Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 3588 word(s) 3588 2020-12-09 06:15:18 |
2 format correction -170 word(s) 3418 2021-01-04 01:58:27 | |
3 format correction -170 word(s) 3418 2021-01-04 02:06:41 | |
4 Spelling and grammar corrections Meta information modification 3418 2021-01-04 14:48:25 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Dondi, A.; Biagi, C. Respiratory Syncytial Virus Vaccines. Encyclopedia. Available online: https://encyclopedia.pub/entry/5969 (accessed on 27 July 2024).
Dondi A, Biagi C. Respiratory Syncytial Virus Vaccines. Encyclopedia. Available at: https://encyclopedia.pub/entry/5969. Accessed July 27, 2024.
Dondi, Arianna, Carlotta Biagi. "Respiratory Syncytial Virus Vaccines" Encyclopedia, https://encyclopedia.pub/entry/5969 (accessed July 27, 2024).
Dondi, A., & Biagi, C. (2021, January 03). Respiratory Syncytial Virus Vaccines. In Encyclopedia. https://encyclopedia.pub/entry/5969
Dondi, Arianna and Carlotta Biagi. "Respiratory Syncytial Virus Vaccines." Encyclopedia. Web. 03 January, 2021.
Respiratory Syncytial Virus Vaccines
Edit

Respiratory Syncytial Virus (RSV) is the main cause of acute respiratory tract infections in infants and it also induces significant disease in the elderly, with a potentially severe clinical course and a large number of deaths in developing countries and of intensive care hospitalizations worldwide. To date, prevention strategies against RSV infection are based on hygienic measures and passive immunization with humanized monoclonal antibodies limited to selected high-risk children due to their high costs. The development of a safe and effective vaccine is a global health need and an important objective of research in this field. A growing number of RSV vaccine candidates in different formats (particle-based vaccines, vector-based vaccines, subunit vaccines, and live-attenuated vaccines) are being developed and are now at different stages. While waiting for commercially available safe and effective vaccines, immune prophylaxis in selected groups of high-risk populations is still mandatory.

RSV vaccine prevention

1. Epidemiology

Respiratory syncytial virus (RSV) is often responsible for severe seasonal respiratory disease in infants and elderly people, and it causes a great burden on health systems worldwide. RSV clinical presentation in children involves respiratory infections ranging from mild upper to severe lower respiratory tract infections (LRTI), including pneumonia or bronchiolitis [1][2][3][4]; in fact, it was estimated that RSV may cause up to 22% of all severe acute LRTI in young children. A recent systematic review [3] reported that worldwide in 2015, RSV caused 33.1 million episodes of LRTI, resulting in nearly 3.2 million hospitalizations and 59,600 in-hospital deaths in children younger than 5 years. In infants younger than 6 months, RSV caused 1.4 million hospital admissions and 27,300 in-hospital deaths [3]. The estimated overall RSV mortality in 2015 was 118,200 deaths in infants younger than 5 years. Moreover, RSV is responsible for pulmonary morbidity and hospitalizations also in the elderly and high-risk adults [5], causing more than 17,000 deaths for underlying pneumonia and circulatory complications [6]. Risk factors for severe RSV infections in pediatric populations that may require hospitalization and Intensive Care Unit admission include prematurity, chronic lung disease of prematurity or bronchopulmonary dysplasia, hemodynamically significant congenital heart disease, congenital or acquired immunodeficiency, and severe neuromuscular disease.

2. Molecular Characteristics of Respiratory Syncytial Virus

RSV is an enveloped, non-segmented, negative-sense, single-stranded RNA virus of the Orthopneumovirus genus and Pneumoviridae family. The envelope of RSV includes four proteins associated with the lipid double layer: the matrix (M) protein, the small hydrophobic (SH) protein, and the two glycosylated surface proteins: F (Fusion) and G (attachment glycoprotein). The proteins, which are directly involved in infectivity and development of the respiratory disease, are F and G: the G protein determines the attachment of the virus to host epithelial cells, while the F protein is involved in the entry of the virus, through the fusion of viral and cellular membranes, as well as the subsequent insertion of the viral RNA into the host cell; the F protein is also responsible for fusion of infected cells with adjacent cells, resulting in the formation of the characteristic syncytia [7]. Moreover, both F and G proteins induce the neutralizing antibody immune response by the host [8]. Three types of epitopes have been identified in the G protein: (1) conserved epitopes, detectable in all viral strains; (2) group-specific epitopes, expressed only by the same antigenic group; and (3) strain-specific epitopes, which are present only in specific strains of the same antigenic group and expressed in the C-terminal hypervariable region of the G protein ectodomain [9]. The F glycoprotein is derived from an inactive precursor containing three hydrophobic peptides: (1) the N-terminal signal peptide, mediating translocation of the nascent polypeptide into the lumen of the endoplasmic reticulum; (2) the transmembrane region near the C-terminus, which links the F protein to the cell and viral membranes; and (3) the so-called fusion peptide, which inserts into the target cell membrane and determines the fusion process. The binding of the prefusion F protein to the cell surface is followed by its activation and structural changes, which determine the fusion of the membrane of the virus with the airway epithelial cells of the host and lead to the formation of syncytia. There are two major subgroups of RSV, A and B, usually both detectable during the same epidemic season, even if temporal and geographic clustering may occur [10][11]. RSV infections with Group A have a higher incidence and higher transmissibility than Group B [12]. The antigenic variability between the two groups is determined by differences in the G glycoprotein (35% homology between the G glycoprotein of strains A and B) [13]. For this reason, many antibodies targeting the G protein may be subtype-specific, while antibodies against the F protein have a neutralizing effect both against RSV A and B. Vaccine candidates against RSV use different antigenic targets [14]. Most vaccines in clinical trials use the F protein, because it is highly conserved and facilitates viral fusion with host cells. Current vaccine candidates use pre-F and post-F as vaccine antigens. Site zero (∅) of the pre-F protein has been recently discovered and is one of the major antigenic targets [15]. Other less frequent vaccine antigens, used alone or in combination with others, include the RSV envelope associated glycoprotein G and the SH protein, as well as the internal proteins: nucleocapsid (N), M and M2-1. Besides the F protein, the G protein is the only other target for neutralizing antibodies on the viral surface.

3. Prophylaxis measures

Prevention of RSV infection is based on primary and secondary prophylaxis. Preventive measures are critical because an etiologic treatment against RSV is not available. Primary prophylaxis is essentially based on hygienic measures aimed to prevent the diffusion of respiratory virus infections (handwashing, use of face masks in case of symptoms, disinfection of objects and surfaces) [16]. Secondary prophylaxis is based on the administration of monoclonal antibodies (mAbs) to high-risk patients during the epidemic season. The only commercially available mAb is palivizumab, while more recent ones are being studied in ongoing clinical trials [17][18][19]. No vaccine is currently available for active immunization against RSV, even if several are a matter of ongoing clinical trials [14]. One of the hardest challenges in the development of a safe and effective vaccine is enhanced respiratory disease (ERD), a side-effect that occurred in the 1960s subsequently to the administration of formalin-inactivated RSV vaccines [20][21]. Moreover, it is still a matter of concern if a vaccine determines absolute protection against RSV infections; encouraging data demonstrate that cell-mediated immunity, mucosal IgA, and serum neutralizing antibodies are inversely related to disease severity [14].

4. Target Populations

Considering the epidemiology of RSV, preventive measures are particularly useful for target populations. Currently, passive immunization with mAbs is reserved for high-risk infants [17], as detailed in national and international guidelines. Risk factors considered in the guidelines for prophylaxis include severe prematurity, congenital heart diseases, bronchopulmonary dysplasia, severe respiratory, and neuromuscular diseases. The clinical trials of vaccines against RSV are run on pediatric populations, old adults, or pregnant women with the aim to prevent RSV infections in high-risk populations (Table 1). RSV was also observed to affect long-term respiratory morbidity since young infants with RSV LRTI subsequently had an increased risk for asthma and recurrent wheezing [22]. For this reason, the development of a safe and effective vaccine may be useful to prevent the onset of respiratory morbidity if extended to cohorts of naive infants during the first months of life. Since severe RSV infections often involve newborns and very young infants, the strategy of vaccination during pregnancy may be useful to determine an effective immunity at early ages when active immunization is not feasible, as maternal antibodies are transferred efficiently through the placenta. Vaccines in clinical development are grouped into four categories: particle-based, vector-based, subunit, and live-attenuated or chimeric vaccines.

Table 1. Summary of the RSV vaccines in clinical development by the target population. Only the most advanced trial for a specific target group is reported. RSV: respiratory syncytial virus; NIAID: National Institute of Allergy and Infectious Diseases.

Target Population

Vaccine Name

Sponsor

Vaccine Type

Clinical Trial Phase

Pregnant women

ResVax

Novavax

Particle-based

III

RSV F DS-Cav1

VRC-RSVRGP084-00-VP

NIAID

Subunit

I

RSV vaccine

Pfizer

Subunit

II

GSK3888550A

GlaxoSmithKline

Subunit

II

Children

RSV F nanoparticle

Novavax

Particle-based

I

SynGEM

Mucosis

Particle-based

II

Ad26.RSV.preF

Janssen

Vector-based

I/IIa

ChAd155-RSV

GlaxoSmithKline

Vector-based

II

MEDI-534

MedImmune (AstraZeneca)

Vector-based

I

SeVRSV

NIAID

Vector-based

I

RSV MEDI ΔM2-2

NIAID

Live-attenuated

I/II

RSV LID ΔM2-2

NIAID

Live-attenuated

I

RSV cps2

NIAID

Live-attenuated

I

RSV LID cp ΔM2-2

NIAID

Live-attenuated

I

RSV LID ΔM2-2 1030s

NIAID

Live-attenuated

I

RSV D46/NS2/N/ΔM2-2-HindIII

NIAID

Live-attenuated

I

RSV ΔNS2 Δ1313 I1314L

NIAID

Live-attenuated

I

RSV 6120/ΔNS1;

RSV 6120/F1/G2/ΔNS1

NIAID

Live-attenuated

I

RSV ΔNS2/Δ1313/I1314L;

RSV 6120/ΔNS2/1030s;

RSV 276

NIAID

Live-attenuated

I/II

RSV 6120/∆NS2/1030s

NIAID

Live-attenuated

I

RSV ΔNS2/Δ1313/I1314L;

RSV 276

NIAID

Live-attenuated

I

RSV D46 cpΔM2-2

NIAID

Live-attenuated

I

MEDI-559

MedImmune (AstraZeneca)

Live-attenuated

I/II

MV-012-968

Meissa Vaccines

Live-attenuated

I

rBCG-N-hRSV

Pontificia Universidad Catolica de Chile

Chimeric

I

Elderly

RSV F nanoparticle

Novavax

Particle-based

III

SynGEM

Mucosis

Particle-based

II

MVA-BN RSV

Bavarian Nordic

Vector-based

II

VXA-RSVf oral

Vaxart

Vector-based

I

Ad26.RSV.preF

Janssen

Vector-based

II

PanAd3-RSV and MVA-RSV

ReiThera

Vector-based

I

DPX-RSV

Dalhousie University

Subunit

I

RSV vaccine

Pfizer

Subunit

II

GSK3844766A

GlaxoSmithKline

Subunit

I

5. Particle-Based Vaccines

Particle-based vaccines are synthesized by self-assembling nanoscopic particles that expose multiple copies of a selected antigen on their surface and mimic the native virions: thanks to the high copy number of the selected antigen and the immune-boosting properties of the particle matrix, these vaccines elicit strong humoral and cellular immune responses [23]. Moreover, the lack of the viral genome required for replication makes them safe. To date, two nanoparticle-based RSV vaccines have been tested in clinical trials. The RSV F nanoparticle vaccine, developed by Novavax, is being evaluated in women of childbearing age, pre-school children (2–6 years old) and the elderly (≥ 60 years old). [24] In Phase I clinical trials, it has proven to be well-tolerated and highly immunogenic in all the target populations. [25][26][27][28] ResVax is a maternal RSV F nanoparticle vaccine with an aluminium phosphate adjuvant which is being developed to protect infants from RSV disease via maternal immunization. ResVax has been shown to be safe in a Phase II trial enrolling 50 healthy third-trimester pregnant women and to elicit RSV neutralizing antibodies and palivizumab-competing antibodies that are efficiently transferred to the infants. [29] This maternal vaccine has been the object a Phase III multi-country, randomized, placebo-controlled trial evaluating its efficacy against RSV-LRTI in infants born in the RSV season to 4363 women who received the vaccine or placebo between 28 and 36 weeks of pregnancy. [30] ResVax failed to meet the primary outcome of prevention of medically significant LRTI. However, it showed 44% efficacy in reducing RSV-LRTI hospitalization, 39.4% efficacy in reducing RSV-specific medically significant LRTI and 58.8% efficacy in reducing RSV-related severe hypoxemia in young infants (< 3 months of age). In addition, pneumonia was 49.4% less common in infants of the vaccinees than the placebo recipients. An additional Phase III clinical trial to confirm the efficacy of ResVax will thus be conducted by Novavax. [31] Regarding older adults, the non-adjuvanted RSV F nanoparticle vaccine failed to demonstrate efficacy in a 2015 Phase III clinical trial which enrolled 11,850 adults ≥60 years of age. [32] Novavax conducted a second Phase II clinical trial in the elderly to evaluate the safety and immunogenicity of single- or two-dose regimens of the RSV F vaccine with and without adjuvants (aluminium phosphate or Matrix-M1). The study showed that both adjuvants increased the magnitude, duration and quality of the immune response versus the non-adjuvanted RSV F vaccine. [33] These data support the inclusion of the RSV F vaccine adjuvant formulations in future elderly trials.

The other nanoparticle-based RSV vaccine that has been tested in clinical trials is SynGEM, developed by Mucosis. SynGEM is a mucosal vaccine containing the RSV F protein in the prefusion conformation bound to a bacterium-like particle (BLP) derived from Lactococcus lactis. BLP has the role to present the vaccine antigen in a more natural conformation and to boost the immune system of the virus. [34] The safety and tolerability of SynGEM have been evaluated in a Phase I trial and it was generally well-tolerated and induced persistent antibody responses in healthy adults. [35] However, no detectable RSV neutralizing antibodies were found, despite preclinical data that had shown protection. [36] Future studies are needed to optimize the immunogenicity of SynGEM.

Particle-based vaccines are promising both for young infants through the immunization of pregnant mothers and for the elderly. ResVax has already reached Phase III clinical trials for pregnant women, and hopefully new trials will soon allow its marketing approval. On the other hand, for elderly people, the use of RSV F vaccine adjuvant formulations in future trials may increase the magnitude, duration and quality of the induced immune response.

6. Vector-Based Vaccines

Vector-based vaccines use a carrier vector to deliver RSV antigens and induce an immune response against RSV components exploiting the adjuvant effect of the vector. Due to the chimeric nature of the vectors, there is no risk of reversion to wild-type RSV and of ERD. However, the presence of pre-existing anti-vector immunity or its potential development may challenge the clinical use of these vaccines. To date, eight RSV vector-based vaccines have been tested in clinical trials. The MVA-BN-RSV vaccine has been developed by Bavarian Nordic and it is based on a non-replicating modified vaccinia Ankara (MVA) virus; it displays the RSV surface proteins F and G (for both A and B subtypes) and two internal RSV proteins N and M2. It has passed phase I and II clinical trials, [37][38] and a phase III trial on elderly people is planned for 2021. ReiThera developed two novel, recombinant, viral-vectored vaccine candidates for RSV, PanAd3-RSV and MVA-RSV. Both vaccines use RSV F, N and M2 proteins as antigens, delivered, respectively, by a replication-defective simian Adenovirus (PanAd3) and MVA vector. PanAd3-RSV and MVA-RSV were well tolerated and immunogenic in healthy adults [39] and in the elderly [40], but their production has been halted since 2015. Three other virus-vectored vaccines use an Adenovirus vector to deliver RSV antigens. VXA-RSV-f, developed by Vaxart, is a molecularly adjuvanted Adenovirus serotype 5 based RSV vaccine encoding the F protein and is being developed for older adults, even if the results of an already concluded phase I trial have not been published yet. Janssen developed Ad26.RSV.Pre-F vaccine for the elderly and children, based on the human Adenovirus strain 26 vector expressing the F protein stabilized in the pre-fusion conformation. [41] A Phase II study evaluated the co-administration of a seasonal influenza vaccine with the Ad26.RSV.preF vaccine in 180 healthy elderly people, showing an acceptable safety profile and no evidence of interference in the immune response. [42] Results of two Phase I/IIa studies [43][44] evaluating the safety and tolerability of Ad26.RSV.preF in children should be published soon. Another Adenovirus-based vaccine, ChAd155-RSV, intended for children, is currently in clinical development. [45] Two vector based-vaccines use the parainfluenza virus (PIV) to display RSV antigens, with the aim to induce immunity against both viruses: MEDI-534 and the recombinant Sendai virus vectored RSV (SeVRSV). However, these vaccines are still under evaluation in Phase I trials. [46][47]

Vector-based vaccines are potentially good candidates for the pediatric population because there is no risk of reversion to wild-type RSV and of ERD. These vaccines are still in the initial stage concerning their evaluation. The eight vaccines that have been tested in clinical trials are based on MVA, Adenovirus, bovine PIV or Sendai Virus vectors. The MVA-BN-RSV vaccine is the only one that has passed Phase 2 clinical trial, and a Phase 3 trial in the elderly has been planned for initiation in 2021.

7. Subunit Vaccines

Subunit vaccines are created with RSV protein fragments. They are poorly immunogenic due to their non-replicating nature and their limited components; therefore, booster administration and adjuvants are often necessary to make them effective. [48] This type of vaccine primarily induces a CD4 T cell response, with a higher risk for vaccine-ERD in seronegative infants, [49][50] so subunit vaccines are under development only for pregnant women and older people that have already had a previous RSV infection and that are not a risk of developing ERD. [51] VRC-RSVRGP084-00-VP, a prefusion F-based RSV vaccine, has been tested in healthy adults with or without an aluminum adjuvant, with the aim of monitoring safety, tolerability, and immunogenicity. [52] Preliminary results that were published in 2019 are promising, [53] but the Phase I trial has recently been concluded (April 2020), and the results are expected to be published soon. Trials testing another subunit vaccine candidate (with and without adjuvant) on healthy adults of various age groups should be completed at the end of 2020 and in 2021. [54][55][56] Meanwhile, the Phase IIb trial, concluded in December 2019, recruited a group of healthy non-pregnant women (18–49 years old) to which this vaccine candidate was given together with diphtheria/tetanus/pertussis vaccine. [57] DepoVax (DPX)-RSV is a candidate based on the ectodomain of the SH protein, presented with a novel lipid-based formulation that ensures a prolonged exposure of the antigen; Phase I results are promising, [58] but Phase II has not yet started. BARS13 is a candidate based on the G glycoprotein: the Phase I study has involved healthy adult volunteers, but the results have not yet been published. [59] Two more candidates are under evaluation: GSK3888550A, with pregnant women as the target population, and GSK3844766A, designed for older adults. [60]

Subunit vaccines can be considered a safe choice: they do not contain live viruses that could return to a virulent state and they cannot induce an exceeding immune response. However, owing to concerns of ERD associated with protein-based vaccines, they are potentially good candidates only for pregnant women and the elderly. Observing the concluded trials and the studies in progress, induction of protective immunity, and obtaining F protein conformation stability remain the major unsolved problems for creating an effective subunit vaccine.

8. Live-attenuated and chimeric vaccines

Live-attenuated RSV vaccines (RSV-LAVs) are produced with versions of RSV that are able to replicate but have been modified to discourage severe disease. ERD has not been observed with RSV-LAVs or replicating vaccine vectors: for this reason, these candidates can be considered safe for naive-RSV infants. [49][61] RSV-LAVs have also other benefits: the ability to replicate in the respiratory tract despite the presence of maternal antibodies, the capacity to promote both a humoral and cellular immune response, and the possibility to be administered as nasal drops, which are less invasive and better tolerated in children. [62][63] Most vaccines are in Phase I and no candidates of this type have progressed beyond Phase II clinical trials. However, RSV-LAVs represent promising candidates worthy of further investigation.

The only chimeric vaccine candidate is rBCG-N-hRSV. [64] It consists of the bacillus Calmette-Guerin (BCG) vaccine expressing the nucleoprotein (N) of RSV. It is delivered via a BCG strain and induces a Th1 and a humoral response. It is the only LAV that combines protection against two respiratory pathogens, Mycobacterium tuberculosis and RSV. Furthermore, it could be safe for administration to newborn babies. A Phase I clinical trial on adults has been conducted, even if the results have not been reported yet. [65]

9. Conclusions

The development of an effective vaccine to protect high-risk groups from severe RSV infections is of critical importance but still challenging. Different antigens and vaccine formats should be considered for different target populations (children, the elderly, and pregnant women). To date, the formats that are being evaluated are particle-based vaccines, vector-based vaccines, subunit vaccines, and LAVs. Currently, the only vaccine that has reached Phase III clinical trials is a maternal RSV F nanoparticle vaccine, which showed efficacy in reducing hospitalization and RSV-LRTI in young infants, but did not meet the desired primary outcome, so future trials are needed to confirm its efficacy. The same vaccine with adjuvants was safe and immunogenic in the elderly, and future clinical trials will evaluate its efficacy. Only one vector-based vaccine has passed a Phase II clinical trial, and a Phase III trial in the elderly has been planned for initiation in 2021. Subunit vaccines are potentially good candidates for pregnant women and the elderly; induction of protective immunity and obtaining F protein conformation stability remain the major unsolved problems for creating an effective subunit vaccine. Most clinical trials for RSV-LAVs are only in Phase I, but these vaccines represent promising candidates worthy of further investigation as they are able to generate a robust immune response and, thanks to their safety and nature, can be administered also to infants. While waiting for commercially available safe and effective vaccines, immune prophylaxis in selected groups of high-risk populations is still mandatory. In young infants, combining the use of passive immunization via maternal vaccination or mAbs, followed by paediatric active immunization, may be effective to prevent severe RSV infection. Research on this topic is still of utmost importance.

References

  1. Stein, R.T.; Bont, L.J.; Zar, H.; Polack, F.P.; Park, C.; Claxton, A.; Borok, G.; Butylkova, Y.; Wegzyn, C. Respiratory syncytial virus hospitalization and mortality: systematic review and meta‐analysis. Pediatr. Pulmonol. 2017, 52, 556–569, doi:10.1002/ppul.23570.
  2. Iwane, M.K.; Chaves, S.S.; Szilagyi, P.G.; Edwards, K.M.; Hall, C.B.; Staat, M.A.; Brown, C.J.; Griffin, M.R.; Weinberg, G.A.; Poehling, K.A.; et al. Disparities between black and white children in hospitalizations associated with acute respiratory illness and laboratory-confirmed influenza and respiratory syncytial virus in 3 US Counties—2002–2009. Am. J. Epidemiol. 2013, 177, 656–665, doi:10.1093/aje/kws299.
  3. Nair, H.; Nokes, D.J.; Gessner, B.D.; Dherani, M.; Madhi, S.A.; Singleton, R.J.; O’Brien, K.L.; Roca, A.; Wright, P.F.; Bruce, N.; et al. Global burden of acute lower respiratory infections due to respiratory syncytial virus in young children: A systematic review and meta-analysis. Lancet Lond. Engl. 2010, 375, 1545–1555, doi:10.1016/S0140-6736(10)60206-1.
  4. Shi, T.; McAllister, D.A.; O’Brien, K.L.; Simoes, E.A.F.; Madhi, S.A.; Gessner, B.D.; Polack, F.P.; Balsells, E.; Acacio, S.; Aguayo, C.; 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, 946–958, doi:10.1016/S0140-6736(17)30938-8.
  5. Falsey, A.R.; Hennessey, P.A.; Formica, M.A.; Cox, C.; Walsh, E.E. Respiratory Syncytial Virus Infection in Elderly and High-Risk Adults. N. Engl. J. Med.2005, 352, 1749–1759, doi:10.1056/NEJMoa043951.
  6. Thompson, W.W.; Shay, D.K.; Weintraub, E.; Brammer, L.; Cox, N.; Anderson, L.J.; Fukuda, K. Mortality associated with influenza and respiratory syncytial virus in the United States. JAMA 2003, 289, 179–186, doi:10.1001/jama.289.2.179.
  7. Vandini, S.; Biagi, C.; Lanari, M. Respiratory syncytial virus: the influence of serotype and genotype variability on clinical course of infection. Int. J. Mol. Sci. 2017, 18, 1717, doi:10.3390/ijms18081717.
  8. Melero, J.A.; Mas, V.; McLellan, J.S. Structural, antigenic and immunogenic features of respiratory syncytial virus glycoproteins relevant for vaccine development. Vaccine 2017, 35, 461–468, doi:10.1016/j.vaccine.2016.09.045.
  9. Martinez, I.; Valdes, O.; Delfraro, A.; Arbiza, J.; Russi, J.; Melero, J.A. Evolutionary pattern of the G glycoprotein of human respiratory syncytial viruses from antigenic group B: The use of alternative termination codons and lineage diversification. J. Gen. Virol. 1999, 80, 125–130, doi:10.1099/0022-1317-80-1-125.
  10. Hendry, R.M.; Talis, A.L.; Godfrey, E.; Anderson, L.J.; Fernie, B.F.; McIntosh, K. Concurrent circulation of antigenically distinct strains of respiratory syncytial virus during community outbreaks. J. Infect. Dis. 1986, 153, 291–297, doi:10.1093/infdis/153.2.291.
  11. Venter, M.; Madhi, S.A.; Tiemessen, C.T.; Schoub, B.D. Genetic diversity and molecular epidemiology of respiratory syncytial virus over four consecutive seasons in South Africa : Identification of New Subgroup A and B Genotypes. J. Gen. Virol. 2001, 82, 2117–2124.
  12. White, L.J.; Waris, M.; Cane, P.A.; Nokes, D.J.; Medley, G.F. The transmission dynamics of groups A and B Human Respiratory Syncytial Virus (HRSV) in England & Wales and Finland: Seasonality and cross-protection. Epidemiol. Infect. 2005, 133, 279–289.
  13. Johnson, T.R.; Graham, B.S. Contribution of respiratory syncytial virus g antigenicity to vaccine-enhanced illness and the implications for severe disease during primary respiratory syncytial virus infection. Pediatr. Infect. Dis. J. 2004, 23, S46–S57, doi:10.1097/01.inf.0000108192.94692.d2.
  14. Mazur, N.I.; Higgins, D.; Nunes, M.C.; Melero, J.A.; Langedijk, A.C.; Horsley, N.; Buchholz, U.J.; Openshaw, P.J.; McLellan, J.S.; Englund, J.A.; 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–e311, doi:10.1016/S1473-3099(18)30292-5.
  15. Hashimoto, K.; Hosoya, M. Neutralizing epitopes of RSV and Palivizumab resistance in Japan. FUKUSHIMA J. Med. Sci. 2017, 63, 127–134, doi:10.5387/fms.2017-09.
  16. RSV|Prevention|Respiratory Syncytial Virus|CDC. Available online: https://www.cdc.gov/rsv/about/prevention.html (accessed on 1 October 2020).
  17. Resch, B. Product review on the monoclonal antibody Palivizumab for prevention of respiratory syncytial virus infection. Hum. Vaccines Immunother. 2017, 13, 2138–2149, doi:10.1080/21645515.2017.1337614.
  18. Mac, S.; Sumner, A.; Duchesne-Belanger, S.; Stirling, R.; Tunis, M.; Sander, B. Cost-Effectiveness of Palivizumab for respiratory syncytial virus: A systematic review. Pediatrics 2019, 143, doi:10.1542/peds.2018-4064.
  19. Griffin, M.P.; Yuan, Y.; Takas, T.; Domachowske, J.B.; Madhi, S.A.; Manzoni, P.; Simões, E.A.F.; Esser, M.T.; Khan, A.A.; Dubovsky, F.; et al. Nirsevimab study group. Single-Dose Nirsevimab for prevention of RSV in preterm infants. N. Engl. J. Med. 2020, 383, 415–425, doi:10.1056/NEJMoa1913556.
  20. Kim, H.W.; Canchola, J.G.; Brandt, C.D.; Pyles, G.; Chanock, R.M.; Jensen, K.; Parrott, R.H. Respiratory Syncytial Virus disease in infants despite prior administrationof antigenic inactivated vaccine 12. Am. J. Epidemiol. 1969, 89, 422–434, doi:10.1093/oxfordjournals.aje.a120955.
  21. Kapikian, A.Z.; Mitchell, R.H.; Chanock, R.M.; Shvedoff, R.A.; Stewart, C.E. An epidemiologic study of altered clinical reactivity to Respiratory Syncytial (RS) Virus infection in childern previously vaccinated with an inactivated RS Virus vaccine. Am. J. Epidemiol. 1969, 89, 405–421, doi:10.1093/oxfordjournals.aje.a120954.
  22. Blanken, M.O.; Rovers, M.M.; Molenaar, J.M.; Winkler-Seinstra, P.L.; Meijer, A.; Kimpen, J.L.L.; Bont, L. Dutch RSV neonatal network. respiratory Syncytial Virus and recurrent wheeze in healthy preterm infants. N. Engl. J. Med. 2013, 368, 1791–1799, doi:10.1056/NEJMoa1211917.
  23. Recombinant Nanoparticle Rsv F Vaccine for Respiratory Syncytial Virus. . WO2013049342A1. Retrieved 2020-12-11
  24. Hotcherl Jeong; Baik L Seong; Exploiting virus-like particles as innovative vaccines against emerging viral infections. Journal of Microbiology 2017, 55, 220-230, 10.1007/s12275-017-7058-3.
  25. Greg Glenn; Louis F. Fries; D. Nigel Thomas; Gale Smith; Eloi Kpamegan; Hanxin Lu; David C. Flyer; Dewal Jani; Somia P. Hickman; Pedro A. Piedra; et al. A Randomized, Blinded, Controlled, Dose-Ranging Study of a Respiratory Syncytial Virus Recombinant Fusion (F) Nanoparticle Vaccine in Healthy Women of Childbearing Age. The Journal of Infectious Diseases 2015, 213, 411-422, 10.1093/infdis/jiv406.
  26. Allison August; Greg Glenn; Eloi Kpamegan; Somia P. Hickman; Dewal Jani; Hanxin Lu; D. Nigel Thomas; Judy Wen; Pedro A Piedra; Louis F. Fries; et al. A Phase 2 randomized, observer-blind, placebo-controlled, dose-ranging trial of aluminum-adjuvanted respiratory syncytial virus F particle vaccine formulations in healthy women of childbearing age. Vaccine 2017, 35, 3749-3759, 10.1016/j.vaccine.2017.05.045.
  27. Louis Fries; Vivek Shinde; Jeffrey J. Stoddard; D. Nigel Thomas; Eloi Kpamegan; Hanxin Lu; Gale Smith; Somia P. Hickman; Pedro Piedra; Gregory M. Glenn; et al. Immunogenicity and safety of a respiratory syncytial virus fusion protein (RSV F) nanoparticle vaccine in older adults. Immunity & Ageing 2017, 14, 1-14, 10.1186/s12979-017-0090-7.
  28. Novavax Announces Positive Top-Line Data From RSV F Vaccine Phase 1 Clinical Trial in Pediatrics | NovavaxInc. . Novavax. Retrieved 2020-12-11
  29. Flor M Muňoz; Geeta K Swamy; Somia P Hickman; Sapeckshita Agrawal; Pedro A Piedra; Gregory M Glenn; Nita Patel; Allison M August; Iksung Cho; Louis Fries; et al. Safety and Immunogenicity of a Respiratory Syncytial Virus Fusion (F) Protein Nanoparticle Vaccine in Healthy Third-Trimester Pregnant Women and Their Infants. The Journal of Infectious Diseases 2019, 220, 1802-1815, 10.1093/infdis/jiz390.
  30. Shabir A. Madhi; Fernando P. Polack; Pedro A. Piedra; Flor M. Munoz; Adrian A. Trenholme; Eric A.F. Simões; Geeta K. Swamy; Sapeckshita Agrawal; Khatija Ahmed; Allison August; et al.Abdullah H. BaquiAnna CalvertJanice ChenIksung ChoMark F. CottonClare L. CutlandJanet A. EnglundAmy FixBernard GonikLaura HammittPaul T. HeathJoanne N. De JesusChristine E. JonesAsma KhalilDavid W. KimberlinRomina LibsterConrado J. LlapurMarilla LuceroGonzalo Pérez MarcHelen S. MarshallMasebole S. MasenyaFederico Martinón-TorresJennifer K. MeeceTerry M. NolanAyman OsmanKirsten P. PerrettJoyce S. PlestedPeter C. RichmondMatthew D. SnapeJulie H. ShakibVivek ShindeTanya StoneyD. Nigel ThomasAlan T. TitaMichael W. VarnerManu VatishKeith VrbickyJudy WenKhalequ ZamanHeather J. ZarGregory M. GlennLouis F. Fries Respiratory Syncytial Virus Vaccination during Pregnancy and Effects in Infants. New England Journal of Medicine 2020, 383, 426-439, 10.1056/nejmoa1908380.
  31. Novavax Provides Updates on the Global Pathways to Licensure for ResVaxTM | Novavax Inc. . Novavax. Retrieved 2020-12-11
  32. Novavax Announces Topline RSV F Vaccine Data from Two Clinical Trials in Older Adults | Novavax Inc. . Novavax. Retrieved 2020-12-11
  33. Novavax Announces Positive Topline Data from Phase 2 Older Adult Trial and Provides Path Forward for RSV F Vaccine Programs | Novavax Inc . Novavax. Retrieved 2020-12-11
  34. Natalija Van Braeckel-Budimir; Bert Jan Haijema; Kees Leenhouts; Bacterium-Like Particles for Efficient Immune Stimulation of Existing Vaccines and New Subunit Vaccines in Mucosal Applications. Frontiers in Immunology 2013, 4, 282, 10.3389/fimmu.2013.00282.
  35. Stephanie Ascough; Iris Vlachantoni; Mohini Kalyan; Bert-Jan Haijema; Sanna Wallin-Weber; Margriet Dijkstra-Tiekstra; Muhammad S. Ahmed; Maarten Van Roosmalen; Roberto Grimaldi; Qibo Zhang; et al.Kees LeenhoutsPeter J. OpenshawChristopher Chiu Local and Systemic Immunity against Respiratory Syncytial Virus Induced by a Novel Intranasal Vaccine. A Randomized, Double-Blind, Placebo-controlled Clinical Trial. American Journal of Respiratory and Critical Care Medicine 2019, 200, 481-492, 10.1164/rccm.201810-1921oc.
  36. Alan Rigter; Ivy Widjaja; Hanneke Versantvoort; Frank E. J. Coenjaerts; Maarten Van Roosmalen; Kees Leenhouts; Peter J. M. Rottier; Bert Jan Haijema; Cornelis A. M. De Haan; A Protective and Safe Intranasal RSV Vaccine Based on a Recombinant Prefusion-Like Form of the F Protein Bound to Bacterium-Like Particles. PLOS ONE 2013, 8, e71072, 10.1371/journal.pone.0071072.
  37. Nathaly Samy; Daniela Reichhardt; Darja Schmidt; Liddy M Chen; Günter Silbernagl; Sanja Vidojkovic; Thomas Ph Meyer; Elke Jordan; Tatiana Adams; Heinz Weidenthaler; et al.Daria StroukovaSonja De CarliPaul Chaplin Safety and immunogenicity of novel modified vaccinia Ankara-vectored RSV vaccine: A randomized phase I clinical trial. Vaccine 2020, 38, 2608-2619, 10.1016/j.vaccine.2020.01.055.
  38. Elke Jordan; Steven J Lawrence; Thomas P H Meyer; Darja Schmidt; Stephanie Schultz; Jutta Mueller; Daria Stroukova; Brigitte Koenen; Robert Gruenert; Guenter Silbernagl; et al.Sanja VidojkovicLiddy M ChenHeinz WeidenthalerNathaly SamyPaul Chaplin Broad Antibody and Cellular Immune Response From a Phase 2 Clinical Trial With a Novel Multivalent Poxvirus-Based Respiratory Syncytial Virus Vaccine. The Journal of Infectious Diseases 2020, XX, 1-11, 10.1093/infdis/jiaa460.
  39. Christopher A Green; Elisa Scarselli; Charles J. Sande; Amber J. Thompson; Catherine M. De Lara; Kathryn S. Taylor; Kathryn Haworth; Mariarosaria Del Sorbo; Brian Angus; Loredana Siani; et al.Stefania Di MarcoCinzia TraboniAntonella FolgoriStefano CollocaStefania CaponeAlessandra VitelliRiccardo CortesePaul KlenermanAlfredo NicosiaAndrew J. Pollard Chimpanzee adenovirus– and MVA-vectored respiratory syncytial virus vaccine is safe and immunogenic in adults. Science Translational Medicine 2015, 7, 300ra126-300ra126, 10.1126/scitranslmed.aac5745.
  40. Christopher A Green; Charles J. Sande; Elisa Scarselli; Stefania Capone; Alessandra Vitelli; Alfredo Nicosia; Laura Silva-Reyes; Amber J. Thompson; Catherine M. De Lara; Kathryn S. Taylor; et al.Kathryn HaworthClaire L. HutchingsTamsin CargillBrian AngusPaul KlenermanAndrew J. Pollard Novel genetically-modified chimpanzee adenovirus and MVA-vectored respiratory syncytial virus vaccine safely boosts humoral and cellular immunity in healthy older adults. Journal of Infection 2019, 78, 382-392, 10.1016/j.jinf.2019.02.003.
  41. Myra N. Widjojoatmodjo; Lies Bogaert; Bob Meek; Roland Zahn; Jort Vellinga; Jerome Custers; Jan Serroyen; Katarina Radošević; Hanneke Schuitemaker; Recombinant low-seroprevalent adenoviral vectors Ad26 and Ad35 expressing the respiratory syncytial virus (RSV) fusion protein induce protective immunity against RSV infection in cotton rats. Vaccine 2015, 33, 5406-5414, 10.1016/j.vaccine.2015.08.056.
  42. Jerald Sadoff; Els De Paepe; Wouter Haazen; Edmund Omoruyi; Arangassery R Bastian; Christy Comeaux; Esther Heijnen; Cynthia Strout; Hanneke Schuitemaker; Benoit Callendret; et al. OUP accepted manuscript. The Journal of Infectious Diseases 2020, XX, 1-10, 10.1093/infdis/jiaa409.
  43. A Randomized, Double-Blind, Phase 1/2a Study to Evaluate the Safety, Tolerability and Immunogenicity of Ad26.RSV.PreF in Adults 18 to 50 Years of Age, RSV-Seropositive Toddlers 12 to 24 Months of Age . ClinicalTrials.gov. Retrieved 2021-1-3
  44. A Randomized, Controlled, Observer-Blind, Phase 1/2a Study to Evaluate the Safety, Reactogenicity, and Immunogenicity of Ad26.RSV.PreF in RSV-Seronegative Toddlers 12 to 24 Months of Age . ClinicalTrials.gov. Retrieved 2021-1-3
  45. A Study to Evaluate Safety, Reactogenicity and Immunogenicity of GSK Biologicals’ RSV Investigational Vaccine Based on Viral Proteins Encoded by Chimpanzee-Derived Adenovector (ChAd155-RSV) (GSK3389245A) in RSV-Seropositive Infants . ClinicalTrials.gov. Retrieved 2021-1-3
  46. David I. Bernstein; Elissa Malkin; Nazha Abughali; Judith Falloon; Tingting Yi; Filip Dubovsky; Phase 1 Study of the Safety and Immunogenicity of a Live, Attenuated Respiratory Syncytial Virus and Parainfluenza Virus Type 3 Vaccine in Seronegative Children. The Pediatric Infectious Disease Journal 2012, 31, 109-114, 10.1097/inf.0b013e31823386f1.
  47. A Phase I Double-Blind Placebo Controlled Trial to Evaluate the Safety and Immunogenicity of Intranasal Sendai Virus Vectored Respiratory Syncytial Virus (SeVRSV) Vaccine in Healthy Adults . ClinicalTrials.gov. Retrieved 2021-1-3
  48. Noushin Jaberolansar; Istvan Toth; Paul R Young; Mariusz Skwarczynski; Recent advances in the development of subunit-based RSV vaccines. Expert Review of Vaccines 2015, 15, 53-68, 10.1586/14760584.2016.1105134.
  49. Hyun Wha Kim; Jose G. Canchola; Carl D. Brandt; Gloria Pyles; Robert M. Chanock; Keith Jensen; Robert H. Parrott; RESPIRATORY SYNCYTIAL VIRUS DISEASE IN INFANTS DESPITE PRIOR ADMINISTRATION OF ANTIGENIC INACTIVATED VACCINE12. American Journal of Epidemiology 1969, 89, 422-434, 10.1093/oxfordjournals.aje.a120955.
  50. Albert Z. Kapikian; Reginald H. Mitchell; Robert M. Chanock; Ruth A. Shvedoff; C. Eleanor Stewart; AN EPIDEMIOLOGIC STUDY OF ALTERED CLINICAL REACTIVITY TO RESPIRATORY SYNCYTIAL (RS) VIRUS INFECTION IN CHILDREN PREVIOUSLY VACCINATED WITH AN INACTIVATED RS VIRUS VACCINE. American Journal of Epidemiology 1969, 89, 405-421, 10.1093/oxfordjournals.aje.a120954.
  51. Natalie I Mazur; Deborah Higgins; Marta C Nunes; José A. Melero; Annefleur C Langedijk; Nicole Horsley; Ursula J Buchholz; Peter J Openshaw; Jason S McLellan; Janet A Englund; et al.Asuncion MejiasRuth A KarronEric Af SimõesIvana KnezevicOctavio RamiloPedro A PiedraHelen Y. ChuAnn R FalseyHarish NairLeyla Kragten-TabatabaieAnne GreenoughEugenio BaraldiNikolaos G PapadopoulosJohan VekemansFernando P PolackMair PowellAshish SatavEdward E WalshRenato T SteinBarney S. GrahamLouis J. Bont The respiratory syncytial virus vaccine landscape: lessons from the graveyard and promising candidates. The Lancet Infectious Diseases 2018, 18, e295-e311, 10.1016/s1473-3099(18)30292-5.
  52. A Phase I Randomized, Open-Label Clinical Trial to Evaluate Dose, Safety, Tolerability and Immunogenicity of a Stabilized Prefusion RSV F Subunit Protein Vaccine, VRC-RSVRGP084-00-VP (DS-Cav1), Alone or with Alum Adjuvant, in Healthy Adults . ClinicalTrials.gov. Retrieved 2021-1-3
  53. Michelle C. Crank; Tracy J. Ruckwardt; Man Chen; Kaitlyn M. Morabito; Emily Phung; Pamela J. Costner; LaSonji A. Holman; Somia P. Hickman; Nina M. Berkowitz; Ingelise J. Gordon; et al.Galina V. YamshchikovMartin R. GaudinskiAzad KumarLauren A. ChangSyed M. MoinJuliane P. HillAnthony T. DiPiazzaRichard M. SchwartzLisa KueltzoJonathan W. CooperPeifeng ChenJudith A. SteinKevin CarltonJason G. GallMartha NasonPeter D. KwongGrace L. ChenJohn R. MascolaJason S. McLellanJulie E. LedgerwoodBarney S. Grahamthe VRC 317 Study Team A proof of concept for structure-based vaccine design targeting RSV in humans. Science 2019, 365, 505-509, 10.1126/science.aav9033.
  54. Iebe Rossey; Xavier Saelens; Vaccines against human respiratory syncytial virus in clinical trials, where are we now?. Expert Review of Vaccines 2019, 18, 1053-1067, 10.1080/14760584.2019.1675520.
  55. A Study to Evaluate the Safety and Immunogenicity of an Adjuvanted RSV Vaccine in Healthy Older Adults . ClinicalTrials.gov. Retrieved 2021-1-3
  56. A Study to Describe the Safety and Immunogenicity of a RSV Vaccine in Healthy Adults . ClinicalTrials.gov. Retrieved 2021-1-3
  57. A STUDY OF A RSV VACCINE WHEN GIVEN TOGETHER WITH TDAP IN HEALTHY NONPREGNANT WOMEN AGED BETWEEN 18 TO 49 YEARS . ClinicalTrials.gov. Retrieved 2021-1-3
  58. Joanne M. Langley; Lisa D Macdonald; Genevieve M Weir; Donna MacKinnon-Cameron; Lingyun Ye; Shelly McNeil; Bert Schepens; Xavier Saelens; Marianne M Stanford; Scott A Halperin; 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. Journal of Infectious Diseases 2018, 218, 378-387, 10.1093/infdis/jiy177.
  59. A Phase I, first in human (FIH), randomised, double-blind, placebo-controlled, dose-escalation study to evaluate safety, reactogenicity and immunogenicity of the recombinant respiratory syncytial virus vaccines (BARS13) when administered intramuscularly (IM) to healthy adult volunteers. . Australian New Zealand Clinical Trials Register. Retrieved 2021-1-3
  60. RSV Clinical Trial Tracker . PATH. Retrieved 2021-1-3
  61. Peter F. Wright; Ruth A. Karron; Robert B. Belshe; Jian R. Shi; Valerie B. Randolph; Peter L. Collins; Alice F. O'shea; William C. Gruber; Brian R. Murphy; The absence of enhanced disease with wild type respiratory syncytial virus infection occurring after receipt of live, attenuated, respiratory syncytial virus vaccines. Vaccine 2007, 25, 7372-7378, 10.1016/j.vaccine.2007.08.014.
  62. Ruth A. Karron; Ursula J. Buchholz; Peter L. Collins; Live-Attenuated Respiratory Syncytial Virus Vaccines. Current Topics in Microbiology and Immunology 2013, 372, 259-284, 10.1007/978-3-642-38919-1_13.
  63. Jorge A. Soto; Laura M. Stephens; Kody A. Waldstein; Gisela Canedo-Marroquín; Steven M. Varga; Alexis M. Kalergis; Current Insights in the Development of Efficacious Vaccines Against RSV. Frontiers in Immunology 2020, 11, 1507, 10.3389/fimmu.2020.01507.
  64. Pablo F. Céspedes; Emma Rey-Jurado; Janyra A. Espinoza; Claudia A. Rivera; Gisela Canedo-Marroquín; Susan M. Bueno; Alexis M. Kalergis; A single, low dose of a cGMP recombinant BCG vaccine elicits protective T cell immunity against the human respiratory syncytial virus infection and prevents lung pathology in mice. Vaccine 2017, 35, 757-766, 10.1016/j.vaccine.2016.12.048.
  65. A Study to Assess Safety, Tolerability and Immunogenicity of the Live Attenuated hRSV Vaccine rBCG-N-hRSV (EVA-VRS01) . ClinicalTrials.gov. Retrieved 2021-1-3
More
Information
Subjects: Virology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 774
Revisions: 4 times (View History)
Update Date: 04 Jan 2021
1000/1000
Video Production Service