First-in-human, Randomized, Double-blind Clinical Trial of Differentially Adjuvanted PAMVAC, A Vaccine Candidate to Prevent Pregnancy-associated Malaria

Benjamin Mordmüller, Mihály Sulyok, Diane Egger-Adam, Mafalda Resende, Willem A de Jongh, Mette H Jensen, Helle Holm Smedegaard, Sisse B Ditlev, Max Soegaard, Lars Poulsen, Charlotte Dyring, Carlos Lamsfus Calle, Annette Knoblich, Javier Ibáñez, Meral Esen, Philippe Deloron, Nicaise Ndam, Saadou Issifou, Sophie Houard, Randall F Howard, Steven G Reed, Odile Leroy, Adrian J F Luty, Thor G Theander, Peter G Kremsner, Ali Salanti, Morten A Nielsen, Benjamin Mordmüller, Mihály Sulyok, Diane Egger-Adam, Mafalda Resende, Willem A de Jongh, Mette H Jensen, Helle Holm Smedegaard, Sisse B Ditlev, Max Soegaard, Lars Poulsen, Charlotte Dyring, Carlos Lamsfus Calle, Annette Knoblich, Javier Ibáñez, Meral Esen, Philippe Deloron, Nicaise Ndam, Saadou Issifou, Sophie Houard, Randall F Howard, Steven G Reed, Odile Leroy, Adrian J F Luty, Thor G Theander, Peter G Kremsner, Ali Salanti, Morten A Nielsen

Abstract

Background: Malaria in pregnancy has major impacts on mother and child health. To complement existing interventions, such as intermittent preventive treatment and use of impregnated bed nets, we developed a malaria vaccine candidate with the aim of reducing sequestration of asexual "blood-stage" parasites in the placenta, the major virulence mechanism.

Methods: The vaccine candidate PAMVAC is based on a recombinant fragment of VAR2CSA, the Plasmodium falciparum protein responsible for binding to the placenta via chondroitin sulfate A (CSA). Healthy, adult malaria-naive volunteers were immunized with 3 intramuscular injections of 20 μg (n = 9) or 50 μg (n = 27) PAMVAC, adjuvanted with Alhydrogel or glucopyranosyl lipid adjuvant in stable emulsion (GLA-SE) or in a liposomal formulation with QS21 (GLA-LSQ). Allocation was random and double blind. The vaccine was given every 4 weeks. Volunteers were observed for 6 months following last immunization.

Results: All PAMVAC formulations were safe and well tolerated. A total of 262 adverse events (AEs) occurred, 94 (10 grade 2 and 2 grade 3) at least possibly related to the vaccine. No serious AEs occurred. Distribution and severity of AEs were similar in all arms. PAMVAC was immunogenic in all participants. PAMVAC-specific antibody levels were highest with PAMVAC-GLA-SE. The antibodies inhibited binding of VAR2CSA expressing P. falciparum-infected erythrocytes to CSA in a standardized functional assay.

Conclusions: PAMVAC formulated with Alhydrogel or GLA-based adjuvants was safe, well tolerated, and induced functionally active antibodies. Next, PAMVAC will be assessed in women before first pregnancies in an endemic area.

Clinical trials registration: EudraCT 2015-001827-21; ClinicalTrials.gov NCT02647489.

Keywords: VAR2CSA; first-in-human; malaria vaccine; phase 1 clinical trial; pregnancy-associated malaria.

© The Author(s) 2019. Published by Oxford University Press for the Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
Study flow chart. Abbreviations: GLA-LSQ, glucopyranosyl lipid adjuvant containing QS21/saponin; GLA-SE, glucopyranosyl lipid adjuvant in stable emulsion; PAMVAC, pregnancy-associated malaria vaccine.
Figure 2.
Figure 2.
Pregnancy-associated malaria vaccine (PAMVAC) immunoglobulin G (IgG) over time. Enzyme-linked immunosorbent assay was used to measure IgG antibody reactivity generated against PAMVAC. The y-axis shows the area under the curve (AUC) calculated for each individual at day 7, 28, 35, 56, 63, 84, and 252. The AUC unit was derived from recording the optical density values in plasma after 11 two-fold dilutions (from 1/100 to 1/102 400), yielding a titration curve under which the area was calculated. Shown are the AUC values for individuals who received the 20-µg dose (upper panel) and 50-µg dose (lower panel) according to adjuvant and days after first vaccination. Each individual was vaccinated at time 0, 28, and 56 days (vertical dotted lines). Abbreviations: GLA-LSQ, glucopyranosyl lipid adjuvant containing QS21/saponin; GLA-SE, glucopyranosyl lipid adjuvant in stable emulsion.
Figure 3.
Figure 3.
Functional activity of induced antibodies. The binding inhibitory activity of plasma against erythrocytes infected with VAR2CSA expressing FCR3 parasites binding to chondroitin sulfate A (CSA). Samples from individuals enrolled in the study, a pool of plasma from naive Danes, and a pool of plasma from multiparous women living in a highly endemic area in Benin. The plasma was tested at day -1 at a dilution of 1:10 and at day 84 at a 1:20 dilution. Briefly, CSA was coated at the bottom of 96 well plates. Infected erythrocytes were added along with plasma. The plates were washed using a washing robot (Biomek 2000). Values shown are the percentage inhibition according to binding in wells without plasma after subtraction of background binding values in wells without CSA. The upper and lower horizontal lines indicate the positive and negative control plasma samples. All samples were run in triplicate. The 50-µg group is indicated by triangles; 20-µg group is indicated by circles. See also Supplementary Figure S2. Abbreviations: GLA-LSQ, glucopyranosyl lipid adjuvant containing QS21/saponin; GLA-SE, glucopyranosyl lipid adjuvant in stable emulsion.
Figure 4.
Figure 4.
Correlation matrix of immunoglobulin G (IgG) reactivity between VAR2CSA variants. Alhydrogel (A), glucopyranosyl lipid adjuvant containing QS21/saponin (B), and glucopyranosyl lipid adjuvant in stable emulsion (GLA-SE) (C). The shading is proportional to the level of cross-reactivity measured by correlating the reactivity of IgG in plasma from individuals in each arm of the study. Numbers indicated according to the shading are r2-values, denoting how close the data are to the fitted regression line. Note the overall higher correlation in the GLA-SE adjuvanted group. See also Supplementary Figure S4.

References

    1. Moore KA, Simpson JA, Scoullar MJL, McGready R, Fowkes FJI. Quantification of the association between malaria in pregnancy and stillbirth: a systematic review and meta-analysis. Lancet Glob Health 2017; 5:e1101–12.
    1. Miller LH, Baruch DI, Marsh K, Doumbo OK. The pathogenic basis of malaria. Nature 2002; 415:673–9.
    1. Baruch DI, Pasloske BL, Singh HB, et al. . Cloning the P. falciparum gene encoding PfEMP1, a malarial variant antigen and adherence receptor on the surface of parasitized human erythrocytes. Cell 1995; 82:77–87.
    1. Su XZ, Heatwole VM, Wertheimer SP, et al. . The large diverse gene family var encodes proteins involved in cytoadherence and antigenic variation of Plasmodium falciparum-infected erythrocytes. Cell 1995; 82:89–100.
    1. Bull PC, Lowe BS, Kortok M, Molyneux CS, Newbold CI, Marsh K. Parasite antigens on the infected red cell surface are targets for naturally acquired immunity to malaria. Nat Med 1998; 4:358–60.
    1. Nielsen MA, Staalsoe T, Kurtzhals JA, et al. . Plasmodium falciparum variant surface antigen expression varies between isolates causing severe and nonsevere malaria and is modified by acquired immunity. J Immunol 2002; 168:3444–50.
    1. Marsh K, Howard RJ. Antigens induced on erythrocytes by P. falciparum: expression of diverse and conserved determinants. Science 1986; 231:150–3.
    1. Salanti A, Staalsoe T, Lavstsen T, et al. . Selective upregulation of a single distinctly structured var gene in chondroitin sulphate A-adhering Plasmodium falciparum involved in pregnancy-associated malaria. Mol Microbiol 2003; 49:179–91.
    1. Ayres PM, Mandel CT, Pehrson C, et al. . Placental sequestration of Plasmodium falciparum malaria parasites is mediated by the interaction between VAR2CSA and chondroitin sulfate A on Syndecan-1. PLoS Pathog 2016; 12:e1005831.
    1. Fried M, Nosten F, Brockman A, Brabin BJ, Duffy PE. Maternal antibodies block malaria. Nature 1998; 395:851–2.
    1. Ricke CH, Staalsoe T, Koram K, et al. . Plasma antibodies from malaria-exposed pregnant women recognize variant surface antigens on Plasmodium falciparum-infected erythrocytes in a parity-dependent manner and block parasite adhesion to chondroitin sulfate A. J Immunol 2000; 165:3309–16.
    1. Salanti A, Dahlbäck M, Turner L, et al. . Evidence for the involvement of VAR2CSA in pregnancy-associated malaria. J Exp Med 2004; 200:1197–203.
    1. Tuikue Ndam NG, Salanti A, Le-Hesran JY, et al. . Dynamics of anti-VAR2CSA immunoglobulin G response in a cohort of Senegalese pregnant women. J Infect Dis 2006; 193:713–20.
    1. Brolin KJ, Persson KE, Wahlgren M, Rogerson SJ, Chen Q. Differential recognition of P. falciparum VAR2CSA domains by naturally acquired antibodies in pregnant women from a malaria endemic area. PLoS One 2010; 5:e9230.
    1. Tutterrow YL, Salanti A, Avril M, et al. . High avidity antibodies to full-length VAR2CSA correlate with absence of placental malaria. PLoS One 2012; 7:e40049.
    1. Mayor A, Kumar U, Bardají A, et al. . Improved pregnancy outcomes in women exposed to malaria with high antibody levels against Plasmodium falciparum. J Infect Dis 2013; 207:1664–74.
    1. Ndam NT, Denoeud-Ndam L, Doritchamou J, et al. . Protective antibodies against placental malaria and poor outcomes during pregnancy, Benin. Emerg Infect Dis 2015; 21:813–23.
    1. Duffy PE, Fried M. Antibodies that inhibit Plasmodium falciparum adhesion to chondroitin sulfate A are associated with increased birth weight and the gestational age of newborns. Infect Immun 2003; 71:6620–3.
    1. Dechavanne S, Srivastava A, Gangnard S, et al. . Parity-dependent recognition of DBL1X-3X suggests an important role of the VAR2CSA high-affinity CSA-binding region in the development of the humoral response against placental malaria. Infect Immun 2015; 83:2466–74.
    1. Nielsen MA, Resende M, de Jongh WA, et al. . The influence of sub-unit composition and expression system on the functional antibody response in the development of a VAR2CSA based Plasmodium falciparum placental malaria vaccine. PLoS One 2015; 10:e0135406.
    1. Pinto VV, Ditlev SB, Jensen KE, et al. . Differential induction of functional IgG using the Plasmodium falciparum placental malaria vaccine candidate VAR2CSA. PLoS One 2011; 6:e17942.
    1. Barfod L, Dobrilovic T, Magistrado P, et al. . Chondroitin sulfate A-adhering Plasmodium falciparum-infected erythrocytes express functionally important antibody epitopes shared by multiple variants. J Immunol 2010; 185:7553–61.
    1. Doritchamou J, Bigey P, Nielsen MA, et al. . Differential adhesion-inhibitory patterns of antibodies raised against two major variants of the NTS-DBL2X region of VAR2CSA. Vaccine 2013; 31:4516–22.
    1. Doritchamou J, Sabbagh A, Jespersen JS, et al. . Identification of a major dimorphic region in the functionally critical N-terminal ID1 domain of VAR2CSA. PLoS One 2015; 10:e0137695.
    1. Fernandez P, Petres S, Mécheri S, Gysin J, Scherf A. Strain-transcendent immune response to recombinant Var2CSA DBL5-ε domain block P. falciparum adhesion to placenta-derived BeWo cells under flow conditions. PLoS One 2010; 5:e12558.
    1. Fried M, Avril M, Chaturvedi R, et al. . Multilaboratory approach to preclinical evaluation of vaccine immunogens for placental malaria. Infect Immun 2013; 81:487–95.
    1. Schmiegelow C, Matondo S, Minja DTR, et al. . Plasmodium falciparum infection early in pregnancy has profound consequences for fetal growth. J Infect Dis 2017; 216:1601–10.
    1. Bonhoeffer J, Imoukhuede EB, Aldrovandi G, et al. ; Brighton Collaboration Clinical Trial Protocol Working Group Template protocol for clinical trials investigating vaccines–focus on safety elements. Vaccine 2013; 31:5602–20.
    1. Nielsen MA, Salanti A. High-throughput testing of antibody-dependent binding inhibition of placental malaria parasites. Methods Mol Biol 2015; 1325:241–53.
    1. Gbédandé K, Fievet N, Viwami F, et al. ; Multi-centre research paper Clinical development of a VAR2CSA-based placental malaria vaccine PAMVAC: quantifying vaccine antigen-specific memory B & T cell activity in Beninese primigravidae. Vaccine 2017; 35:3474–81.
    1. Hviid L, Salanti A. VAR2CSA and protective immunity against pregnancy-associated Plasmodium falciparum malaria. Parasitology 2007; 134:1871–6.
    1. Rogerson SJ, Hviid L, Duffy PE, Leke RF, Taylor DW. Malaria in pregnancy: pathogenesis and immunity. Lancet Infect Dis 2007; 7:105–17.
    1. Tuikue-Ndam N, Deloron P. Developing vaccines to prevent malaria in pregnant women. Expert Opin Biol Ther 2015; 15:1173–82.
    1. Pehrson C, Heno KK, Adams Y, et al. . Comparison of functional assays used in the clinical development of a placental malaria vaccine. Vaccine 2017; 35:610–8.
    1. McCall MBB, Kremsner PG, Mordmüller B. Correlating efficacy and immunogenicity in malaria vaccine trials. Semin Immunol 2018; 39:52–64.
    1. Cockburn IA, Seder RA. Malaria prevention: from immunological concepts to effective vaccines and protective antibodies. Nat Immunol 2018; 19:1199–211.
    1. Draper SJ, Angov E, Horii T, et al. . Recent advances in recombinant protein-based malaria vaccines. Vaccine 2015; 33:7433–43.
    1. Palladini A, Thrane S, Janitzek CM, et al. . Virus-like particle display of HER2 induces potent anti-cancer responses. Oncoimmunology 2018; 7:e1408749.
    1. Thrane S, Janitzek CM, Matondo S, et al. . Bacterial superglue enables easy development of efficient virus-like particle based vaccines. J Nanobiotechnology 2016; 14:30.

Source: PubMed

3
Iratkozz fel