3,967
Views
19
CrossRef citations to date
0
Altmetric
Editorial

Antibody-dependent enhancement and vaccine development

, , &
Pages 409-412 | Published online: 09 Jan 2014

The development of protective antibodies has long been the basis for vaccine development against a large range of infective agents. However, the discovery of the phenomena of antibody-dependent enhancement (ADE) in the 1960s gave rise to the concern that the development of antibodies could, at times, exacerbate the reaction to a natural infection by the microbe. Clinical evidence of exacerbation of disease by previous vaccination, infection or the presence of maternal antibodies is available for some microbes and hypotheses have been made about other agents. The phenomena of ADE must be considered during the development of new vaccines for a range of infective agents to ensure that the vaccines are protective and do not harm the recipients, particularly in light of new insights into the alteration of intracellular signaling pathways post-Fc-γR engagement by a microbe–antibody complex.

It is a long-held view in immunology that antibodies are of the utmost benefit in defending the host from microbial infection. Antibody responses to a pathogen are often measured as an indicator of a properly functioning immune system. The antibody response indicates that the infectious agent is recognized by the adaptive immune response, will be cleared from the host and the immune memory established. Since Jenner vaccinated James Phipps with cowpox in 1796 to protect against smallpox, vaccine strategies have sought to induce antibody responses and produce B-cell memory to protect individuals from future infections by the same microbe, modify the severity of the disease and reduce the transmission of microbes between hosts.

However, there is another perspective on this traditional view that has been around for over 30 years and has gained renewed energy over the past 5 years with a number of fresh insights. ADE has historically been associated with virus infection. Hawkes first described ADE with the arboviruses, Murray Valley encephalitis, West Nile, Japanese encephalitis and Getah viruses in 1964 Citation[1]. He found that viral titers in cell culture were greater if the initial infection was in the presence of virus-specific antibody (usually immunoglobulin [Ig]G) at subneutralizing concentrations. Given that subneutralizing concentrations are a normal phase of antibody responses, there was understandable concern that a suboptimal antibody response may exacerbate infection and this disease, rather than protect the host.

Other viruses have since been found to also exhibit ADE. For example, the more severe responses to Dengue virus infection, Dengue hemorrhagic fever and Dengue shock syndrome were found to be 15–80-times more likely in secondary infections than in primary infections Citation[2]. Furthermore, a prospective study in Thai children found that those with pre-existing antibodies that were capable of enhancing Dengue virus type 2 growth in human monocytes in vitro and were at a greater risk of more severe disease following a second infection with Dengue virus Citation[3].

Heterotypic anti-Dengue-virus antibodies enhance the uptake of virions into monocytic cell lines and primary human monocytes in vitro through an interaction with cell surface Ig receptors Citation[4]. Yang and colleagues found that interferon (IFN)-γ was the major cytokine expressed in Dengue 2 infections in the absence of heterotypic antibodies, but that IFN-γ production was suppressed in the presence of heterotypic antibodies Citation[5]. Chen and colleagues found evidence of decreased IFN-γ levels in more severely affected patients, that is, those with Dengue hemorrhagic fever compared with those with Dengue fever Citation[6]. Furthermore, the authors found that subneutralizing antibodies were able to enhance virus replication, interleukin (IL)-4 levels appeared to be increased in the ADE reactions and there was an association between ADE and an increase in prostaglandin (PG)E2Citation[7]. Later work found that Dengue hemorrhagic fever patients had significantly higher IL-10 levels and a lower Th1 response Citation[6]. Similarly, ADE has also been associated with neurovirulence of the arboviruses, yellow fever and Japanese encephalitis viruses in mice Citation[8] and enhance the replication of tick-borne encephalitis virus in vitroCitation[9]. In vitro studies Citation[10,11] have linked ADE to a variety of HIV strains in different cell types, raising the possibility that antibodies produced in trial subjects in response to HIV vaccines could enhance future natural HIV infections. This concern has led to expert meetings to discuss the challenges posed by suboptimal vaccination responses to HIV vaccines Citation[12].

Early trials with formalin-inactivated respiratory syncytial virus (FI-RSV) found that the vaccines not only did not protect children against RSV but also appeared to increase the attack rate and clinical severity of later natural infection with RSV Citation[13–15], possibly contributing to some deaths Citation[16]. It has been suggested that exacerbation of disease after RSV vaccination and, possibly also in the presence of maternal antibodies, was also due to ADE mechanisms during later natural infections with RSV Citation[13].

Studies with BALB/c mice have been undertaken in order to elucidate the mechanisms of this response to vaccination. A similar enhancement is also seen when mice are sensitized with the RSV-G glycoprotein Citation[16] or vaccinia virus expressing the secreted form of the RSV G glycoprotein Citation[17]. The severe disease that followed challenge with RSV was associated with elevated levels of IL-4, -5, -13 and eotaxin Citation[18]. Interestingly, the secondary response to RSV infection following vaccination with FI-RSV mainly utilizes IL-4-mediated mechanisms Citation[17].

The recent insights into ADE have revealed the crucial role of intracellular signaling as an underpinning molecular mechanism that explains the high growth of progeny virus in ADE-infected cells postinfection. The signaling impact of ADE is remarkable in that it has been found to be highly specific for key defense pathways, allowing an early and temporary suppression of IFN, tumor necrosis factor (TNF) and inducible nitric oxide synthase (NOS2) expression, leading to a cellular environment less threatening to virus survival. This is a seminal development in the understanding of ADE. The traditional view is that enhanced virus growth post-ADE infection is a function of the enhanced uptake of the virus through the Fc-receptors (or in some cases, complement receptors); the observations on post-ADE infection intracellular signaling show another dimension, namely the specific suppression of antiviral protein expression. Work on Ross River virus (RRV) infection in macrophages linked the suppression of IFN, TNF and NOS2 to the disruption of signal transducer and activation of transcription (STAT) complexes, immunoregulatory factor (IRF)-1 and necrosis factor (NF)-κB Citation[19,20].

The modulation of cell signaling processes post-infection has also been observed for the intracellular parasite Leishmania. The primary hosts – vertebrates – are infected by sandflies. The parasite causes disease that can manifest in the skin, nasal–oral membranes or the spleen, liver or bone-marrow Citation[21]. Leishmania cells have two morphologies depending on whether they exist in the vertebrate host or the sandfly vector. The vertebrate form has no flagellum (amastigote) and the sandfly form has an anterior flagellum (promastigote). The promastigote is introduced into the vertebrate during sandfly feeding Citation[22] and then enters phagocytic cells, predominantly macrophages, through the fibronectin receptor, the mannose-fucose receptor and the complement receptors (CR)1 and 3 Citation[23] and transforms to the amastigote. These then replicate inside the phagolysosome of the cell, are able to infect neighboring macrophages Citation[24] and are responsible for sustaining infection Citation[25].

Kima and colleagues demonstrated that maintenance of Leishmania infections was impaired in the absence of antibody, thus indicting a role for ADE in the pathogenesis of leishmaniases Citation[23]. Amastigotes with host-derived IgG on their surface and these IgG-opsonized amastigotes have increased virulence. Miles and colleagues demonstrated, in humans and mice, that infection with Leishmania major in the presence of IgG-immune complexes results in an inability to resolve infection in the host Citation[24]. Kane and Mosser proposed that IgG forms an immune complex with the amastigote, which then ligates to Fcγ receptors to induce IL-10 production in macrophages and, in turn, inhibits macrophage activation and increases parasite growth Citation[22]. This is supported by the findings of Kima and colleagues that the Fcγ receptor is required for amastigote parasite entry Citation[23]. Increased IL-10 has been associated with visceral and cutaneous leishmanias and IL-10 has been flagged as having an important role in the regulation of the immune response to this intracellular parasite Citation[22] and links ADE with the Th1/Th2 immune response. Uzonna and colleagues showed that pathophysiological states are associated with characteristic Th1 or Th2 immune responses and that mice infected with subclinical infections that maintained a Th1 immune response were resistant to a higher infectious challenge Citation[26]. A Th2 immune response and the production of IL-4 and -10, increase disease persistence by antagonizing the capacity of macrophages to produce NOS2 and consequently generate NO Citation[25]. Gantt and colleagues propose that NO and superoxide contribute to the intracellular eradication of Leishmania chagasi in both human and mouse models Citation[27]. In light of these studies, an antibody-based vaccine may not be effective in controlling leishmanias and may, in fact, contribute to disease through the development of immune complexes. Furthermore, as suggested by Uzonna and colleagues, clearance of the parasite may lead to loss of immunological memory and therefore loss of resistance Citation[26]. Vaccination leading to clearance of the parasite may require continual and frequent exposure to leishmanias antigens to maintain the Th1 response and immunity.

A critical feature of this observation was the parasite-associated enhancement of IL-10 expression, a finding that supported earlier observations with the RRV-macrophage model of ADE Citation[20]. The RRV study proposed that the role of pathogen-mediated stimulation of IL-10 was to globally downregulate the expression of early inflammatory and antiviral Th1 cytokines, such as TNF Citation[20]. Increased levels of IL-10 have also been observed for Ebola, Junin, Lassa virus and Yellow fever virus infections Citation[28].

Post-ADE alterations to cell signaling are not simply a matter of neutralizing key cellular defense proteins, but also for potentiating the expression of other host genes that assist the survival of the microbe. So far, the pathogen-stimulated alteration of cellular signaling pathways are connected with intracellular microbes, which makes biological sense given the microbe lifecycle.

An ADE mechanism for bacteria has also been observed but the mechanism involved enhanced bacterial adherence to mucosal surfaces. Streptococcus pneumoniae is a Gram-positive coccus. Although a commensal organism of the nasopharynx, it is also a major opportunistic human pathogen Citation[29]. A major function of IgA is to stop bacterial adhesion and, therefore, colonization of host mucosal surfaces Citation[30]. A recent paper by Weiser and colleagues suggests that the specific antibacterial antibody, IgA1, the major form of IgA in the upper-respiratory tract, was cleaved by pneumoccocal IgA1 protease, resulting in a survival advantage for the bacteria Citation[31]. The protease produced Fab (antigen-binding) antibody fragments, the variable regions of which have an ionic charge, increasing adherence to host epithelial cells by the IgA(Fab) complex. This antibody-mediated mechanism may allow pneumococcus to persist on mucosal surfaces in the respiratory tract for extended periods.

In terms of vaccination strategies, the role of ADE and the subsequent downstream impact on signaling inside the infected cells has a much broader consequence beyond the early, enhanced growth of the microbe. What impact the early alteration of innate immune protein expression has on future adaptive responses in terms of microbe/parasite clearance and the formation of immunological memory, is yet to be fully understood.

Early vaccination strategies for some microbes have been successful and have grown to produce highly significant results, for example, Jenner’s original work with cowpox eventually led to the eradication of smallpox from the planet. However, many vaccine trials have been unsuccessful in producing the desired immunity and some have actually harmed participants. Although many vaccine failures are owing to a lack of antibody production or nonprotective antibodies being produced, ADE raises concerns that subneutralizing antibody production may enhance subsequent natural infections and increase disease severity. Vaccines are a routine medical intervention performed on healthy individuals, so ADE needs to be considered seriously during vaccine development to ensure that vaccines protect individuals and do not exacerbate disease following subsequent infections.

References

  • Hawkes RA. Enhancement of the infectivity of arboviruses by specific antisera produced in domestic fowls. Aust. J. Exp. Biol. Med. Sci.42, 465–482 (1964).
  • Halstead SB. Immune enhancement of viral infection. Prog. Allergy31, 301–364 (1982).
  • Kliks SC, Nisalak A, Brandt WE, Wahl L, Burke DS. Antibody-dependent enhancement of Dengue virus growth in human monocytes as a risk factor for Dengue hemorrhagic fever. Am. J. Trop. Med. Hyg.40, 444–451(1989).
  • Morens DM, Halstead SB. Measurement of antibody-dependent infection enhancement of four Dengue virus serotypes by monoclonal and polyclonal antibodies. J. Gen. Virol.71, 2909–2914 (1990).
  • Yang KD, Yeh W-T, Yang M-Y, Chen R-F, Shaio M-F. Antibody-dependent enhancement of heterotypic Dengue infections involved in suppression of IFNγ production. J. Med. Virol.63, 150–157 (2001).
  • Chen R-F, Liu J-W, Yeh W-T et al. Altered T helper 1 reaction but not increase of virus load in patients with Dengue hemorrhagic fever. FEMS Immunol. Med. Micro.44, 43–50 (2005).
  • Chen R, Yeh W, Yang M-Y, Yang KD. A model of the real-time correlation of viral titers with immune reactions in antibody-dependent enhancement of Dengue-2 infections. FEMS Immunol. Med. Micro.30, 1–7 (2001).
  • Gould EA, Buckley A. Antibody-dependent enhancement of yellow fever and Japanese encephalitis virus neurovirulence. J. Gen. Virol.70, 1605–1608 (1989).
  • Phillpotts RJ, Stephenson JR, Porterfield JS. Antibody-dependent enhancement of tick-borne encephalitis virus infectivity. J. Gen. Virol.66, 1831–1837 (1985).
  • Fust G. Enhancing antibodies in HIV infection. Parasitology115, S127–S140 (1997).
  • Homsy J, Meyer M, Levy JA. Serum enhancement of human immunodeficiency virus (HIV) infection correlates with disease in HIV-infected individuals. J. Virol.64, 1437–1440 (1990).
  • Mascola JR, Mathieson BJ, Zack PM, Walker MC, Halstead BS, Burke DS. Summary report: workshop on the potential risks of antibody-dependent enhancement in human HIV vaccine trials. AIDS Res. Hum. Retroviruses9, 1175–1184 (1993).
  • Chin J, Magoffin RL, Shearer LA, Schieble JH, Lennette EH Field evaluation of a respiratory syncytial virus vaccine and a trivalent parainfluenza virus vaccine in a pediatric population. Am. J. Epidemiol.89, 449–463 (1969).
  • Fulginiti VA, Eller JJ, Sieber OF, Joyner JW, Minamitani M, Meiklejohn G. Respiratory virus immunization. I. A field trial of two inactivated respiratory virus vaccines; an aqueous trivalent parainfluenza virus vaccine and an alum-precipitated respiratory syncytial virus vaccine. Am. J. Epidemiol.89(4), 435–448 (1969).
  • Kim HW, Canchola JG, Brandt CD et al. Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine. Am. J. Epidemiol.89, 422–434 (1969).
  • Haynes LM, Jones LP, Barskey A, Anderson LJ, Tripp RA. Enhanced disease and pulmonary eosinophilia associated with formalin-inactivated respiratory syncytial virus vaccination are linked to G glycoprotein CX3C–CX3CR1 interaction and expression of substance P. J. Virol.77, 9831–9844 (2003).
  • Johnson TR, Graham BS. Secreted respiratory syncytial virus G glycoprotein induces interleukin-5 (IL-5), IL-13, and eosinophilia by an IL-4-independent mechanism. J. Virol.73, 8485–8495 (1999).
  • Johnson TR, Johnson JE, Roberts SR, Wertz GW, Parker RA, Graham BS. Priming with secreted glycoprotein G of respiratory syncytial virus (RSV) augments interleukin-5 production and tissue eosinophilia after RSV challenge. J. Virol.72, 2871–2880 (1998).
  • Lidbury BA, Mahalingam S. Specific ablation of antiviral gene expression in macrophages by antibody-dependent enhancement of Ross River virus infection. J. Virol.74, 8376–8381 (2000).
  • Mahalingam S, Lidbury BA. Suppression of lipopolysaccharide-induced antiviral transcription factor (STAT-1 and NF-κB) complexes by antibody-dependent enhancement of macrophage infection by Ross River virus. Proc. Natl Acad. Sci. USA99, 13819–13824 (2002).
  • Berman J. Current treatment approaches to leishmaniasis. Curr. Opin. Infect. Dis.16, 397–401 (2003).
  • Kane MM, Mosser DM. The role of IL-10 in promoting disease progression in leishmaniasis. J. Immunol.166, 1141–1147 (2001).
  • Kima PE, Constant SL, Hannum L et al. Internalization of Leishmania mexicana complex amastigotes via the Fc receptor is required to sustatin infection in murine cutaneous leishmaniasis. J. Exp. Med.191, 1063–1067 (2000).
  • Miles SA, Conrad SM, Alves RG, Jeronimo SMB, Mosser DM. A role for IgG immune complexes during infection with the intracellular pathogen Leishmania.J. Exp. Med.201, 747–754 (2005).
  • Melby PC. Recent developments in leishmaniasis. Curr. Opin. Infect. Dis.15, 485–490 (2002).
  • Uzonna JE, Wei G, Yurkowski D, Bretscher P. Immune elimination of Leishmania major in mice: implications for immune memory, vaccination, and reactivation disease. J. Immunol.167, 6967–6974 (2001).
  • Gantt KR, Goldman TL, McCormick ML et al. Oxidative responses of human and murine macrophages during phagocytosis of Leishmania chagasi. J. Immunol.167, 893–901 (2001).
  • Geisbert TW, Jahrling PB Exotic emerging viral diseases: progress and challenges. Nat. Med.10, S110–S121 (2004).
  • Jedrzejas MJ. Unveiling molecular mechanisms of pneumococcal surface protein A interactions with antibodies and lactoferrin. Clin. Chim. Acta367, 1–10 (2006).
  • Kilian M, Reinholdt J, Lomholt H, Poulsen K, Frandsen EV. Biological significance of IgA1 proteases in bacterial coloization and pathogenesis: critical evaluation of experimental evidence. APMIS104, 321–338 (1996).
  • Weiser JN, Bae D, Fasching C, Scamurra RW, Ratner AJ, Janoff EN. Antibody-enhanced pneumococcal adherence requires IgA1 protease. Proc. Natl Acad. Sci. USA100, 4215–4220 (2003).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.