Cross-reactive antibodies enhance live attenuated virus infection for increased immunogenicity

Kuan Rong Chan, Xiaohui Wang, Wilfried A A Saron, Esther Shuyi Gan, Hwee Cheng Tan, Darren Z L Mok, Summer Li-Xin Zhang, Yie Hou Lee, Cui Liang, Limin Wijaya, Sujoy Ghosh, Yin Bun Cheung, Steven R Tannenbaum, Soman N Abraham, Ashley L St John, Jenny G H Low, Eng Eong Ooi, Kuan Rong Chan, Xiaohui Wang, Wilfried A A Saron, Esther Shuyi Gan, Hwee Cheng Tan, Darren Z L Mok, Summer Li-Xin Zhang, Yie Hou Lee, Cui Liang, Limin Wijaya, Sujoy Ghosh, Yin Bun Cheung, Steven R Tannenbaum, Soman N Abraham, Ashley L St John, Jenny G H Low, Eng Eong Ooi

Abstract

Vaccination has achieved remarkable successes in the control of childhood viral diseases. To control emerging infections, however, vaccines will need to be delivered to older individuals who, unlike infants, probably have had prior infection or vaccination with related viruses and thus have cross-reactive antibodies against the vaccines. Whether and how these cross-reactive antibodies impact live attenuated vaccination efficacy is unclear. Using an open-label randomized trial design, we show that subjects with a specific range of cross-reactive antibody titres from a prior inactivated Japanese encephalitis vaccination enhanced yellow fever (YF) immunogenicity upon YF vaccination. Enhancing titres of cross-reactive antibodies prolonged YF vaccine viraemia, provoked greater pro-inflammatory responses, and induced adhesion molecules intrinsic to the activating Fc-receptor signalling pathway, namely immune semaphorins, facilitating immune cell interactions and trafficking. Our findings clinically demonstrate antibody-enhanced infection and suggest that vaccine efficacy could be improved by exploiting cross-reactive antibodies.

Conflict of interest statement

The authors declare no competing financial interests.

Figures

Figure 1. Clinical trial overview.
Figure 1. Clinical trial overview.
a, Consort diagram showing the flow of participants through each stage of the randomized trial. b, Diagrammatic representation of the study. Subjects in the treatment group were vaccinated with Ixiaro followed by Stamaril at 1 month (Group 1, red), 4 months (Group 2, blue) or 9 months (Group 3, cyan) apart, indicated by §. The control group (Group 4) received YF vaccination only. c, Anti-JE antibody titres of subjects from Group 1 (n = 23), Group 2 (n = 11) and Group 3 (n = 14) before YF vaccination as measured by PRNT. *P < 0.05, **P < 0.01 (unpaired t-test).
Figure 2. Cross-reactive antibodies enhance YF vaccine…
Figure 2. Cross-reactive antibodies enhance YF vaccine infection to produce prolonged viraemia and improved immunogenicity.
a,b, Correlation between anti-JE (day 0) (a) and anti-JE (1 month) (b) versus anti-YF (1 month) specific antibody titres for Groups 1–3 (n = 34) as measured by PRNT. Dotted lines indicate the 99% confidence interval of the mean titre of Group 4 and the enhancing group, defined as subjects with titre greater than the 99% confidence interval of Group 4. c,d, Pre-YF immune sera from the treatment group (Groups 1–3; n = 48) (c) or the control group (n = 20) (d) were diluted 1:10 and incubated with YF17D before infecting THP-1 monocytes. YF17D plaque titres, represented by pre-vaccination serum mediated YF17D infection, were correlated with the log-transformed YF antibody titres (1 month). eg, YF RNA levels at days 1, 3 and 7 after YF vaccination for Group 4 subjects (n = 20) (e) and the non-enhancing group (n = 40) (f) and enhancing group (n = 8) (g). h,j, YF RNA levels across the different groups at day 3 (h) and day 7 (j) post-YF vaccination. Error bars indicate standard error of the mean. i,k, Correlation of log-transformed YF antibody titres (1 month) with log-transformed YF RNA levels observed on day 3 (i) and day 7 (k) (n = 48). *P < 0.05, **P < 0.01 (paired t-test for eg, unpaired t-test for h,j).
Figure 3. Cross-reactive antibodies differentially regulate the…
Figure 3. Cross-reactive antibodies differentially regulate the production of inflammatory lipids and metabolites during YF vaccination.
a,c,e, Serum levels of arachidonic acid (a), linoleic acid (c) and 12-HETE (e) in Group 4 (black bars) (n = 7), the non-enhancing group (blue bars) (n = 16) and the enhancing group (red bars) (n = 7) at days 0, 1, 3 and 7 post-YF vaccination. Box plot whiskers indicate the range of the data. b,d,f, Pearson correlation (r) of anti-YF antibody titres (1 month), as measured by PRNT, with log2-transformed values of fold changes (day 7 versus day 0) of arachidonic acid (b), linoleic acid (d) and 12-HETE (f) (n = 23). *P < 0.05, **P < 0.01, ***P < 0.001 (paired t-test).
Figure 4. Cross-reactive antibodies upregulate immune semaphorins…
Figure 4. Cross-reactive antibodies upregulate immune semaphorins via activating FcγR ligation.
a, GSEA analysis of the enriched signalling pathways in enhancing group (n = 6) compared with subjects from non-enhancing and control groups (n = 23) at day 3 post YF-vaccination. Normalized enrichment scores were computed by GSEA and these pathways have gene-set nominal P values < 0.05 (n = 21). b, GSEA analysis of enriched signalling pathways at day 3 post YF-vaccination that correlate with YF antibody titres for subjects with prior JE vaccination (black bars) (n = 21). White bars indicate corresponding enrichment scores for the control group (n = 7). Nominal gene-set P values were <0.05 for black bars and >0.05 for white bars. PLC, phospholipase C. SHC, Src homology 2 containing transforming protein. c, Immune semaphorin genes (in red) identified by GSEA to be enriched in subjects from the enhancing group. d, Pearson correlation (r) of YF antibody titres, as measured by PRNT, with log2-transformed values of fold changes (day 3 versus day 0) of SEMA4A, SEMA6A, SEMA7A and ITGA1 (n = 25). P values indicate significance of the slope. e, Localization and expression of SEMA4A (green) in THP-1 exposed to media, h4G2, YF17D or h4G2-opsonized YF17D (ADE) for 24 h. DAPI staining is blue. Values in white indicate the corrected total cell fluorescence of SEMA4A, normalized to DAPI. Scale bars, 20 µm. f, SEMA4A protein expression levels in primary monocytes exposed to media, YF17D or YF17D opsonized with pre-YF vaccination serum (1:10) from an enhancing group subject for 24 h. Relative expression is normalized to GAPDH. g, SEMA4A expression when YF17D opsonized with pre-vaccination serum from an enhancing group subject is added to primary monocytes blocked with either isotype antibodies or anti-FcγRI and FcγRII antibodies. h, SEMA4A protein expression in THP-1 when FcγRI, FcγRII or both receptors are ligated for 6 h. ik, Quantitative PCR analysis of SEMA4A and SEMA7A in primary monocytes (i) and dendritic cells (j,k) when FcγRI, FcγRII or both receptors are ligated for 6 h (n = 4). Values were corrected for expression of the control gene (GAPDH). *P < 0.05, **P < 0.01 (unpaired t-test). Error bars indicate standard deviation of the data.
Figure 5. Cross-reactive antibodies increase DC activation,…
Figure 5. Cross-reactive antibodies increase DC activation, migration and semaphorin expression for improved antigen presentation during YF vaccination.
a, Diagrammatic representation showing how cross-reactive antibodies from mice immunized with Ixiaro were transferred to non-immunized mice to study their effects on YF vaccination. b,c, Total (b, CD11c+) and activated DCs (c, CD11c+CD80+CD86+) in DLNs after YF vaccination with pretreatment of different doses of cross-reactive JE IgG (n = 6). df, Percentage of SEMA4A+ DCs (d), representative histograms (from n = 6) showing SEMA4A staining on DCs (e) and mean fluorescence intensity (MFI) of SEMA4A+ staining on DCs (f) with pretreatment of different doses of cross-reactive JE IgG (n = 6). g, SEMA4A MFI of LN cells (n = 6). *P < 0.05, **P < 0.01 (unpaired t-test). Error bars indicate standard deviation of the data.

References

    1. Pike BL. The origin and prevention of pandemics. Clin. Infect. Dis. 2010;50:1636–1640. doi: 10.1086/652860.
    1. Fauci AS, Morens DM. Zika virus in the Americas—yet another arbovirus threat. N. Engl. J. Med. 2016;374:601–604. doi: 10.1056/NEJMp1600297.
    1. WHO Ebola Response Team. Ebola virus disease in West Africa—the first 9 months of the epidemic and forward projections. N. Engl. J. Med. 2014;371:1481–1495. doi: 10.1056/NEJMoa1411100.
    1. D'Argenio DA, Wilson CB. A decade of vaccines: integrating immunology and vaccinology for rational vaccine design. Immunity. 2010;33:437–440. doi: 10.1016/j.immuni.2010.10.011.
    1. Rappuoli R, Mandl CW, Black S, De Gregorio E. Vaccines for the twenty-first century society. Nat. Rev. Immunol. 2011;11:865–872. doi: 10.1038/nri3085.
    1. Henao-Restrepo AM. Efficacy and effectiveness of an rVSV-vectored vaccine expressing Ebola surface glycoprotein: interim results from the Guinea ring vaccination cluster-randomised trial. Lancet. 2015;386:857–866. doi: 10.1016/S0140-6736(15)61117-5.
    1. Buchbinder SP. Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-of-concept trial. Lancet. 2008;372:1881–1893. doi: 10.1016/S0140-6736(08)61591-3.
    1. Davenport FM, Hennessy AV, Francis T., Jr Epidemiologic and immunologic significance of age distribution of antibody to antigenic variants of influenza virus. J. Exp. Med. 1953;98:641–656. doi: 10.1084/jem.98.6.641.
    1. Hadinegoro SR. Efficacy and long-term safety of a dengue vaccine in regions of endemic disease. N. Engl. J. Med. 2015;373:1195–1206. doi: 10.1056/NEJMoa1506223.
    1. Villar L. Efficacy of a tetravalent dengue vaccine in children in Latin America. N. Engl. J. Med. 2015;372:113–123. doi: 10.1056/NEJMoa1411037.
    1. Capeding MR. Clinical efficacy and safety of a novel tetravalent dengue vaccine in healthy children in Asia: a phase 3, randomised, observer-masked, placebo-controlled trial. Lancet. 2014;384:1358–1365. doi: 10.1016/S0140-6736(14)61060-6.
    1. Halstead SB, Mahalingam S, Marovich MA, Ubol S, Mosser DM. Intrinsic antibody-dependent enhancement of microbial infection in macrophages: disease regulation by immune complexes. Lancet Infect. Dis. 2010;10:712–722. doi: 10.1016/S1473-3099(10)70166-3.
    1. Chan KR. Leukocyte immunoglobulin-like receptor B1 is critical for antibody-dependent dengue. Proc. Natl Acad. Sci. USA. 2014;111:2722–2727. doi: 10.1073/pnas.1317454111.
    1. Boonnak K, Dambach KM, Donofrio GC, Tassaneetrithep B, Marovich MA. Cell type specificity and host genetic polymorphisms influence antibody-dependent enhancement of dengue virus infection. J. Virol. 2011;85:1671–1683. doi: 10.1128/JVI.00220-10.
    1. Campi-Azevedo AC. Subdoses of 17DD yellow fever vaccine elicit equivalent virological/immunological kinetics timeline. BMC Infect. Dis. 2014;14:391. doi: 10.1186/1471-2334-14-391.
    1. Subramanian A. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102.
    1. Gaucher D. Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses. J. Exp. Med. 2008;205:3119–3131. doi: 10.1084/jem.20082292.
    1. Querec TD. Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat. Immunol. 2009;10:116–125. doi: 10.1038/ni.1688.
    1. McKinney EF, Lee JC, Jayne DR, Lyons PA, Smith KG. T-cell exhaustion, co-stimulation and clinical outcome in autoimmunity and infection. Nature. 2015;523:612–616. doi: 10.1038/nature14468.
    1. Acuto O, Michel F. CD28-mediated co-stimulation: a quantitative support for TCR signalling. Nat. Rev. Immunol. 2003;3:939–951. doi: 10.1038/nri1248.
    1. Boyman O, Sprent J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat. Rev. Immunol. 2012;12:180–190. doi: 10.1038/nri3156.
    1. Suzuki K, Kumanogoh A, Kikutani H. Semaphorins and their receptors in immune cell interactions. Nat. Immunol. 2008;9:17–23. doi: 10.1038/ni1553.
    1. Takamatsu H, Kumanogoh A. Diverse roles for semaphorin-plexin signaling in the immune system. Trends Immunol. 2012;33:127–135. doi: 10.1016/j.it.2012.01.008.
    1. Kolodkin AL, Matthes DJ, Goodman CS. The semaphorin genes encode a family of transmembrane and secreted growth cone guidance molecules. Cell. 1993;75:1389–1399. doi: 10.1016/0092-8674(93)90625-Z.
    1. Kumanogoh A. Class IV semaphorin Sema4A enhances T-cell activation and interacts with Tim-2. Nature. 2002;419:629–633. doi: 10.1038/nature01037.
    1. Suzuki K. Semaphorin 7A initiates T-cell-mediated inflammatory responses through α1β1 integrin. Nature. 2007;446:680–684. doi: 10.1038/nature05652.
    1. van Rijn A. Semaphorin 7A promotes chemokine-driven dendritic cell migration. J. Immunol. 2016;196:459–468. doi: 10.4049/jimmunol.1403096.
    1. Daynes RA, Dowell T, Araneo BA. Platelet-derived growth factor is a potent biologic response modifier of T cells. J. Exp. Med. 1991;174:1323–1333. doi: 10.1084/jem.174.6.1323.
    1. Gavalas NG. VEGF directly suppresses activation of T cells from ascites secondary to ovarian cancer via VEGF receptor type 2. Br. J. Cancer. 2012;107:1869–1875. doi: 10.1038/bjc.2012.468.
    1. Ziogas AC. VEGF directly suppresses activation of T cells from ovarian cancer patients and healthy individuals via VEGF receptor Type 2. Int. J. Cancer. 2012;130:857–864. doi: 10.1002/ijc.26094.
    1. Liu Y. Regulated expression of FcgammaR in human dendritic cells controls cross-presentation of antigen-antibody complexes. J. Immunol. 2006;177:8440–8447. doi: 10.4049/jimmunol.177.12.8440.
    1. Guzman MG. Dengue hemorrhagic fever in Cuba, 1981: a retrospective seroepidemiologic study. Am. J. Trop. Med. Hyg. 1990;42:179–184. doi: 10.4269/ajtmh.1990.42.179.
    1. de Alwis R. Dengue viruses are enhanced by distinct populations of serotype cross-reactive antibodies in human immune sera. PLoS Pathogens. 2014;10:e1004386. doi: 10.1371/journal.ppat.1004386.
    1. Halstead SB, O'Rourke EJ. Dengue viruses and mononuclear phagocytes. I. Infection enhancement by non-neutralizing antibody. J. Exp. Med. 1977;146:201–217. doi: 10.1084/jem.146.1.201.
    1. Olkowski S. Reduced risk of disease during postsecondary dengue virus infections. J. Infect. Dis. 2013;208:1026–1033. doi: 10.1093/infdis/jit273.
    1. Thanachartwet V. Identification of clinical factors associated with severe dengue among Thai adults: a prospective study. BMC Infect. Dis. 2015;15:420. doi: 10.1186/s12879-015-1150-2.
    1. Ben Mkaddem S. Shifting FcγRIIA-ITAM from activation to inhibitory conFig.uration ameliorates arthritis. J. Clin. Invest. 2014;124:3945–3959. doi: 10.1172/JCI74572.
    1. Aloulou M. IgG1 and IVIg induce inhibitory ITAM signaling through FcγRIII controlling inflammatory responses. Blood. 2012;119:3084–3096. doi: 10.1182/blood-2011-08-376046.
    1. Chan KR. Ligation of Fc γ receptor IIB inhibits antibody-dependent enhancement of dengue virus infection. Proc. Natl Acad. Sci. USA. 2011;108:12479–12484. doi: 10.1073/pnas.1106568108.
    1. Lennartz MR, Brown EJ. Arachidonic acid is essential for IgG Fc receptor-mediated phagocytosis by human monocytes. J. Immunol. 1991;147:621–626.
    1. Stachowska E. Conjugated linoleic acids can change phagocytosis of human monocytes/macrophages by reduction in Cox-2 expression. Lipids. 2007;42:707–716. doi: 10.1007/s11745-007-3072-2.
    1. Middleton MK, Rubinstein T, Pure E. Cellular and molecular mechanisms of the selective regulation of IL-12 production by 12/15-lipoxygenase. J. Immunol. 2006;176:265–274. doi: 10.4049/jimmunol.176.1.265.
    1. Calder PC, Grimble RF. Polyunsaturated fatty acids, inflammation and immunity. Eur. J. Clin. Nutr. 2002;56:S14–S19. doi: 10.1038/sj.ejcn.1601478.
    1. Pulendran B. Systems vaccinology: probing humanity's diverse immune systems with vaccines. Proc. Natl Acad. Sci. USA. 2014;111:12300–12306. doi: 10.1073/pnas.1400476111.
    1. Casanova JL, Abel L. The genetic theory of infectious diseases: a brief history and selected illustrations. Annu. Rev. Genom. Hum. Genet. 2013;14:215–243. doi: 10.1146/annurev-genom-091212-153448.
    1. Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI. Human nutrition, the gut microbiome and the immune system. Nature. 2011;474:327–336. doi: 10.1038/nature10213.
    1. Goh KC. Molecular determinants of plaque size as an indicator of dengue virus attenuation. Sci. Rep. 2016;6:26100. doi: 10.1038/srep26100.
    1. Jafari H. Polio eradication. Efficacy of inactivated poliovirus vaccine in India. Science. 2014;345:922–925. doi: 10.1126/science.1255006.
    1. Bass JW. Booster vaccination with further live attenuated measles vaccine. JAMA. 1976;235:31–34. doi: 10.1001/jama.1976.03260270017018.
    1. Low JG. The role of pre-existing cross-reactive antibodies in determining the efficacy of vaccination in humans: study protocol for a randomized controlled trial. Trials. 2015;16:147. doi: 10.1186/s13063-015-0651-z.
    1. Domingo C. Advanced yellow fever virus genome detection in point-of-care facilities and reference laboratories. J. Clin. Microbiol. 2012;50:4054–4060. doi: 10.1128/JCM.01799-12.
    1. Cui L. Serum metabolome and lipidome changes in adult patients with primary dengue infection. PLoS Negl. Trop. Dis. 2013;7:e2373. doi: 10.1371/journal.pntd.0002373.
    1. Hanson BJ. Passive immunoprophylaxis and therapy with humanized monoclonal antibody specific for influenza A H5 hemagglutinin in mice. Respir. Res. 2006;7:126. doi: 10.1186/1465-9921-7-126.

Source: PubMed

3
Subscribe