Elsevier

Vaccine

Volume 27, Issue 15, 26 March 2009, Pages 2171-2176
Vaccine

Cell-mediated immunity elicited by the blood stage malaria vaccine apical membrane antigen 1 in Malian adults: Results of a Phase I randomized trial

https://doi.org/10.1016/j.vaccine.2009.01.097Get rights and content

Abstract

The development of a safe and effective malaria vaccine is impeded by the complexity of the Plasmodium life cycle. A vaccine that elicits both cell-mediated and humoral immune responses might be needed for protection against this multistage parasitic infection. Apical membrane antigen 1 (AMA-1) plays a key role in erythrocytic invasion but is also expressed in sporozoites and in late stage liver schizonts, where it may provide a target of protective cell-mediated immunity (CMI). A Phase 1 trial of a vaccine consisting of recombinant AMA-1 protein and the Adjuvant system AS02A was conducted in 60 Malian adults aged 18–55 years who were randomized to receive either half-dose (25 μg/0.25 ml) or full dose (50 μg/0.5 ml) FMP2.1/AS02A or a control rabies vaccine. Interleukin 5 (IL-5) and interferon-γ (IFN-γ) production as evaluated by ELISpot and lymphocyte proliferation were measured after in vitro AMA-1 stimulation of peripheral blood mononuclear cells (PBMCs) collected on Days 0 and 90. Post-FMP2.1/AS02A immunization mean stimulation indices were significantly elevated as were the number of IL-5 spot forming cells (SFC)/106 PBMC, but no difference was noted in INF-γ production between the AMA-1/AS02A vaccinated group and the rabies group. These results provide evidence that complex immune responses can be induced by this vaccination strategy and add further impetus for the continuing clinical evaluation of this vaccine.

Introduction

Apical membrane antigen 1 (AMA-1) is an asexual blood-stage antigen that has been identified in all Plasmodium species examined and is a leading malaria vaccine candidate [1], [2], [3]. Although its precise biological function remains undefined, AMA-1 appears to play a role in erythrocytic invasion possibly by orienting merozoites and mediating close junctional apical contact [4], [5] or direct erythrocytic binding [6]. The protein is comprised of an ectodomain in which non-overlapping groups of disulfide bonds define four separate domains (pro-domain and domains I, II, and III) [7]. Correct conformational folding is critical for the invocation of protective or inhibitory antibodies, implying that protection is directed at epitopes dependent upon correct disulfide bonding [7], [8], [9].

Antibodies against Plasmodium falciparum AMA-1 (PfAMA-1) inhibit antigen processing and merozoite invasion of erythrocytes [10] as well as P. falciparum growth in vitro [11], [12]. Passive immunization of P. chabaudi-infected mice with AMA-1-specific polyclonal antibodies prevents lethal parasitemia [8]. Protective immunity against malaria has been elicited after immunization of mice and monkeys with recombinant AMA-1 [8], [9], [13], [14], [15]. Seroprevalence studies have demonstrated the presence of antibodies to AMA-1 in malaria endemic areas [16], [17] and in a longitudinal study of subjects parasitemic at entry, antibodies to AMA-1 were significantly associated with fewer episodes of clinical malaria [18].

While antibody-mediated immunity to AMA-1 appears to be critical for immune protection, AMA-1-specific T cell-mediated immunity (CMI) might also play a significant role in protective immunity. AMA-1 is also present on the sporozoite surface and a strong T cell response against AMA-1 may have an inhibitory effect on sporozoite invasion and exoerythrocytic stage development in the liver [19]. Just before the liver phase where CMI plays a key role in host defense, AMA-1 is translocated to the surface of the sporozoite, undergoes proteolytic cleavage and is essential to hepatocyte invasion [20], [21]. T cells specific for AMA-1 may regulate antibody production as demonstrated in immunized BALB/c mice [9]. Evidence of antibody-independent protective CMI can be demonstrated by the finding that AMA-1 immunized mice and immunized B-cell-deficient mice, which are then depleted of CD4+ T cells, experience a loss of immunity unrelated to changes in antibody levels. CD4+ T cells specific for a conserved AMA-1 cryptic epitope transferred into athymic mice resulted in partial protection from parasite challenge [22]. T cell epitopes that elicit proliferative responses in Kenyan volunteers have been demonstrated in P. falciparum AMA-1 [23]. Strong T cell responses to AMA-1 were also seen in irradiated sporozoite-immunized volunteers protected against experimental malaria challenge [24]. Thus, while antibodies play a crucial role in protection post-AMA-1 immunization, antibody-independent CMI to AMA-1 contributes separately to the overall protective immune response in mice and might also do so in humans.

Immunization strategies that induce both a strong humoral response and CMI may be desirable in development of an AMA-1-based malaria vaccine. A Phase 1 trial of AMA-1/AS02A in malaria-naïve adults conducted at the Walter Reed Army Institute for Research (WRAIR) revealed strong humoral and cellular responses [3]. In the subsequent, presently described trial we measured CMI in semi-immune Malian adults after immunization with three doses of AMA-1 (FMP2.1/AS02A). Enhanced AMA-1-specific CMI was demonstrated in vaccine recipients as compared to control volunteers. These results, together with those demonstrating strong anti-AMA-1 antibody responses [25] provide evidence that multifaceted immune responses can be induced by this vaccination strategy and add further impetus for the continuing clinical evaluation of this vaccine.

Section snippets

Study design and vaccine administration

The clinical study was conducted at the Bandiagara Malaria Project research clinic in Bandiagara, a rural town of 13,634 inhabitants in the Dogon country in central Mali. Malaria transmission is seasonal and heavy with children aged less than 10 years having an average of two clinical malaria episodes per transmission season [26] and severe malaria affecting 2.3% of children less than 6 years of age [27]. The malaria transmission season extends from July to December. Venous blood was obtained

Lymphocyte proliferation

Lymphocyte proliferation assays were performed with PBMC from volunteers on Day 0 and 90. Fifty-five of 60 volunteers had PBMC from both time points (19 controls receiving RabAvert®, 18 volunteers receiving the half-dose FMP2.1/AS02A and 18 volunteers receiving the full dose FMP2.1/AS02A). Three participants had insufficient cells collected for analysis and two participants’ PBMC did not respond to the positive control stimuli. The Day 0 geometric mean SI for all participants with paired

Discussion

The FMP2.1/AS02A malaria vaccine appears to induce detectable cellular as well as humoral responses in malaria semi-immune adults. Measurable T cell-mediated immune responses (IL-5 production and lymphocyte proliferative responses) were detected in semi-immune malaria-exposed adults who received the AMA-1 vaccine. A balanced Th1/T2 cytokine response has been described in preclinical monkey trials after vaccination with AS02A adjuvanted to P. falciparum MSP142 antigen as compared to other

Acknowledgements

Vaccine production and laboratory assays were supported by the United States Agency for International Development, Washington, DC and by the Military Infectious Diseases Research Program, Fort Detrick, MD. The authors would like to thank the laboratory of Urszula Krzych for help in developing laboratory assay methodology, and Ms. Lisa Ware, Project Manager at WRAIR for her support. We would expressly like to acknowledge the population of Bandiagara who so graciously consented to participate in

References (35)

  • V.A. Stewart et al.

    Pre-clinical evaluation of new adjuvant formulations to improve the immunogenicity of the malaria vaccine RTS S/AS02A

    Vaccine

    (2006)
  • E.M. Malkin et al.

    Phase 1 clinical trial of apical membrane antigen 1: an asexual blood-stage vaccine for Plasmodium falciparum malaria

    Infect Immun

    (2005)
  • G.H. Mitchell et al.

    Apical membrane antigen 1, a major malaria vaccine candidate, mediates the close attachment of invasive merozoites to host red blood cells

    Infect Immun

    (2004)
  • K. Kato et al.

    Domain III of Plasmodium falciparum apical membrane antigen 1 binds to the erythrocyte membrane protein Kx

    Proc Natl Acad Sci USA

    (2005)
  • F.H. Amante et al.

    A cryptic T cell epitope on the apical membrane antigen 1 of Plasmodium chabaudi adami can prime for an anamnestic antibody response: implications for malaria vaccine design

    J Immunol

    (1997)
  • S. Dutta et al.

    Invasion-inhibitory antibodies inhibit proteolytic processing of apical membrane antigen 1 of Plasmodium falciparum merozoites

    Proc Natl Acad Sci USA

    (2003)
  • A.N. Hodder et al.

    Specificity of the protective antibody response to apical membrane antigen 1

    Infect Immun

    (2001)
  • Cited by (18)

    • Strain-specific Plasmodium falciparum multifunctional CD4<sup>+</sup> T cell cytokine expression in Malian children immunized with the FMP2.1/AS02<inf>A</inf> vaccine candidate

      2016, Vaccine
      Citation Excerpt :

      Among AMA1 vaccine recipients, 18 of 21 children demonstrated significantly increased AMA1 antigen-specific levels of IFN-γ, TNF-α, and IL-2 derived from CD4+ T cells by D150 compared to 0/10 who received the control rabies vaccine over the course of a malaria transmission season. These findings are in contrast to data collected from a Malian adult population via ELISpot after the same FMP2.1/AS02A vaccine regimen in which all participants, including control rabies vaccine recipients, demonstrated an AMA1-specific IFN-γ production [24]. The high baseline of cell-mediated IFN-γ production in Malian adults versus the pediatric population may be the result of a natural exposure to malaria inducing a lymphocyte response not yet acquired in children.

    • Targeting of Toll-like receptors: A decade of progress in combating infectious diseases

      2011, The Lancet Infectious Diseases
      Citation Excerpt :

      RTS,S formulated with the AS01 adjuvant system—containing MPL and QS21 in liposomes18—had superior immunogenicity and comparable safety and sustained effectiveness compared with the AS02-adjuvant candidate vaccine in both preclinical and clinical studies.33,34 The AMA1-based FMP2.1/AS02 P falciparum malaria vaccine elicited potent humoral and cellular immune responses with promising safety and tolerability profiles in malaria-naive and malaria-exposed children and adults.35–38 The TLR4 agonist RC-529 (Ribi.529; Corixa, Seattle, WA, USA), a fully synthetic acylated monosaccharide mimetic of MPL, has shown great promise as a potent adjuvant in both preclinical and clinical studies.39–41

    • Identification and characterization of autoantibody-producing B220<sup>low</sup> B (B-1) cells appearing in malarial infection

      2010, Cellular Immunology
      Citation Excerpt :

      Many immunologists and clinicians are familiar with immunological responses mediated by conventional T and B cells and have considered that such mediation might also occur in malarial infection. Indeed, immunologists in the field of parasitology tend to use vaccination against malaria parasites in animals and humans [1–9]. However, we have noticed that malarial infection does not activate conventional T and B cells, but rather activates unconventional T cells (i.e., NK1.1−TCRint cells) [10–13].

    • A field trial to assess a blood-stage malaria vaccine

      2011, New England Journal of Medicine
      Citation Excerpt :

      Alternatively, the postvaccination rise in antibody levels may reflect enhancement of some other protective mechanism. We previously reported that FMP2.1/AS02A elicited cellular immune responses in Malian adults.19 Studies of cellular immune responses, IgG subclass, antibody avidity, and serologic growth inhibition may identify immune correlates of protection and inform future decisions about vaccine development for clinical use.

    View all citing articles on Scopus
    View full text