Nasal IgA Provides Protection against Human Influenza Challenge in Volunteers with Low Serum Influenza Antibody Titre

Victoria M W Gould, James N Francis, Katie J Anderson, Bertrand Georges, Alethea V Cope, John S Tregoning, Victoria M W Gould, James N Francis, Katie J Anderson, Bertrand Georges, Alethea V Cope, John S Tregoning

Abstract

In spite of there being a number of vaccines, influenza remains a significant global cause of morbidity and mortality. Understanding more about natural and vaccine induced immune protection against influenza infection would help to develop better vaccines. Virus specific IgG is a known correlate of protection, but other factors may help to reduce viral load or disease severity, for example IgA. In the current study we measured influenza specific responses in a controlled human infection model using influenza A/California/2009 (H1N1) as the challenge agent. Volunteers were pre-selected with low haemagglutination inhibition (HAI) titres in order to ensure a higher proportion of infection; this allowed us to explore the role of other immune correlates. In spite of HAI being uniformly low, there were variable levels of H1N1 specific IgG and IgA prior to infection. There was also a range of disease severity in volunteers allowing us to compare whether differences in systemic and local H1N1 specific IgG and IgA prior to infection affected disease outcome. H1N1 specific IgG level before challenge did not correlate with protection, probably due to the pre-screening for individuals with low HAI. However, the length of time infectious virus was recovered from the nose was reduced in patients with higher pre-existing H1N1 influenza specific nasal IgA or serum IgA. Therefore, IgA contributes to protection against influenza and should be targeted in vaccines.

Keywords: Human Infection Challenge study; IgA; influenza; nasal; vaccine.

Figures

FIGURE 1
FIGURE 1
Human influenza infection in clinical volunteers. Patients were inoculated intranasally with 3.5 × 106 TCID50 influenza A/California/2009 (H1N1). Samples were collected daily and assessed for influenza by culture or rtPCR, and disease signs and symptoms. Cultured virus presented as days virus positive (A), influenza RNA rtPCR data presented as area under the curve plotted over the time course of infection (B), signs and symptoms are presented as cumulative total (C). Correlation plots of days virus positive against detectable Influenza RNA (D) or signs and symptoms (E). (A–C) Are presented as binned frequencies. Data presented are combined from two studies, n = 47 volunteers.
FIGURE 2
FIGURE 2
Infection with influenza increases H1 serum IgG antibody responses, but IgG does not correlate with protection. Healthy adult human volunteers were infected intranasally with different doses of H1N1 influenza (low dose: 3.5 × 104 TCID50, Medium dose: 3.5 × 105 TCID50 and high dose: 3.5 × 106 TCID50). H1 specific IgG responses were measured by ELISA in serum on day-1 and day 29 of infection and presented as absolute values (A), fold change based on the infecting dose (B). HAI was assessed at day-1 and day 29 (C) and compared with IgG titre on day 29 (D). IgG ELISA titre was compared to days culture positive (E) and cumulative signs and symptoms (F). (A–D) Represent data from a single study n = 29 volunteers, (E,F) Represent data from two studies combined n = 47.
FIGURE 3
FIGURE 3
Infection with influenza increases H1 serum IgA antibody responses. Healthy adult human volunteers were infected intranasally with different doses of H1N1 influenza. H1 specific IgA responses were measured by ELISA in serum on day-1 and day 29 of infection and presented as absolute values (A) and fold change based on the infecting dose (B). Serum IgA and IgG were compared at day-1 (C) and day 29 (D). Data from a single study n = 29 volunteers.
FIGURE 4
FIGURE 4
H1N1 specific IgA in nasal wash and serum correlate weakly with protection. H1N1 specific IgA and total IgA were measured in the nasal wash samples collected on day-1 prior to infection (A). H1N1 specific IgA responses were compared between nasal wash and serum (B). Days culture positive virus was recoverable compared to serum IgA (C) and nasal IgA (D). Data pooled from two studies, n = 47 volunteers.
FIGURE 5
FIGURE 5
H3N2 specific antibody does not correlate with protection. H3N2 specific IgG (A), HAI (C) and IgA (D) were measured in serum on day-1 and day 29, fold change in IgG response compared between H1N1 and H3N2 IgG (B). Days culture positive virus was recoverable compared to H3N2 specific serum IgA (E) and nasal IgA (F). Data pooled from two studies, n = 47 volunteers.

References

    1. Ambrose C. S., Wu X., Jones T., Mallory R. M. (2012). The role of nasal IgA in children vaccinated with live attenuated influenza vaccine. Vaccine 30 6794–6801. 10.1016/j.vaccine.2012.09.018
    1. Asahi Y., Yoshikawa T., Watanabe I., Iwasaki T., Hasegawa H., Sato Y., et al. (2002). Protection against influenza virus infection in polymeric ig receptor knockout mice immunized intranasally with adjuvant-combined vaccines. J. Immunol. 168 2930–2938. 10.4049/jimmunol.168.6.2930
    1. Barría M. I., Garrido J. L., Stein C., Scher E., Ge Y., Engel S. M., et al. (2013). Localized mucosal response to intranasal live attenuated influenza vaccine in adults. J. Infect. Dis. 207 115–124. 10.1093/infdis/jis641
    1. Belshe R. B., Gruber W. C., Mendelman P. M., Mehta H. B., Mahmood K., Reisinger K., et al. (2000). Correlates of immune protection induced by live, attenuated, cold-adapted, trivalent, intranasal influenza virus vaccine. J. Infect. Dis. 181 1133–1137. 10.1086/315323
    1. Bergin P., Langat R., Omosa-Manyonyi G., Farah B., Ouattara G., Park H., et al. (2016). Assessment of anti-HIV-1 antibodies in oral and nasal compartments of volunteers from three different populations. J. Acquir. Immune Defic. Syndr. 73 130–137. 10.1097/QAI.0000000000001094
    1. Black S., Nicolay U., Vesikari T., Knuf M., Del Giudice G., Della Cioppa G., et al. (2011). Hemagglutination inhibition antibody titers as a correlate of protection for inactivated influenza vaccines in children. Pediatr. Infect. Dis. J. 30 1081–1085. 10.1097/INF.0b013e3182367662
    1. Brown T. A., Murphy B. R., Radl J., Haaijman J. J., Mestecky J. (1985). Subclass distribution and molecular form of immunoglobulin A hemagglutinin antibodies in sera and nasal secretions after experimental secondary infection with influenza A virus in humans. J. Clin. Microbiol. 22 259–264.
    1. Carrat F., Vergu E., Ferguson N. M., Lemaitre M., Cauchemez S., Leach S., et al. (2008). Time lines of infection and disease in human influenza: a review of volunteer challenge studies. Am. J. Epidemiol. 167 775–785. 10.1093/aje/kwm375
    1. Clements M. L., Betts R. F., Tierney E. L., Murphy B. R. (1986). Serum and nasal wash antibodies associated with resistance to experimental challenge with influenza A wild-type virus. J. Clin. Microbiol. 24 157–160.
    1. Clements M. L., Murphy B. R. (1986). Development and persistence of local and systemic antibody responses in adults given live attenuated or inactivated influenza A virus vaccine. J. Clin. Microbiol. 23 66–72.
    1. Everitt A. R., Clare S., Mcdonald J. U., Kane L., Harcourt K., Ahras M., et al. (2013). Defining the range of pathogens susceptible to Ifitm3 restriction using a knockout mouse model. PLoS ONE 8:e80723 10.1371/journal.pone.0080723
    1. Everitt A. R., Clare S., Pertel T., John S. P., Wash R. S., Smith S. E., et al. (2012). IFITM3 restricts the morbidity and mortality associated with influenza. Nature (London) 484 519–523. 10.1038/nature10921
    1. Frise R., Bradley K., Van Doremalen N., Galiano M., Elderfield R. A., Stilwell P., et al. (2016). Contact transmission of influenza virus between ferrets imposes a looser bottleneck than respiratory droplet transmission allowing propagation of antiviral resistance. Sci. Rep. 6:29793 10.1038/srep29793
    1. Grohskopf L. A., Sokolow L. Z., Broder K. R., Olsen S. J., Karron R. A., Jernigan D. B., et al. (2016). Prevention and control of seasonal influenza with vaccines. MMWR Recomm. Rep. 65 1–54. 10.15585/mmwr.rr6505a1
    1. Habibi M. S., Jozwik A., Makris S., Dunning J., Paras A., Devincenzo J. P., et al. (2015). Impaired antibody-mediated protection and defective IgA B-cell memory in experimental infection of adults with respiratory syncytial virus. Am. J. Respir. Crit. Care Med. 191 1040–1049. 10.1164/rccm.201412-2256OC
    1. Hobson D., Curry R. L., Beare A. S., Ward-Gardner A. (1972). The role of serum haemagglutination-inhibiting antibody in protection against challenge infection with influenza A2 and B viruses. J. Hyg. 70 767–777. 10.1017/S0022172400022610
    1. Lambert L., Kinnear E., Mcdonald J. U., Grodeland G., Bogen B., Stubsrud E., et al. (2016). DNA vaccines encoding antigen targeted to MHC Class II induce influenza-specific CD8+ T cell responses, enabling faster resolution of influenza disease. Front. Immunol. 7:321 10.3389/fimmu.2016.00321
    1. Mastelic Gavillet B., Eberhardt C. S., Auderset F., Castellino F., Seubert A., Tregoning J. S., et al. (2015). MF59 Mediates Its B cell adjuvanticity by promoting T follicular helper cells and thus germinal center responses in adult and early life. J. Immunol. 194 4836–4845. 10.4049/jimmunol.1402071
    1. Mazanec M. B., Coudret C. L., Fletcher D. R. (1995). Intracellular neutralization of influenza virus by immunoglobulin A anti-hemagglutinin monoclonal antibodies. J. Virol. 69 1339–1343.
    1. Mbawuike I. N., Pacheco S., Acuna C. L., Switzer K. C., Zhang Y., Harriman G. R. (1999). Mucosal immunity to influenza without IgA: an IgA knockout mouse model. J. Immunol. 162 2530–2537.
    1. Pillai P. S., Molony R. D., Martinod K., Dong H., Pang I. K., Tal M. C., et al. (2016). Mx1 reveals innate pathways to antiviral resistance and lethal influenza disease. Science 352 463–466. 10.1126/science.aaf3926
    1. Piralla A., Daleno C., Pariani E., Conaldi P., Esposito S., Zanetti A., et al. (2013). Virtual quantification of influenza A virus load by real-time RT-PCR. J. Clin. Virol. 56 65–68. 10.1016/j.jcv.2012.09.011
    1. Reber A., Katz J. (2013). Immunological assessment of influenza vaccines and immune correlates of protection. Expert Rev. Vaccines 12 519–536.10.1586/erv.13.35
    1. Renegar K. B., Small P. A., Jr., Boykins L. G., Wright P. F. (2004). Role of IgA versus IgG in the control of influenza viral infection in the murine respiratory tract. J. Immunol. 173 1978–1986. 10.4049/jimmunol.173.3.1978
    1. Rossen R. D., Butler W. T., Waldman R. H., Alford R. H., Hornick R. B., Togo Y., et al. (1970). The proteins in nasal secretion. II. A longitudinal study of IgA and neutralizing antibody levels in nasal washings from men infected with influenza virus. JAMA 211 1157–1161. 10.1001/jama.1970.03170070027005
    1. Russell R. F., Mcdonald J. U., Ivanova M., Zhong Z., Bukreyev A., Tregoning J. S. (2015). Partial attenuation of respiratory syncytial virus with a deletion of a small hydrophobic gene is associated with elevated interleukin-1beta responses. J. Virol. 89 8974–8981. 10.1128/JVI.01070-15
    1. Russell R. F., Mcdonald J. U., Lambert L., Tregoning J. S. (2016). Use of the microparticle NanoSiO2 as an adjuvant to boost vaccine immune responses in neonatal mice against influenza. J. Virol. 90 4735–4744. 10.1128/JVI.03159-15
    1. Seibert C. W., Rahmat S., Krause J. C., Eggink D., Albrecht R. A., Goff P. H., et al. (2013). Recombinant IgA is sufficient to prevent influenza virus transmission in guinea pigs. J. Virol. 87 7793–7804. 10.1128/JVI.00979-13
    1. Sridhar S., Begom S., Bermingham A., Hoschler K., Adamson W., Carman W., et al. (2013). Cellular immune correlates of protection against symptomatic pandemic influenza. Nat. Med. 19 1305–1312. 10.1038/nm.3350
    1. Sridhar S., Brokstad K. A., Cox R. J. (2015). Influenza vaccination strategies: comparing inactivated and live attenuated influenza vaccines. Vaccines (Basel) 3 373–389. 10.3390/vaccines3020373
    1. Tamura S., Funato H., Hirabayashi Y., Suzuki Y., Nagamine T., Aizawa C., et al. (1991). Cross-protection against influenza A virus infection by passively transferred respiratory tract IgA antibodies to different hemagglutinin molecules. Eur. J. Immunol. 21 1337–1344. 10.1002/eji.1830210602
    1. Wagner D. K., Clements M. L., Reimer C. B., Snyder M., Nelson D. L., Murphy B. R. (1987). Analysis of immunoglobulin G antibody responses after administration of live and inactivated influenza A vaccine indicates that nasal wash immunoglobulin G is a transudate from serum. J. Clin. Microbiol. 25 559–562.
    1. Watson J. M., Francis J. N., Mesens S., Faiman G. A., Makin J., Patriarca P., et al. (2015). Characterisation of a wild-type influenza (A/H1N1) virus strain as an experimental challenge agent in humans. Virol. J. 12 13 10.1186/s12985-015-0240-5
    1. Wright P. F., Hoen A. G., Ilyushina N. A., Brown E. P., Ackerman M. E., Wieland-Alter W., et al. (2016). Correlates of immunity to influenza as determined by challenge of children with live, attenuated influenza vaccine. Open Forum Infect. Dis. 3:ofw108 10.1093/ofid/ofw108
    1. Wright P. F., Murphy B. R., Kervina M., Lawrence E. M., Phelan M. A., Karzon D. T. (1983). Secretory immunological response after intranasal inactivated influenza A virus vaccinations: evidence for immunoglobulin A memory. Infect. Immun. 40 1092–1095.

Source: PubMed

3
Subskrybuj