19. https://clinicaltrials.gov/ct2/show/NCT04519073 (accessed April 12, 2022). [Google Scholar]
- 77.Chen GL, Mithani R, Schaefers K, et al. VT&T-53: Safety and Immunogenicity of mRNA-1345, an mRNA-Based Vaccine Against Respiratory Syncytial Virus in Adults. In: RSVVW’21 Abstract Booklet. Virtual: RSVVW’21: 6th ReSViNet Conference: November 10–12, 2021. Available from: https://www.resvinet.org/uploads/2/2/2/7/22271200/abstract_booklet_rsvvw21.pdf, 2021: 45. [Google Scholar]
- 78.ClinicalTrials.gov. Identifier: NCT05127434, A Study to Evaluate the Safety and Efficacy of mRNA-1345 Vaccine Targeting Respiratory Syncytial Virus (RSV) in Adults ≥60 Years of Age - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2021. Nov 19. https://clinicaltrials.gov/ct2/show/NCT05127434 (accessed April 12, 2022). [Google Scholar]
- 79.Samy N, Reichhardt D, Schmidt D, et al. Safety and immunogenicity of novel modified vaccinia Ankara-vectored RSV vaccine: A randomized phase I clinical trial. Vaccine 2020; 38: 2608–19. [DOI] [PubMed] [Google Scholar]
- 80.Weidenthaler H, Schultz S, Sanos S, et al. VT&T-77: Efficacy, Safety And Immunogenicity Of The Recombinant MVA-BN-RSV Vaccine Against Respiratory Syncytial Virus (RSV) Infection In A Human Challenge Trial (HCT) In Healthy Adult Participants. In: RSVVW’21 Abstract Booklet. Virtual: RSVVW’21: 6th ReSViNet Conference: November 10–12, 2021. Available from: https://www.resvinet.org/uploads/2/2/2/7/22271200/abstract_booklet_rsvvw21.pdf, 2021: 59. [Google Scholar]
- 81.Jordan E, Lawrence SJ, Meyer TPH, et al. Broad Antibody and Cellular Immune Response from a Phase 2 Clinical Trial with a Novel Multivalent Poxvirus-Based Respiratory Syncytial Virus Vaccine. J Infect Dis 2021; 223: 1062–72. [DOI] [PubMed] [Google Scholar]
- 82.ClinicalTrials.gov. Identifier NCT05238025, Phase 3 MVA-BN-RSV Vaccine Efficacy Trial. Bethesda Natl. Libr. Med. (US). 2022. Feb 14. https://clinicaltrials.gov/ct2/show/NCT05238025 (accessed April 12, 2022). [Google Scholar]
- 83.Krarup A, Truan D, Furmanova-Hollenstein P, et al. A highly stable prefusion RSV F vaccine derived from structural analysis of the fusion mechanism. Nat Commun 2015; 6: 8143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.van der Fits L, Bolder R, Heemskerk-van der Meer M, et al. Adenovector 26 encoded prefusion conformation stabilized RSV-F protein induces long-lasting Th1-biased immunity in neonatal mice. NPJ vaccines 2020; 5: 49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Williams K, Bastian AR, Feldman RA, et al. Phase 1 Safety and Immunogenicity Study of a Respiratory Syncytial Virus Vaccine With an Adenovirus 26 Vector Encoding Prefusion F (Ad26.RSV.preF) in Adults Aged ≥60 Years. J Infect Dis 2020; 222: 979–88. [DOI] [PubMed] [Google Scholar]
- 86.Sadoff J, De Paepe E, DeVincenzo J, et al. Prevention of Respiratory Syncytial Virus Infection in Healthy Adults by a Single Immunization of Ad26.RSV.preF in a Human Challenge Study. J Infect Dis 2021; Online ahe: jiab003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Falsey AR, Williams K, Gymnopoulou E, et al. Efficacy and Immunogenicity of an Ad26.RSV.preF-based Vaccine in the Prevention of RT-PCR-confirmed RSV-mediated Lower Respiratory Tract Disease in Adults Aged ≥65 Years: A Randomized, Placebo-controlled, Phase 2b Study. Open Forum Infect Dis 2021; 8: S812–S812. [Google Scholar]
- 88.Falsey AR, Williams K, Efi G, et al. VT&T-58: Safety and tolerability of an Ad26.RSV.preF-based vaccine: a randomized, double-blind, placebo-controlled, phase 2b study in adults aged >65 years. In: RSVVW’21 Abstract Booklet. Virtual: RSVVW’21: 6th ReSViNet Conference: November 10–12, 2021. Available from: https://www.resvinet.org/uploads/2/2/2/7/22271200/abstract_booklet_rsvvw21.pdf, 2021: 48. [Google Scholar]
- 89.Falsey AR, Williams K, Efi G, et al. VT&T-57: Efficacy, safety, and immunogenicity of AD26.RSV.preF-based vaccine in adults aged >65 years with or without addditional risk factors for RSV-mediated lower respiratory tract disease. In: RSVVW’21 Abstract Booklet. Virtual: RSVVW’21: 6th ReSViNet Conference: November 10–12, 2021. Available from: https://www.resvinet.org/uploads/2/2/2/7/22271200/abstract_booklet_rsvvw21.pdf, 2021: 47. [Google Scholar]
- 90.Sadoff J, De Paepe E, Haazen W, et al. Safety and Immunogenicity of the Ad26.RSV.preF Investigational Vaccine Coadministered With an Influenza Vaccine in Older Adults. J Infect Dis 2021; 223: 699–708. [DOI] [PubMed] [Google Scholar]
- 91.ClinicalTrails.gov. Identifier: NCT04908683, A Study of an Adenovirus Serotype 26 Pre-fusion Conformation-stabilized F Protein (Ad26. RSV. preF) Based Respiratory Syncytial Virus (RSV) Vaccine in the Prevention of Lower Respiratory Tract Disease in Adults Aged 60 Years and O. Bethesda Natl. Libr. Med. (US). 2021. Jun 1. https://clinicaltrials.gov/ct2/show/NCT04908683 (accessed April 12, 2022). [Google Scholar]
- 92.Snape M. Phase 1 safety and immunogenicity study of a respiratory syncytial virus vaccine with an adenovirus 26 vector encoding prefusion F (Ad26.RSV.preF) in adults aged ≥60 years. In: ISIRV RSV Webinar Series: RSV Vaccines and Therapies. Virtual: Available from: https://isirv.org/site/index.php/9-events/514-rsv-webinar-series, 2022. DOI: 10.1093/infdis/jiaa193. [DOI] [PubMed] [Google Scholar]
- 93.Klok FA, Pai M, Huisman MV, Makris M. Vaccine-induced immune thrombotic thrombocytopenia. Lancet Haematol 2022; 9: e73–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Brodsky FM. Monoclonal antibodies as magic bullets. Pharm Res 1988; 5: 1–9. [DOI] [PubMed] [Google Scholar]
- 95.Griffin MP, Yuan Y, Takas T, et al. Single-Dose Nirsevimab for Prevention of RSV in Preterm Infants. N Engl J Med 2020; 383: 415–25. [DOI] [PubMed] [Google Scholar]
- 96.Hammitt LL, Dagan R, Yuan Y, et al. Nirsevimab for Prevention of RSV in Healthy Late-Preterm and Term Infants. N Engl J Med 2022; 386: 837–46. [DOI] [PubMed] [Google Scholar]
- 97.Zhu Q, Lu B, Mctamney P, et al. Prevalence and Significance of Substitutions in the Fusion Protein of Respiratory Syncytial Virus Resulting in Neutralization Escape From Antibody MEDI8897. J Infect Dis 2018; 218: 572–80. [DOI] [PubMed] [Google Scholar]
- 98.Tang A, Chen Z, Cox KS, et al. A potent broadly neutralizing human RSV antibody targets conserved site IV of the fusion glycoprotein. Nat Commun 2019; 10: 4153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Maas BM, Lommerse J, Plock N, et al. VT&T-65: Forward and reverse translational approaches to predict efficacy of the neutralizing respiratory syncytial virus (RSV) antibody MK-1654. In: RSVVW’21 Abstract Booklet. Virtual: RSVVW’21: 6th ReSViNet Conference: November 10–12, 2021. Available from: https://www.resvinet.org/uploads/2/2/2/7/22271200/abstract_booklet_rsvvw21.pdf, 2021: 53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Maas BM, Lommerse J, Plock N, et al. Forward and reverse translational approaches to predict efficacy of neutralizing respiratory syncytial virus (RSV) antibody prophylaxis. EBioMedicine 2021; 73: 103651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.ClinicalTrials.gov. Identifier: NCT05118386, Safety, Tolerability, and Pharmacokinetics of RSV Monoclonal Antibody RSM01 in Healthy Adults - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2021. Nov 12. https://clinicaltrials.gov/ct2/show/NCT05118386 (accessed April 12, 2022). [Google Scholar]
- 102.Ananworanich J, Heaton PM. Bringing Preventive RSV Monoclonal Antibodies to Infants in Low- and Middle-Income Countries: Challenges and Opportunities. Vaccines 2021; 9: 961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Jones RGA, Martino A. Targeted localized use of therapeutic antibodies: a review of non-systemic, topical and oral applications. Crit Rev Biotechnol 2016; 36: 506–20. [DOI] [PubMed] [Google Scholar]
- 104.ClinicalTrials.gov. Identifier: NCT04540627, Exploratory Study to Estimate the Prophylactic Efficacy of Palivizumab in Healthy Adult Participants Inoculated With RSV - Full Text View - ClinicalTrials.gov. Bethesda Natl. Libr. Med. (US). 2020. Sep 7. https://clinicaltrials.gov/ct2/show/NCT04540627 (accessed April 12, 2022). [Google Scholar]
- 105.Mazur NI, Martinón-Torres F, Baraldi E, et al. Lower respiratory tract infection caused by respiratory syncytial virus: current management and new therapeutics. Lancet Respir Med 2015; 3: 888–900. [DOI] [PubMed] [Google Scholar]
- 106.DeVincenzo JP, El Saleeby CM, Bush AJ. Respiratory syncytial virus load predicts disease severity in previously healthy infants. J Infect Dis 2005; 191: 1861–8. [DOI] [PubMed] [Google Scholar]
- 107.Chu HY, Englund JA. Maternal immunization. Clin Infect Dis 2014; 59: 560–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Nunes MC, Cutland CL, Jones S, et al. Duration of infant protection against influenza illness conferred by maternal immunization secondary analysis of a randomized clinical trial. JAMA Pediatr 2016; 170: 840–7. [DOI] [PubMed] [Google Scholar]
- 109.Lee B. Update on rotavirus vaccine underperformance in low- to middle-income countries and next-generation vaccines. Hum Vaccin Immunother 2021; 17: 1787–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Ahonkhai V, Martins SF, Portet A, Lumpkin M, Hartman D. Speeding Access to Vaccines and Medicines in Low- and Middle-Income Countries: A Case for Change and a Framework for Optimized Product Market Authorization. PLoS One 2016; 11: e0166515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Li X, Willem L, Antillon M, Bilcke J, Jit M, Beutels P. Health and economic burden of respiratory syncytial virus (RSV) disease and the cost-effectiveness of potential interventions against RSV among children under 5 years in 72 Gavi-eligible countries. BMC Med 2020; 18: 82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Scheltema NM, Gentile A, Lucion F, et al. Global respiratory syncytial virusassociated mortality in young children (RSV GOLD): a retrospective case series. Lancet Glob Heal 2017; 5: e984–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Supplement 3: Community Mortality Burden Associated with Respiratory Syncytial Virus in Low- and Middle-Income Countries. Clin Infect Dis 2021; 73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Zhang S, Akmar LZ, Bailey F, et al. Cost of Respiratory Syncytial Virus-Associated Acute Lower Respiratory Infection Management in Young Children at the Regional and Global Level: A Systematic Review and Meta-Analysis. J Infect Dis 2020; 222: S680–7. [DOI] [PubMed] [Google Scholar]
- 115.Joyce MG, Mclellan JS, Stewart-Jones GBE, Sastry M, Yang Y, Graham BS, Kwong PD. Crystal Structure of Prefusion-stabilized RSV F Variant DS-Cav1 at pH 5.5: pdb4MMU. 2013. [Google Scholar]
- 116.McLellan JS, Chen M, Joyce MG, et al. Structure-based design of a fusion glycoprotein vaccine for respiratory syncytial virus. Science (80- ) 2013; 342: 592–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.McLellan JS, Yang Y, Graham BS, Kwong PD. Structure of respiratory syncytial virus fusion glycoprotein in the postfusion conformation reveals preservation of neutralizing epitopes. J Virol 2011; 85: 7788–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.McLellan JS, Yongping Y, Graham BS, Kwong PD. Structure of the RSV F protein in the post-fusion conformation: pdb3RRT. 2015. [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.