A live RSV vaccine with engineered thermostability is immunogenic in cotton rats despite high attenuation

Christopher C Stobart, Christina A Rostad, Zunlong Ke, Rebecca S Dillard, Cheri M Hampton, Joshua D Strauss, Hong Yi, Anne L Hotard, Jia Meng, Raymond J Pickles, Kaori Sakamoto, Sujin Lee, Michael G Currier, Syed M Moin, Barney S Graham, Marina S Boukhvalova, Brian E Gilbert, Jorge C G Blanco, Pedro A Piedra, Elizabeth R Wright, Martin L Moore, Christopher C Stobart, Christina A Rostad, Zunlong Ke, Rebecca S Dillard, Cheri M Hampton, Joshua D Strauss, Hong Yi, Anne L Hotard, Jia Meng, Raymond J Pickles, Kaori Sakamoto, Sujin Lee, Michael G Currier, Syed M Moin, Barney S Graham, Marina S Boukhvalova, Brian E Gilbert, Jorge C G Blanco, Pedro A Piedra, Elizabeth R Wright, Martin L Moore

Abstract

Respiratory syncytial virus (RSV) is a leading cause of infant hospitalization and there remains no pediatric vaccine. RSV live-attenuated vaccines (LAVs) have a history of safe testing in infants; however, achieving an effective balance of attenuation and immunogenicity has proven challenging. Here we seek to engineer an RSV LAV with enhanced immunogenicity. Genetic mapping identifies strain line 19 fusion (F) protein residues that correlate with pre-fusion antigen maintenance by ELISA and thermal stability of infectivity in live RSV. We generate a LAV candidate named OE4 which expresses line 19F and is attenuated by codon-deoptimization of non-structural (NS1 and NS2) genes, deletion of the small hydrophobic (SH) gene, codon-deoptimization of the attachment (G) gene and ablation of the secreted form of G. OE4 (RSV-A2-dNS1-dNS2-ΔSH-dGm-Gsnull-line19F) exhibits elevated pre-fusion antigen levels, thermal stability, immunogenicity, and efficacy despite heavy attenuation in the upper and lower airways of cotton rats.

Conflict of interest statement

M.L.M. co-founded Meissa Vaccines, Inc. and serves as Chief Scientific Officer for the Company. M.L.M., C.C.S., A.L.H., J.M. and C.A.R. are co-inventors of RSV vaccine technology subject to evaluation in this paper. The vaccine technology has been optioned to Meissa by Emory University. The remaining authors declare no competing financial interests.

Figures

Figure 1. MPE8 and D25 ELISAs.
Figure 1. MPE8 and D25 ELISAs.
(a) Ratio of direct ELISA using MPE8, a pre-F-specific mAb, to direct ELISA using motavizumab, a total F mAb. Values are normalized to strain A2. For A2-line19F mutants, the asterisks show significant differences compared with A2-line19F. (b) Ratio of direct ELISA using D25, another pre-F-specific mAb, to direct ELISA using motavizumab. All graphs represent the means+s.d.'s of at least two experimental replicates, and data were analysed by one-way ANOVA. When significant, P values are shown as a bracket between groups (P<0.0005) or by asterisk when compared with A2-line19F (*P<0.05; **P<0.005; ***P<0.0005).
Figure 2. Thermal stability assays.
Figure 2. Thermal stability assays.
Thermal inactivation was carried out either by incubation of virus at 4 °C (a,c,d) or 37 °C (b). The viruses in c labelled 79, 191, 357, 371, 557 and 357/371 represent A2-line19F containing substitutions at these indicated positions with A2 residues. The virus in c labelled DB1-357/371 represents DB1 with substitutions of line 19F residues at positions 357 and 371. Viruses were harvested at the indicated time points and titrated by FFU or PFU assays. All graphs represent the means+s.d.'s of at least two experimental replicates combined, and data were analysed by two-way ANOVA (*P<0.05; **P<0.005; ***P<0.0005; ****P<0.00005).
Figure 3. Design of live-attenuated vaccine OE4…
Figure 3. Design of live-attenuated vaccine OE4 and expression of viral proteins.
(a) Schematic of RSV LAV OE4 genome including codon deoptimization of the NS1, NS2 and G genes, deletion of the SH gene, and incorporation of the line 19F gene. (b) Western blotting of Vero cells infected with A2 (white), OE4 (green) or OE4 expressing wild-type G (OE4-wtG, grey) for NS1, NS2, N and G. An A2-Gnull mutant was included as a control. (c) Western densitometry analyses were normalized to A2 expression levels. (d) Western blotting of Vero cells infected with mock, A2-line19F (white), OE4-wtG (grey) or OE4 (green) for F, N and GAPDH. (e) Densitometry results were normalized to A2 expression levels. Densitometry results represent the means+s.d.'s of at least two experimental replicates and representative blots are shown. Statistical analyses were performed by one-way ANOVA (***P<0.0005; ****P<0.00005). d, codon-deoptimized; F, fusion protein; G, attachment glycoprotein; L, large polymerase; M, matrix; mK2, monomeric Katushka2; N, nucleoprotein; NS1/NS2, nonstructural proteins 1 and 2; P, phosphoprotein; SH, small hydrophobic protein.
Figure 4. Immunogold labelling of RSV surface…
Figure 4. Immunogold labelling of RSV surface glycoproteins F and G.
(a) Representative TEM images of BEAS-2B cells infected at an MOI of 10 with A2 (black) or OE4 (green) and labelled with MPE8 (pre-F mAb), 131-2A (post-F mAb), Motavizumab (total F mAb) or 131-2G (G mAb) and probed with gold-labelled secondary antibodies. (b) Quantification of the amount of immunogold particles per measured membrane length per virion. For each labeling condition, more than 100 virions (graph data points) were evaluated for each virus. The red lines represent the mean particle densities along the membrane for each condition. Significant differences are indicated by ***P<0.0005 determined by t-test with Welch's correction. Scale bars represent 200 nm.
Figure 5. Cryo-electron tomography of RSV virions…
Figure 5. Cryo-electron tomography of RSV virions and subvolume averaging of the F glycoprotein.
(ac) Tomographic slices (6.14 nm) of A2, OE4 and A2-heat (55 °C for 30 min) virions showing overall virus structure and the organization of surface glycoproteins (insets). Inset in OE4 is rotated 180°. Scale bars are 200 nm for A2 and OE4, and 100 nm for A2-heat. (dl) Subvolume averages and modelling of RSV F structures in pre- and post-fusion conformations. Central slices (6.14 Å in thickness) of the averaged structures lowpass filtered to 40 Å for A2 (d), OE4 (e) and A2-heat (f). Quasi-atomic models generated by fitting the RSV pre-fusion F (PDB ID 4JHW) and RSV post-fusion F (PDB ID 3RRT) crystal structures into the subvolume averages, with side views (gi) and top views (jl) for A2 (g,j), OE4 (h,k) and A2-heat (i,l). Note the height difference between the ectodomain of A2/OE4 and A2-heat. The measurements were made from the top of the membrane to the top of the head domain. Scale bars, 10 nm (df); 5 nm (gl).
Figure 6. OE4 replication in immortalized and…
Figure 6. OE4 replication in immortalized and primary cell cultures.
Vero (a), BEAS-2B (b), primary normal human bronchial epithelial cells differentiated at air–liquid interface (NHBE) (c), primary human tracheobronchial airway cells differentiated at air–liquid interface (HAE) (d) were infected with A2-line19F (black), OE4 (green) and in HAE, OE4+wtG (green dash) at MOI=0.01 (Vero and BEAS-2B), MOI=2.6 (NHBE), or MOI=6.7 (HAE). Samples were titrated by fluorescent focus unit (FFU) assays on Vero cells. (e) Representative images of infected HAE cultures. Scale bar represents 200 μm. Graphs depict the means±s.e.s of the means combined from three experiments (Vero and BEAS-2B), from two donors in duplicate (NHBE), or from six cultures from a single donor per virus (HAE). When significant, P values are shown relative to A2-line19F (*P<0.05; by two-way ANOVA).
Figure 7. Attenuation and efficacy of OE4…
Figure 7. Attenuation and efficacy of OE4 in mice and cotton rats.
(a) Lung viral loads were determined in mice inoculated i.n. with 106 FFU of A2, A2-line19F or OE4 at the indicated time points. (b) Serum nAb titres were measured in mice inoculated with 106 FFU of A2, A2-line19F or OE4. (c) Mice were inoculated with 106 FFU of A2, A2-line19F or OE4 then challenged with 105 PFU of A2-line19F on day 102. Lung viral loads were determined day 4 post challenge. (ac) Graphs represent combined data from two experiments of 5–10 mice per group. (d) Mice (five per group) were inoculated with mock, A2-line19F, OE4 or A2-del-M2-2, and lungs were harvested 8 days post inoculation for histological quantification of airway mucin expression. Each dot represents an airway, and graph shows >300 airways per group in one of two experiments with similar results. (e,f) Viral load on day 4 in cotton rat lung homogenates (n=3) (e) and nasal washes (n=3) (f) following i.n. inoculation with 105 FFU of A2, OE4 or A2-del-M2-2. (g) Cotton rats (six per group) were inoculated with mock, RSV A(Tracy), OE4 or A2-del-M2-2, and serum nAb titres against representative RSV strains were determined on day 42 post inoculation using pooled sera. EC50 was calculated by non-linear regression, and data represent EC50+upper limit of the 95% confidence interval. (h,i) Cotton rats (five per group) were inoculated with mock, A2, or OE4, challenged on day 42 with 106 FFU of RSV A2-line19F, and viral loads on day 46 were measured in nasal washes (i) and lung lavages (h). *P<0.05; **P<0.005; ***P<0.0005; ****P<0.00005 by one-way (cf,h,i) or two-way (a,b) ANOVA.
Figure 8. Histopathology following RSV challenge in…
Figure 8. Histopathology following RSV challenge in cotton rats.
To evaluate for vaccine attributable enhanced disease post challenge, groups of five cotton rats were inoculated intramuscularly with FI-RSV or intranasally with either mock or OE4. Animals vaccinated with FI-RSV also received a boost on day 21 p.i. All animals were challenged with A2-line19F on day 42 p.i., lungs were harvested 4 days later and histopathology scores were performed (a). Representative haematoxylin and eosin stains for FI-RSV (b), mock (c) and OE4 (d) vaccinated rats are shown. Scale bars represent 200 μm. Data are represented as mean+s.d. *P<0.05, **P<0.005 by two-way ANOVA.

References

    1. Kim H. W. et al.. Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine. Am. J. Epidemiol. 89, 422–434 (1969).
    1. Schickli J. H., Dubovsky F. & Tang R. S. Challenges in developing a pediatric RSV vaccine. Hum. Vaccin. 5, 582–591 (2009).
    1. Connors M. et al.. Cotton rats previously immunized with a chimeric RSV FG glycoprotein develop enhanced pulmonary pathology when infected with RSV, a phenomenon not encountered following immunization with vaccinia--RSV recombinants or RSV. Vaccine 10, 475–484 (1992).
    1. Murphy B. R., Sotnikov A. V., Lawrence L. A., Banks S. M. & Prince G. A. Enhanced pulmonary histopathology is observed in cotton rats immunized with formalin-inactivated respiratory syncytial virus (RSV) or purified F glycoprotein and challenged with RSV 3-6 months after immunization. Vaccine 8, 497–502 (1990).
    1. Wright P. F. et al.. The absence of enhanced disease with wild type respiratory syncytial virus infection occurring after receipt of live, attenuated, respiratory syncytial virus vaccines. Vaccine 25, 7372–7378 (2007).
    1. Collins P. L. & Melero J. A. Progress in understanding and controlling respiratory syncytial virus: still crazy after all these years. Virus Res. 162, 80–99 (2011).
    1. Karron R. A. et al.. A gene deletion that up-regulates viral gene expression yields an attenuated RSV vaccine with improved antibody responses in children. Sci. Transl. Med. 7, 312ra175 (2015).
    1. Meng J., Lee S., Hotard A. L. & Moore M. L. Refining the balance of attenuation and immunogenicity of respiratory syncytial virus by targeted codon deoptimization of virulence genes. MBio 5, e01704–e01714 (2014).
    1. Rostad C. A. et al.. A recombinant respiratory syncytial virus vaccine candidate attenuated by a low-fusion F protein is immunogenic and protective against challenge in cotton rats. J. Virol. 90, 7508–7518 (2016).
    1. McLellan J. S. et al.. Structure of RSV fusion glycoprotein trimer bound to a prefusion-specific neutralizing antibody. Science 340, 1113–1117 (2013).
    1. Ngwuta J. O. et al.. Prefusion F-specific antibodies determine the magnitude of RSV neutralizing activity in human sera. Sci. Transl. Med. 7, 309ra162 (2015).
    1. Krarup A. et al.. A highly stable prefusion RSV F vaccine derived from structural analysis of the fusion mechanism. Nat. Commun. 6, 8143 (2015).
    1. Palomo C. et al.. Influence of respiratory syncytial virus F glycoprotein conformation on induction of protective immune responses. J. Virol. 90, 5485–5498 (2016).
    1. McLellan J. S. et al.. Structure-based design of a fusion glycoprotein vaccine for respiratory syncytial virus. Science 342, 592–598 (2013).
    1. McGinnes Cullen L., Schmidt M. R., Kenward S. A., Woodland R. T. & Morrison T. G. Murine immune responses to virus-like particle-associated pre- and postfusion forms of the respiratory syncytial virus F protein. J. Virol. 89, 6835–6847 (2015).
    1. Liang B. et al.. Enhanced neutralizing antibody response induced by respiratory syncytial virus prefusion F protein expressed by a vaccine candidate. J. Virol. 89, 9499–9510 (2015).
    1. Kiss G. et al.. Structural analysis of respiratory syncytial virus reveals the position of M2-1 between the matrix protein and the ribonucleoprotein complex. J. Virol. 88, 7602–7617 (2014).
    1. Liljeroos L., Krzyzaniak M. A., Helenius A. & Butcher S. J. Architecture of respiratory syncytial virus revealed by electron cryotomography. Proc. Natl Acad. Sci. USA 110, 11133–11138 (2013).
    1. Corti D. et al.. Cross-neutralization of four paramyxoviruses by a human monoclonal antibody. Nature 501, 439–443 (2013).
    1. Hotard A. L. et al.. Identification of residues in the human respiratory syncytial virus fusion protein that modulate fusion activity and pathogenesis. J. Virol. 89, 512–522 (2015).
    1. Moore M. L. et al.. A chimeric A2 strain of respiratory syncytial virus (RSV) with the fusion protein of RSV strain line 19 exhibits enhanced viral load, mucus, and airway dysfunction. J. Virol. 83, 4185–4194 (2009).
    1. Gilman M. S. et al.. Characterization of a prefusion-specific antibody that recognizes a quaternary, cleavage-dependent epitope on the RSV fusion glycoprotein. PLoS Pathog. 11, e1005035 (2015).
    1. Yunus A. S. et al.. Elevated temperature triggers human respiratory syncytial virus F protein six-helix bundle formation. Virology 396, 226–237 (2010).
    1. Ling Z., Tran K. C. & Teng M. N. Human respiratory syncytial virus nonstructural protein NS2 antagonizes the activation of beta interferon transcription by interacting with RIG-I. J. Virol. 83, 3734–3742 (2009).
    1. Spann K. M., Tran K. C. & Collins P. L. Effects of nonstructural proteins NS1 and NS2 of human respiratory syncytial virus on interferon regulatory factor 3, NF-kappaB, and proinflammatory cytokines. J. Virol. 79, 5353–5362 (2005).
    1. Bukreyev A., Whitehead S. S., Murphy B. R. & Collins P. L. Recombinant respiratory syncytial virus from which the entire SH gene has been deleted grows efficiently in cell culture and exhibits site-specific attenuation in the respiratory tract of the mouse. J. Virol. 71, 8973–8982 (1997).
    1. Karron R. A. et al.. Identification of a recombinant live attenuated respiratory syncytial virus vaccine candidate that is highly attenuated in infants. J. Infect. Dis. 191, 1093–1104 (2005).
    1. Whitehead S. S. et al.. Recombinant respiratory syncytial virus bearing a deletion of either the NS2 or SH gene is attenuated in chimpanzees. J. Virol. 73, 3438–3442 (1999).
    1. Karron R. A. et al.. Respiratory syncytial virus (RSV) SH and G proteins are not essential for viral replication in vitro: clinical evaluation and molecular characterization of a cold-passaged, attenuated RSV subgroup B mutant. Proc. Natl Acad. Sci. USA 94, 13961–13966 (1997).
    1. Meng J. et al.. The respiratory syncytial virus attachment glycoprotein contribution to infection depends on the specific fusion protein. J. Virol. 90, 245–253 (2015).
    1. Techaarpornkul S., Barretto N. & Peeples M. E. Functional analysis of recombinant respiratory syncytial virus deletion mutants lacking the small hydrophobic and/or attachment glycoprotein gene. J. Virol. 75, 6825–6834 (2001).
    1. Teng M. N., Whitehead S. S. & Collins P. L. Contribution of the respiratory syncytial virus G glycoprotein and its secreted and membrane-bound forms to virus replication in vitro and in vivo. Virology 289, 283–296 (2001).
    1. Anderson L. J., Bingham P. & Hierholzer J. C. Neutralization of respiratory syncytial virus by individual and mixtures of F and G protein monoclonal antibodies. J. Virol. 62, 4232–4238 (1988).
    1. Polack F. P. et al.. The cysteine-rich region of respiratory syncytial virus attachment protein inhibits innate immunity elicited by the virus and endotoxin. Proc. Natl Acad. Sci. USA 102, 8996–9001 (2005).
    1. Tripp R. A. et al.. CX3C chemokine mimicry by respiratory syncytial virus G glycoprotein. Nat. Immunol. 2, 732–738 (2001).
    1. Bukreyev A. et al.. The secreted form of respiratory syncytial virus G glycoprotein helps the virus evade antibody-mediated restriction of replication by acting as an antigen decoy and through effects on Fc receptor-bearing leukocytes. J. Virol. 82, 12191–12204 (2008).
    1. Johnson T. R., McLellan J. S. & Graham B. S. Respiratory syncytial virus glycoprotein G interacts with DC-SIGN and L-SIGN to activate ERK1 and ERK2. J. Virol. 86, 1339–1347 (2012).
    1. Yi H. et al.. Native immunogold labeling of cell surface proteins and viral glycoproteins for cryo-electron microscopy and cryo-electron tomography applications. J. Histochem. Cytochem. 63, 780–792 (2015).
    1. Mayhew T. M. Mapping the distributions and quantifying the labelling intensities of cell compartments by immunoelectron microscopy: progress towards a coherent set of methods. J. Anat. 219, 647–660 (2011).
    1. Wright P. F. et al.. Growth of respiratory syncytial virus in primary epithelial cells from the human respiratory tract. J. Virol. 79, 8651–8654 (2005).
    1. Hotard A. L. et al.. A stabilized respiratory syncytial virus reverse genetics system amenable to recombination-mediated mutagenesis. Virology 434, 129–136 (2012).
    1. Widjojoatmodjo M. N. et al.. A highly attenuated recombinant human respiratory syncytial virus lacking the G protein induces long-lasting protection in cotton rats. Virol. J. 7, 114 (2010).
    1. Swanson K. A. et al.. A monomeric uncleaved respiratory syncytial virus F antigen retains prefusion-specific neutralizing epitopes. J. Virol. 88, 11802–11810 (2014).
    1. Stokes K. L. et al.. Differential pathogenesis of respiratory syncytial virus clinical isolates in BALB/c mice. J. Virol. 85, 5782–5793 (2011).
    1. Zhang L., Peeples M. E., Boucher R. C., Collins P. L. & Pickles R. J. Respiratory syncytial virus infection of human airway epithelial cells is polarized, specific to ciliated cells, and without obvious cytopathology. J. Virol. 76, 5654–5666 (2002).
    1. Zhao H. et al.. A novel pregnane-type alkaloid from Pachysandra terminalis inhibits methicillin-resistant Staphylococcus aureus in vitro and in vivo. Phytother. Res. 29, 373–380 (2015).
    1. Jin H., Cheng X., Zhou H. Z., Li S. & Seddiqui A. Respiratory syncytial virus that lacks open reading frame 2 of the M2 gene (M2-2) has altered growth characteristics and is attenuated in rodents. J. Virol. 74, 74–82 (2000).
    1. Mastronarde D. N. Dual-axis tomography: an approach with alignment methods that preserve resolution. J. Struct. Biol. 120, 343–352 (1997).
    1. Mastronarde D. N. Automated electron microscope tomography using robust prediction of specimen movements. J. Struct. Biol. 152, 36–51 (2005).
    1. Kremer J. R., Mastronarde D. N. & McIntosh J. R. Computer visualization of three-dimensional image data using IMOD. J. Struct. Biol. 116, 71–76 (1996).
    1. Galaz-Montoya J. G., Flanagan J., Schmid M. F. & Ludtke S. J. Single particle tomography in EMAN2. J. Struct. Biol. 190, 279–290 (2015).
    1. Nicastro D. et al.. The molecular architecture of axonemes revealed by cryoelectron tomography. Science 313, 944–948 (2006).
    1. Frank J. et al.. SPIDER and WEB: processing and visualization of images in 3D electron microscopy and related fields. J. Struct. Biol. 116, 190–199 (1996).
    1. Pettersen E. F. et al.. UCSF Chimera--a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).
    1. McLellan J. S., Yang Y., Graham B. S. & Kwong P. D. Structure of respiratory syncytial virus fusion glycoprotein in the postfusion conformation reveals preservation of neutralizing epitopes. J. Virol. 85, 7788–7796 (2011).
    1. Wang H. et al.. Controlled growth of few-layer hexagonal boron nitride on copper foils using ion beam sputtering deposition. Small 11, 1542–1547 (2015).

Source: PubMed

3
Subskrybuj