Structure-Based Design of Nipah Virus Vaccines: A Generalizable Approach to Paramyxovirus Immunogen Development

Rebecca J Loomis, Guillaume B E Stewart-Jones, Yaroslav Tsybovsky, Ria T Caringal, Kaitlyn M Morabito, Jason S McLellan, Amy L Chamberlain, Sean T Nugent, Geoffrey B Hutchinson, Lisa A Kueltzo, John R Mascola, Barney S Graham, Rebecca J Loomis, Guillaume B E Stewart-Jones, Yaroslav Tsybovsky, Ria T Caringal, Kaitlyn M Morabito, Jason S McLellan, Amy L Chamberlain, Sean T Nugent, Geoffrey B Hutchinson, Lisa A Kueltzo, John R Mascola, Barney S Graham

Abstract

Licensed vaccines or therapeutics are rarely available for pathogens with epidemic or pandemic potential. Developing interventions for specific pathogens and defining generalizable approaches for related pathogens is a global priority and inherent to the UN Sustainable Development Goals. Nipah virus (NiV) poses a significant epidemic threat, and zoonotic transmission from bats-to-humans with high fatality rates occurs almost annually. Human-to-human transmission of NiV has been documented in recent outbreaks leading public health officials and government agencies to declare an urgent need for effective vaccines and therapeutics. Here, we evaluate NiV vaccine antigen design options including the fusion glycoprotein (F) and the major attachment glycoprotein (G). A stabilized prefusion F (pre-F), multimeric G constructs, and chimeric proteins containing both pre-F and G were developed as protein subunit candidate vaccines. The proteins were evaluated for antigenicity and structural integrity using kinetic binding assays, electron microscopy, and other biophysical properties. Immunogenicity of the vaccine antigens was evaluated in mice. The stabilized pre-F trimer and hexameric G immunogens both induced serum neutralizing activity in mice, while the post-F trimer immunogen did not elicit neutralizing activity. The pre-F trimer covalently linked to three G monomers (pre-F/G) induced potent neutralizing antibody activity, elicited responses to the greatest diversity of antigenic sites, and is the lead candidate for clinical development. The specific stabilizing mutations and immunogen designs utilized for NiV were successfully applied to other henipaviruses, supporting the concept of identifying generalizable solutions for prototype pathogens as an approach to pandemic preparedness.

Keywords: G attachment protein; Nipah virus; pandemic preparedness; pre-F/G chimeric immunogen; stabilized prefusion F; structure-based vaccine design.

Copyright © 2020 Loomis, Stewart-Jones, Tsybovsky, Caringal, Morabito, McLellan, Chamberlain, Nugent, Hutchinson, Kueltzo, Mascola and Graham.

Figures

FIGURE 1
FIGURE 1
Structure-based design of a stabilized prefusion NiV F trimer. (A) Structure of prefusion NiV F glycoprotein trimer (PDB ID 5EVM) in green, orange and sand, and with residues that undergo >5 Å conformational change to transition to the postfusion conformation shown in black. GCN4 trimerization (TD) motif is shown in magenta, linked to NiV F residue 488. Zoom insets highlight mutated residues (red) to stabilize the prefusion F structure including L104C-I114C disulfide bond, a S191P helix-breaking proline substitution and a L172F cavity-filling mutation. (B) Two-dimensional class averages of the prefusion F trimer (left), postfusion F trimer (middle) and prefusion F trimer bound to h5B3 Fab (right) obtained by negative-stain electron microscopy (EM). (C) Binding kinetics were measured using a fortéBio Octet Red384 instrument. Table summarizing binding affinities of F designs to monoclonal antibody h5B3. (D) Non-reduced and reduced SDS-PAGE analysis of prefusion and postfusion NiV F glycoproteins.
FIGURE 2
FIGURE 2
Structure-based design of NiV G immunogens and NiV F/G chimeric immunogens. (A) Two-dimensional class averages of NiV G head domain multimer designs (left: trimeric G, center: hexameric G, right: ferritin-G) obtained by negative-stain EM. (B) Negative-stain EM analysis of NiV F/G chimeras, showing pre-F/G (left), G/pre-F (center), and post-F/G (right). TD, trimerization domain.
FIGURE 3
FIGURE 3
Thermodynamic and colloidal stability assessment of Pre-F, Post-F, Hex G, and Pre-F/G constructs. (A) Differential scanning calorimetry; based on the Tm (transition midpoint), the thermodynamic stability of the individual domains can be ranked Post-F>> Hex G > Pre-F, and Pre-F/G. (B) Dynamic light scattering indicates a similar degree of colloidal stability for Hex G, Pre-F and Pre-F/G, while Post-F is extremely colloidally stable, as indicated by lack of change in size within the temperature range of the experiment. (C) Table summarizing the conformational transitions (A) and colloidal stability (B) of Pre-F, Post-F, Hex G, and Pre-F/G.
FIGURE 4
FIGURE 4
Immunogenicity of NiV Pre-F stabilized immunogens, multimeric forms of G and Pre-F/G chimeric immunogens. (A,B) Recognition of pre-F (A) or post-F (B) NiV F proteins by sera from mice immunized twice with NiV F designs or unimmunized. (C,D) Recognition of pre-F NiV F (C) or mono G (D) protein by sera from mice immunized twice with NiV F, NiV G multimers or NiV F/G chimeric designs or unimmunized. (E) Recognition of pre-F NiV F or mono G proteins by sera from mice immunized twice with NiV Hex G or NiV Stalk G. Binding kinetics were measured using a fortéBio Octet Red384 instrument. Line represents mean of all animals in each group ± standard deviation (using GraphPad Prism).
FIGURE 5
FIGURE 5
Neutralization of NiVF/G VSVΔG-luciferase pseudovirus by sera from mice immunized with NiV stabilized pre-F, multimeric forms of G and F/G chimeric immunogens. VSVΔG-luciferase pseudovirus (expresses both NiV FWT and NiV G on surface) neutralization assays were performed on individual mouse sera collected at week 5. The log10 reciprocal IC80 titer for each sample was calculated by curve fitting and non-linear regression using GraphPad Prism. P-values were calculated using one-way ANOVA with Tukey’s multiple comparisons test (***p < 0.001, ****p < 0.0001). Line represents mean of log10 reciprocal IC80 titer ± standard deviation (using GraphPad Prism).
FIGURE 6
FIGURE 6
Application of NiV antigen designs to phylogenetically related Henipaviruses. (A,B) Two-dimensional negative-stain EM class averages of NiV designs applied to Hendra (A) and Cedar (B) viruses. (C) Binding kinetics of HeV F pre-F, post-F, and NiV pre-F/HeV G chimeric immunogen binding to h5B3, NiV pre-F-specific antibody. (D) Binding kinetics of multimeric forms of NiV G, HeV G, CedPV G and NiV pre-F/HeV G chimeric immunogen binding to m102.4, HeV G-specific antibody. (E,F) Recognition of HeV pre-F (E) or HeV Mono G (F) proteins by sera from mice immunized twice with NiV F, G or F/G immunogens. Binding kinetics were measured using a fortéBio Octet Red384 instrument. Line represents mean of all animals in each group ± standard deviation (using GraphPad Prism).

References

    1. Chua KB, Bellini WJ, Rota PA, Harcourt BH, Tamin A, Lam SK, et al. Nipah virus: a recently emergent deadly paramyxovirus. Science. (2000) 288:1432–5. 10.1126/science.288.5470.1432
    1. Drexler JF, Corman VM, Gloza-Rausch F, Seebens A, Annan A, Ipsen A, et al. Henipavirus RNA in African bats. PLoS One. (2009) 4:e6367. 10.1371/journal.pone.0006367
    1. Hayman DT, Wang L-F, Barr J, Baker KS, Suu-Ire R, Broder CC, et al. Antibodies to henipavirus or henipa-like viruses in domestic pigs in Ghana. West Africa. PLoS One. (2011) 6:e25256. 10.1371/journal.pone.0025256
    1. Murray K, Selleck P, Hooper P, Hyatt A, Gould A, Gleeson L, et al. A morbillivirus that caused fatal disease in horses and humans. Science. (1995) 268:94–7. 10.1126/science.7701348
    1. Wang LF, Yu M, Hansson E, Pritchard LI, Shiell B, Michalski WP, et al. The exceptionally large genome of Hendra virus: support for creation of a new genus within the family Paramyxoviridae. J Virol. (2000) 74:9972–9. 10.1128/jvi.74.21.9972-9979.2000
    1. Wu Z, Yang L, Yang F, Ren X, Jiang J, Dong J, et al. Novel henipa-like virus, mojiang paramyxovirus, in rats, China, 2012. Emerg Infect Dis. (2014) 20:1064–6. 10.3201/eid2006.131022
    1. Drexler JF, Corman VM, Müller MA, Maganga GD, Vallo P, Binger T, et al. Bats host major mammalian paramyxoviruses. Nat Commun. (2012) 3:796. 10.1038/ncomms1796
    1. Chua KB, Goh KJ, Wong KT, Kamarulzaman A, Tan PS, Ksiazek TG, et al. Fatal encephalitis due to Nipah virus among pig-farmers in Malaysia. Lancet. (1999) 354:1257–9. 10.1016/S0140-6736(99)04299-3
    1. Mohd NMN, Gan CH, Ong BL. Nipah virus infection of pigs in peninsular Malaysia. Rev Sci Tech. (2000) 19:160–5. 10.20506/rst.19.1.1202
    1. Goh KJ, Tan CT, Chew NK, Tan PS, Kamarulzaman A, Sarji SA, et al. Clinical features of Nipah virus encephalitis among pig farmers in Malaysia. N Engl J Med. (2000) 342:1229–35. 10.1056/NEJM200004273421701
    1. Banerjee S, Gupta N, Kodan P, Mittal A, Ray Y, Nischal N, et al. Nipah virus disease: A rare and intractable disease. Intractable Rare Dis Res. (2019) 8:1–8. 10.5582/irdr.2018.01130
    1. Chadha MS, Comer JA, Lowe L, Rota PA, Rollin PE, Bellini WJ, et al. Nipah virus-associated encephalitis outbreak, Siliguri, India. Emerg Infect Dis. (2006) 12:235–40. 10.3201/eid1202.051247
    1. Ching PK, De Los Reyes VC, Sucaldito MN, Tayag E, Columna-Vingno AB, Malbas FF, Jr., et al. Outbreak of henipavirus infection, Philippines, 2014. Emerg Infect Dis. (2015) 21:328–31.
    1. Harcourt BH, Lowe L, Tamin A, Liu X, Bankamp B, Bowden N, et al. Genetic characterization of Nipah virus, Bangladesh, 2004. Emerg Infect Dis. (2005) 11:1594–7. 10.3201/eid1110.050513
    1. Hsu VP, Hossain MJ, Parashar UD, Ali MM, Ksiazek TG, Kuzmin I, et al. Nipah virus encephalitis reemergence, Bangladesh. Emerg Infect Dis. (2004) 10:2082–7. 10.3201/eid1012.040701
    1. Luby SP, Gurley ES, Hossain MJ. Transmission of human infection with Nipah virus. Clin Infect Dis. (2009) 49:1743–8.
    1. Ochani RK, Batra S, Shaikh A, Asad A. Nipah virus - the rising epidemic: a review. Infez Med. (2019) 27:117–27.
    1. Arankalle VA, Bandyopadhyay BT, Ramdasi AY, Jadi R, Patil DR, Rahman M, et al. Genomic characterization of Nipah virus, West Bengal, India. Emerg Infect Dis. (2011) 17:907–9.
    1. Arunkumar G, Chandni R, Mourya DT, Singh SK, Sadanandan R, Sudan P, et al. Outbreak Investigation of Nipah Virus Disease in Kerala, India, 2018. J Infect Dis. (2019) 219:1867–78. 10.1093/infdis/jiy612
    1. Gurley ES, Montgomery JM, Hossain MJ, Bell M, Azad AK, Islam MR, et al. Person-to-person transmission of Nipah virus in a Bangladeshi community. Emerg Infect Dis. (2007) 13:1031–7. 10.3201/eid1307.061128
    1. Luby SP, Hossain MJ, Gurley ES, Ahmed B-N, Banu S, Khan SU, et al. Recurrent zoonotic transmission of Nipah virus into humans, Bangladesh, 2001-2007. Emerg Infect Dis. (2009) 15:1229–35. 10.3201/eid1508.081237
    1. Sweileh WM. Global research trends of World Health Organization’s top eight emerging pathogens. Global Health. (2017) 13:9. 10.1186/s12992-017-0233-9
    1. Abdullah S, Tan CT. Henipavirus encephalitis. Handb Clin Neurol. (2014) 123:663–70.
    1. Lo MK, Rota PA. The emergence of Nipah virus, a highly pathogenic paramyxovirus. J Clin Virol. (2008) 43:396–400. 10.1016/j.jcv.2008.08.007
    1. Prescott J, De Wit E, Feldmann H, Munster VJ. The immune response to Nipah virus infection. Arch Virol. (2012) 157:1635–41. 10.1007/s00705-012-1352-5
    1. Wong KT, Tan CT. Clinical and pathological manifestations of human henipavirus infection. Curr Top Microbiol Immunol. (2012) 359:95–104. 10.1007/82_2012_205
    1. Clayton BA. Nipah virus: transmission of a zoonotic paramyxovirus. Curr Opin Virol. (2017) 22:97–104. 10.1016/j.coviro.2016.12.003
    1. Clayton BA, Wang LF, Marsh GA. Henipaviruses: an updated review focusing on the pteropid reservoir and features of transmission. Zoonoses Public Health. (2013) 60:69–83. 10.1111/j.1863-2378.2012.01501.x
    1. Gurley ES, Hegde ST, Hossain K, Sazzad H, Hossain MJ, Rahman M, et al. Convergence of humans, bats, trees, and culture in nipah virus transmission, Bangladesh. Emerg Infect Dis. (2017) 23:1446–53. 10.3201/eid2309.161922
    1. AbuBakar S, Chang L-Y, Ali ARM, Sharifah SH, Yusoff K, Zamrod Z. Isolation and molecular identification of Nipah virus from pigs. Emerg Infect Dis. (2004) 10:2228–30. 10.3201/eid1012.040452
    1. Ang BSP, Lim TCC, Wang L. Nipah virus infection. J Clin Microbiol. (2018) 56:e01875-17.
    1. Chua KB. Nipah virus outbreak in Malaysia. J Clin Virol. (2003) 26:265–75.
    1. Field H, Young P, Yob JM, Mills J, Hall L, Mackenzie J. The natural history of Hendra and Nipah viruses. Microbes Infect. (2001) 3:307–14. 10.1016/s1286-4579(01)01384-3
    1. Geisbert TW, Feldmann H, Broder CC. Animal challenge models of henipavirus infection and pathogenesis. Curr Top Microbiol Immunol. (2012) 359:153–77. 10.1007/82_2012_208
    1. Glennon EE, Restif O, Sbarbaro SR, Garnier R, Cunningham AA, Suu-Ire RD, et al. Domesticated animals as hosts of henipaviruses and filoviruses: a systematic review. Vet J. (2018) 233:25–34. 10.1016/j.tvjl.2017.12.024
    1. Yob JM, Field H, Rashdi AM, Morrissy C, van der Heide B, Rota P, et al. Nipah virus infection in bats (order Chiroptera) in peninsular Malaysia. Emerg Infect Dis. (2001) 7:439–41.
    1. Bloom DE, Cadarette D. Infectious disease threats in the twenty-first century: strengthening the global response. Front Immunol. (2019) 10:549. 10.3389/fimmu.2019.00549
    1. Lamb RA, Kolakofsky D. Paramyxoviridae: THE viruses and their replication. 3rd ed In: FieldsBN, KnipeDM, HowleyPM editors. Fundamental virology. (Vol. xv), Philadelphia, PA: Lippincott-Raven; (1996). p.1340.
    1. Tamin A, Harcourt BH, Ksiazek TG, Rollin PE, Bellini WJ, Rota PA. Functional properties of the fusion and attachment glycoproteins of Nipah virus. Virology. (2002) 296:190–200. 10.1006/viro.2002.1418
    1. Bossart KN, Fusco DL, Broder CC. Paramyxovirus entry. Adv Exp Med Biol. (2013) 790:95–127. 10.1007/978-1-4614-7651-1_6
    1. Lamb RA, Paterson RG, Jardetzky TS. Paramyxovirus membrane fusion: lessons from the F and HN atomic structures. Virology. (2006) 344:30–7. 10.1016/j.virol.2005.09.007
    1. Smith EC, Popa A, Chang A, Masante C, Dutch RE. Viral entry mechanisms: the increasing diversity of paramyxovirus entry. FEBS J. (2009) 276:7217–27. 10.1111/j.1742-4658.2009.07401.x
    1. Wang L, Harcourt BH, Yu M, Tamin A, Rota PA, Bellini WJ, et al. Molecular biology of Hendra and Nipah viruses. Microbes Infect. (2001) 3:279–87. 10.1016/s1286-4579(01)01381-8
    1. Chan YP, Lu M, Dutta S, Yan L, Barr J, Flora M, et al. Biochemical, conformational, and immunogenic analysis of soluble trimeric forms of henipavirus fusion glycoproteins. J Virol. (2012) 86:11457–71. 10.1128/JVI.01318-12
    1. Stewart-Jones GBE, Chuang GY, Xu K, Zhou T, Acharya P, Tsybovsky Y, et al. Structure-based design of a quadrivalent fusion glycoprotein vaccine for human parainfluenza virus types 1-4. Proc Natl Acad Sci USA. (2018) 115:12265–70. 10.1073/pnas.1811980115
    1. Wong JJ, Reay GP, Lamb RA, Jardetzky TS. Structure and stabilization of the Hendra virus F glycoprotein in its prefusion form. Proc Natl Acad Sci USA. (2016) 113:1056–61. 10.1073/pnas.1523303113
    1. Xu K, Chan Y-P, Bradel-Tretheway B, Akyol-Ataman Z, Zhu Y, Dutta S, et al. Crystal Structure of the pre-fusion nipah virus fusion glycoprotein reveals a novel hexamer-of-trimers assembly. PLoS Pathog. (2015) 11:e1005322. 10.1371/journal.ppat.1005322
    1. McLellan JS, Chen M, Joyce MG, Sastry M, Stewart-Jones GBE, Yang Y, et al. Structure-based design of a fusion glycoprotein vaccine for respiratory syncytial virus. Science. (2013) 342:592–8. 10.1126/science.1243283
    1. McLellan JS, Chen M, Leung S, Graepel KW, Du X, Yang Y, et al. Structure of RSV fusion glycoprotein trimer bound to a prefusion-specific neutralizing antibody. Science. (2013) 340:1113–7. 10.1126/science.1234914
    1. Crank MC, Ruckwardt TJ, Chen M, Morabito KM, Phung E, Costner PJ, et al. A proof of concept for structure-based vaccine design targeting RSV in humans. Science. (2019) 365:505–9. 10.1126/science.aav9033
    1. Bonaparte MI, Dimitrov AS, Bossart KN, Crameri G, Mungall BA, Bishop KA, et al. Ephrin-B2 ligand is a functional receptor for Hendra virus and Nipah virus. Proc Natl Acad Sci USA. (2005) 102:10652–7. 10.1073/pnas.0504887102
    1. Bossart KN, Wang L-F, Flora MN, Chua KB, Lam SK, Eaton BT, et al. Membrane fusion tropism and heterotypic functional activities of the Nipah virus and Hendra virus envelope glycoproteins. J Virol. (2002) 76:11186–98. 10.1128/jvi.76.22.11186-11198.2002
    1. Bowden TA, Aricescu AR, Gilbert RJC, Grimes JM, Jones EY, Stuart DI. Structural basis of Nipah and Hendra virus attachment to their cell-surface receptor ephrin-B2. Nat Struct Mol Biol. (2008) 15:567–72. 10.1038/nsmb.1435
    1. Negrete OA, Levroney EL, Aguilar HC, Bertolotti-Ciarlet A, Nazarian R, Tajyar S, et al. EphrinB2 is the entry receptor for Nipah virus, an emergent deadly paramyxovirus. Nature. (2005) 436:401–5. 10.1038/nature03838
    1. Xu K, Broder CC, Nikolov DB. Ephrin-B2 and ephrin-B3 as functional henipavirus receptors. Semin Cell Dev Biol. (2012) 23:116–23. 10.1016/j.semcdb.2011.12.005
    1. Xu K, Rajashankar KR, Chan Y-P, Himanen JP, Broder CC, Nikolov DB. Host cell recognition by the henipaviruses: crystal structures of the Nipah G attachment glycoprotein and its complex with ephrin-B3. Proc Natl Acad Sci USA. (2008) 105:9953–8. 10.1073/pnas.0804797105
    1. Bossart KN, Crameri G, Dimitrov AS, Mungall BA, Feng Y-R, Patch JR, et al. Receptor binding, fusion inhibition, and induction of cross-reactive neutralizing antibodies by a soluble G glycoprotein of Hendra virus. J Virol. (2005) 79:6690–702. 10.1128/JVI.79.11.6690-6702.2005
    1. Pallister J, Middleton D, Wang L-F, Klein R, Haining J, Robinson R, et al. A recombinant Hendra virus G glycoprotein-based subunit vaccine protects ferrets from lethal Hendra virus challenge. Vaccine. (2011) 29:5623–30. 10.1016/j.vaccine.2011.06.015
    1. Pallister JA, Klein R, Arkinstall R, Haining J, Long F, White JR, et al. Vaccination of ferrets with a recombinant G glycoprotein subunit vaccine provides protection against Nipah virus disease for over 12 months. Virol J. (2013) 10:237. 10.1186/1743-422X-10-237
    1. Bossart KN, Rockx B, Feldmann F, Brining D, Scott D, LaCasse R, et al. A Hendra virus G glycoprotein subunit vaccine protects African green monkeys from Nipah virus challenge. Sci Transl Med. (2012) 4:146ra107. 10.1126/scitranslmed.3004241
    1. Mire CE, Geisbert JB, Agans KN, Feng Y-R, Fenton KA, Bossart KN, et al. A recombinant Hendra virus G glycoprotein subunit vaccine protects nonhuman primates against Hendra virus challenge. J Virol. (2014) 88:4624–31. 10.1128/JVI.00005-14
    1. Middleton D, Pallister J, Klein R, Feng Y-R, Haining J, Arkinstall R, et al. Hendra virus vaccine, a one health approach to protecting horse, human, and environmental health. Emerg Infect Dis. (2014) 20:372–9. 10.3201/eid2003.131159
    1. Steffen DL, Xu K, Nikolov DB, Broder CC. Henipavirus mediated membrane fusion, virus entry and targeted therapeutics. Viruses. (2012) 4:280–308. 10.3390/v4020280
    1. Tan R, Hodge A, Klein R, Edwards N, Huang JA, Middleton D, et al. Virus-neutralising antibody responses in horses following vaccination with Equivac(R) HeV: a field study. Aust Vet J. (2018) 96:161–6. 10.1111/avj.12694
    1. Weingartl HM, Berhane Y, Caswell JL, Loosmore S, Audonnet JC, Roth JA, et al. Recombinant nipah virus vaccines protect pigs against challenge. J Virol. (2006) 80:7929–38. 10.1128/JVI.00263-06
    1. Guillaume V, Contamin H, Loth P, Georges-Courbot M-C, Lefeuvre A, Marianneau P, et al. Nipah virus: vaccination and passive protection studies in a hamster model. J Virol. (2004) 78:834–40. 10.1128/jvi.78.2.834-840.2004
    1. Ploquin A, Szécsi J, Mathieu C, Guillaume V, Barateau V, Ong KC, et al. Protection against henipavirus infection by use of recombinant adeno-associated virus-vector vaccines. J Infect Dis. (2013) 207:469–78. 10.1093/infdis/jis699
    1. Lo MK, Bird BH, Chattopadhyay A, Drew CP, Martin BE, Coleman JD, et al. Single-dose replication-defective VSV-based Nipah virus vaccines provide protection from lethal challenge in Syrian hamsters. Antiviral Res. (2014) 101:26–9. 10.1016/j.antiviral.2013.10.012
    1. Mire CE, Geisbert JB, Agans KN, Versteeg KM, Deer DJ, Satterfield BA, et al. Use of single-injection recombinant vesicular stomatitis virus vaccine to protect nonhuman primates against lethal nipah virus disease. Emerg Infect Dis. (2019) 25:1144–52. 10.3201/eid2506.181620
    1. Mire CE, Versteeg KM, Cross RW, Agans KN, Fenton KA, Whitt MA, et al. Single injection recombinant vesicular stomatitis virus vaccines protect ferrets against lethal Nipah virus disease. Virol J. (2013) 10:353. 10.1186/1743-422x-10-353
    1. Keshwara R, Shiels T, Postnikova E, Kurup D, Wirblich C, Johnson RF, et al. Erratum: Publisher Correction: rabies-based vaccine induces potent immune responses against Nipah virus. NPJ Vaccines. (2019) 4:18. 10.1038/s41541-019-0112-x
    1. Keshwara R, Shiels T, Postnikova E, Kurup D, Wirblich C, Johnson RF, et al. Rabies-based vaccine induces potent immune responses against Nipah virus. NPJ Vaccines. (2019) 4:15. 10.1126/scitranslmed.3008929
    1. Kurup D, Wirblich C, Feldmann H, Marzi A, Schnell MJ. Rhabdovirus-based vaccine platforms against henipaviruses. J Virol. (2015) 89:144–54. 10.1128/JVI.02308-14
    1. Yoneda M, Georges-Courbot M-C, Ikeda F, Ishii M, Nagata N, Jacquot F, et al. Recombinant measles virus vaccine expressing the Nipah virus glycoprotein protects against lethal Nipah virus challenge. PLoS One. (2013) 8:e58414. 10.1371/journal.pone.0058414
    1. Zhu Z, Bossart KN, Bishop KA, Crameri G, Dimitrov AS, McEachern JA, et al. Exceptionally potent cross-reactive neutralization of Nipah and Hendra viruses by a human monoclonal antibody. J Infect Dis. (2008) 197:846–53.
    1. Zhu Z, Dimitrov AS, Bossart KN, Crameri G, Bishop KA, Choudhry V, et al. Potent neutralization of Hendra and Nipah viruses by human monoclonal antibodies. J Virol. (2006) 80:891–9.
    1. Bossart KN, Geisbert TW, Feldmann H, Zhu Z, Feldmann F, Geisbert JB, et al. A neutralizing human monoclonal antibody protects african green monkeys from hendra virus challenge. Sci Transl Med. (2011) 3:105ra103. 10.1126/scitranslmed.3002901
    1. Bossart KN, Zhu Z, Middleton D, Klippel J, Crameri G, Bingham J, et al. A neutralizing human monoclonal antibody protects against lethal disease in a new ferret model of acute nipah virus infection. PLoS Pathog. (2009) 5:e1000642. 10.1371/journal.ppat.1000642
    1. Dang HV, Chan Y-P, Park Y-J, Snijder J, Da Silva SC, Vu B, et al. An antibody against the F glycoprotein inhibits Nipah and Hendra virus infections. Nat Struct Mol Biol. (2019) 26:980–7. 10.1038/s41594-019-0308-9
    1. Mastronarde DN. Automated electron microscope tomography using robust prediction of specimen movements. J Struct Biol. (2005) 152:36–51. 10.1016/j.jsb.2005.07.007
    1. Tang G, Peng L, Baldwin PR, Mann DS, Jiang W, Rees I, et al. EMAN2: an extensible image processing suite for electron microscopy. J Struct Biol. (2007) 157:38–46. 10.1016/j.jsb.2006.05.009
    1. Scheres SH. RELION: implementation of a Bayesian approach to cryo-EM structure determination. J Struct Biol. (2012) 180:519–30. 10.1016/j.jsb.2012.09.006
    1. Yee-Peng CC, Broder CC. Antibodies Against F Glycoprotein of Hendra and Nipah VIruses. United States PCT/US15/12641. (2016).
    1. Dimitrov DS, Zhongyu Z, Broder CC. Human Monoclonal Antibodies Against Hendra and Nipah Viruses. United States Patent 7,988,971 B2. (2011). 10.1517/14712590902763755
    1. Petersen RL. Strategies using bio-layer interferometry biosensor technology for vaccine research and development. Biosensors (Basel). (2017) 7:49. 10.3390/bios7040049
    1. Tamin A, Harcourt BH, Lo MK, Roth JA, Wolf MC, Lee B, et al. Development of a neutralization assay for Nipah virus using pseudotype particles. J Virol Methods. (2009) 160:1–6. 10.1016/j.jviromet.2009.02.025
    1. Chattopadhyay A, Rose JK. Complementing defective viruses that express separate paramyxovirus glycoproteins provide a new vaccine vector approach. J Virol. (2011) 85:2004–11. 10.1128/JVI.01852-10
    1. Marsh GA, De Jong C, Barr JA, Tachedjian M, Smith C, Middleton D, et al. Cedar virus: a novel Henipavirus isolated from Australian bats. PLoS Pathog. (2012) 8:e1002836. 10.1371/journal.ppat.1002836
    1. Geisbert TW, Mire CE, Geisbert JB, Chan Y-P, Agans KN, Feldmann F, et al. Therapeutic treatment of Nipah virus infection in nonhuman primates with a neutralizing human monoclonal antibody. Sci Transl Med. (2014) 6:242ra82. 10.1126/scitranslmed.3008929
    1. Bossart KN, Wang LF, Eaton BT, Broder CC. Functional expression and membrane fusion tropism of the envelope glycoproteins of Hendra virus. Virology. (2001) 290:121–35. 10.1006/viro.2001.1158
    1. Crameri G, Wang LF, Morrissy C, White J, Eaton BT. A rapid immune plaque assay for the detection of Hendra and Nipah viruses and anti-virus antibodies. J Virol Methods. (2002) 99:41–51. 10.1016/s0166-0934(01)00377-9
    1. Bossart KN, Broder CC. Viral glycoprotein-mediated cell fusion assays using vaccinia virus vectors. Methods Mol Biol. (2004) 269: 309–32.
    1. Anasir MI, Poh CL. Structural vaccinology for viral vaccine design. Front Microbiol. (2019) 10:738. 10.3389/fmicb.2019.00738
    1. Sesterhenn F, Bonet J, Correia BE. Structure-based immunogen design-leading the way to the new age of precision vaccines. Curr Opin Struct Biol. (2018) 51:163–9. 10.1016/j.sbi.2018.06.002
    1. Gilman MS, Castellanos CA, Chen M, Ngwuta JO, Goodwin E, Moin SM, et al. Rapid profiling of RSV antibody repertoires from the memory B cells of naturally infected adult donors. Sci Immunol. (2016) 1:eaaj1879. 10.1126/sciimmunol.aaj1879
    1. Ngwuta JO, Chen M, Modjarrad K, Joyce MG, Kanekiyo M, Kumar A, et al. Prefusion F-specific antibodies determine the magnitude of RSV neutralizing activity in human sera. Sci Transl Med. (2015) 7:309ra162. 10.1126/scitranslmed.aac4241
    1. Battles MB, Más V, Olmedillas E, Cano O, Vázquez M, Rodríguez L, et al. Structure and immunogenicity of pre-fusion-stabilized human metapneumovirus F glycoprotein. Nat Commun. (2017) 8:1528. 10.1038/s41467-017-01708-9
    1. Kirchdoerfer RN, Cottrell CA, Wang N, Pallesen J, Yassine HM, Turner HL, et al. Pre-fusion structure of a human coronavirus spike protein. Nature. (2016) 531:118–21. 10.1038/nature17200
    1. Pallesen J, Wang N, Corbett KS, Wrapp D, Kirchdoerfer RN, Turner HL, et al. Immunogenicity and structures of a rationally designed prefusion MERS-CoV spike antigen. Proc Natl Acad Sci USA. (2017) 114:E7348–57. 10.1073/pnas.1707304114
    1. Broder CC, Xu K, Nikolov DB, Zhu Z, Dimitrov DS, Middleton D, et al. A treatment for and vaccine against the deadly Hendra and Nipah viruses. Antiviral Res. (2013) 100:8–13. 10.1016/j.antiviral.2013.06.012
    1. Guillaume-Vasselin V, Lemaitre L, Dhondt K, Tedeschi L, Poulard A, Charreyre C, et al. Protection from Hendra virus infection with Canarypox recombinant vaccine. NPJ Vaccines. (2016) 1:16003.
    1. Brideau RJ, Walters RR, Stier MA, Wathen MW. Protection of cotton rats against human respiratory syncytial virus by vaccination with a novel chimeric FG glycoprotein. J Gen Virol. (1989) 70(Pt 10):2637–44.
    1. Garg R, Brownlie R, Latimer L, Gerdts V, Potter A, van Drunen Littel-van den Hurk S. Vaccination with a human parainfluenza virus type 3 chimeric FHN glycoprotein formulated with a combination adjuvant induces protective immunity. Vaccine. (2017) 35:7139–46.
    1. Garg R, Brownlie R, Latimer L, Gerdts V, Potter A, van Drunen Littel-van den Hurk S. A chimeric glycoprotein formulated with a combination adjuvant induces protective immunity against both human respiratory syncytial virus and parainfluenza virus type 3. Antiviral Res. (2018) 158:78–87. 10.1016/j.antiviral.2018.07.021
    1. Wathen MW, Brideau RJ, Thomsen DR, Murphy BR. Characterization of a novel human respiratory syncytial virus chimeric FG glycoprotein expressed using a baculovirus vector. J Gen Virol. (1989) 70(Pt 10):2625–35. 10.1099/0022-1317-70-10-2625
    1. Graham BS, Sullivan NJ. Emerging viral diseases from a vaccinology perspective: preparing for the next pandemic. Nat Immunol. (2018) 19:20–8.
    1. Zhang B, Chen L, Silacci C, Thom M, Boyington JC, Druz A, et al. Protection of calves by a prefusion-stabilized bovine RSV F vaccine. NPJ Vaccines. (2017) 2:7.

Source: PubMed

3
Sottoscrivi