Articles
PRIMVAC vaccine adjuvanted with Alhydrogel or GLA-SE to prevent placental malaria: a first-in-human, randomised, double-blind, placebo-controlled study

https://doi.org/10.1016/S1473-3099(19)30739-XGet rights and content

Summary

Background

PRIMVAC is a VAR2CSA-derived placental malaria vaccine candidate aiming to prevent serious clinical outcomes of Plasmodium falciparum infection during pregnancy. We assessed the safety and immunogenicity of PRIMVAC adjuvanted with Alhydrogel or glucopyranosyl lipid adjuvant in stable emulsion (GLA-SE) in French and Burkinabe women who were not pregnant.

Methods

This first-in-human, randomised, double-blind, placebo-controlled, dose escalation trial was done in two staggered phases, a phase 1A trial in 18–35-year-old women who were malaria naive in a hospital in France and a subsequent phase 1B trial in women who were naturally exposed to P falciparum and nulligravid in the clinical site of a research centre in Burkina Faso. Volunteers were recruited into four sequential cohorts receiving PRIMVAC intramuscularly at day 0, 28, and 56: two cohorts in France receiving 20 μg or 50 μg of PRIMVAC and then two in Burkina Faso receiving 50 μg or 100 μg of PRIMVAC. Volunteers were randomly assigned (1:1) to two groups (PRIMVAC adjuvanted with either Alhydrogel or GLA-SE) in France and randomly assigned (2:2:1) to three groups (PRIMVAC adjuvanted with either Alhydrogel, GLA-SE, or placebo) in Burkina Faso. Randomisation was centralised, using stratification by cohort and blocks of variable size, and syringes were masked by opaque labels. The primary endpoint was the proportion of participants with any grade 3 or higher adverse reaction to vaccination up until day 35. Safety at later time points as well as humoral and cellular immunogenicity were assessed in secondary endpoints. This trial is registered with ClinicalTrials.gov, NCT02658253.

Findings

Between April 19, 2016, and July 13, 2017, 68 women (18 in France, 50 in Burkina Faso) of 101 assessed for eligibility were included. No serious adverse event related to the vaccine occurred. PRIMVAC antibody titres increased with each dose and seroconversion was observed in all women vaccinated with PRIMVAC (n=57). PRIMVAC antibody titres reached a peak (geometric mean 11 843·0, optical density [OD] 1·0, 95% CI 7559·8–18 552·9 with 100 μg dose and GLA-SE) 1 week after the third vaccination (day 63). Compared with Alhydrogel, GLA-SE tended to improve the PRIMVAC antibody response (geometric mean 2163·5, OD 1·0, 95% CI 1315·7–3557·7 with 100 μg dose and Alhydrogel at day 63). 1 year after the last vaccination, 20 (71%) of 28 women who were vaccinated with PRIMVAC/Alhydrogel and 26 (93%) of 28 women who were vaccinated with PRIMVAC/GLA-SE still had anti-PRIMVAC antibodies, although antibody magnitude was markedly lower (452·4, OD 1·0, 95% CI 321·8–636·1 with 100 μg dose and GLA-SE). These antibodies reacted with native homologous VAR2CSA expressed by NF54-CSA infected erythrocytes (fold change from baseline at day 63 with 100 μg dose and GLA-SE: 10·74, 95% CI 8·36–13·79). Limited cross-recognition, restricted to sera collected from women that received the 100 μg PRIMVAC dose, was observed against heterologous VAR2CSA variants expressed by FCR3-CSA (fold change from baseline at day 63: 1·49, 95% CI 1·19–1·88) and 7G8-CSA infected erythrocytes (1·2, 1·08–1·34).

Interpretation

PRIMVAC adjuvanted with Alhydrogel or GLA-SE had an acceptable safety profile, was immunogenic, and induced functional antibodies reacting with the homologous VAR2CSA variant expressed by NF54-CSA infected erythrocytes. Cross-reactivity against heterologous VAR2CSA variants was limited and only observed in the higher dose group. An alternate schedule of immunisation, antigen dose, and combinations with other VAR2CSA-based vaccines are envisaged to improve the cross-reactivity against heterologous VAR2CSA variants.

Funding

Bundesministerium für Bildung und Forschung, through Kreditanstalt für Wiederaufbau, Germany; Inserm, and Institut National de Transfusion Sanguine, France; Irish Aid, Department of Foreign Affairs and Trade, Ireland.

Introduction

According to the 2019 WHO malaria report, 228 million cases of malaria occurred in 2018 leading to 405 000 deaths.1 The majority of clinical cases and deaths occurred in sub-Saharan Africa and mainly resulted from Plasmodium falciparum infection. In malaria endemic areas, individuals progressively acquire clinical immunity during childhood and adults are therefore generally protected against the severe clinical outcomes of the disease.2 However, during their first pregnancies, women become once again susceptible to the serious clinical outcomes associated with placental malaria.3 Placental malaria can lead to maternal anaemia, hypertension, and stillbirth. Their infant can have low birthweight due to premature delivery and fetal growth retardation.3 Low birthweight is an important risk factor for neonatal and infant death.4 A modelling study showed that up to 40% of pregnant women in sub-Saharan Africa develop placental malaria.5 Furthermore, P falciparum malaria was responsible for 11% of low birthweight-related infant mortality in sub-Saharan Africa6 and an estimate of 217 026 stillbirths (20% of all stillbirths in sub-Saharan Africa).7 Remarkably, the prevalence of placental malaria sharply drops with successive pregnancies.4 This protection has been associated with the development of antibodies directed towards the surface of infected erythrocytes from placental origin.8 Therefore, a vaccine priming the immunity observed in women who are multigravid could have a large effect on both disease incidence and severity and possibly save around 100 000 lives each year.3

The severe outcomes of placental malaria results from the massive accumulation of infected erythrocytes in the placental intervillous spaces2 through the binding to the placental chondroitin sulfate A (CSA), a binding phenotype not seen outside of the pregnancy context.9 Infected erythrocyte adhesion is mediated by the highly diverse P falciparum erythrocyte membrane protein 1 family (PfEMP1) encoded by the var genes.2

Evidence strongly supports the PfEMP1-VAR2CSA variant as the leading candidate for a placental malaria vaccine. Indeed, VAR2CSA is preferentially expressed by infected erythrocytes from placental origin10 and recombinant VAR2CSA binds to CSA.11 Genetic deletion of var2csa results in the loss of infected erythrocyte adhesion to CSA that cannot be compensated by any other PfEMP1.12 Antisera to recombinant VAR2CSA react to the surface of CSA-binding infected erythrocytes and inhibit their adhesion to CSA.13, 14, 15 Women gradually acquire strain-transcendent antibodies recognising recombinant and native VAR2CSA expressed on infected erythrocytes, which also inhibit infected erythrocyte adhesion to the placenta, thus correlating with placental malaria protection.8, 16, 17 Taken together, these data provide a rational basis for developing a VAR2CSA-derived placental malaria vaccine. Two placental malaria vaccine candidates (PRIMVAC and PAMVAC) are currently under clinical development.18 Following an extensive screening study, PRIMVAC spanning the CSA-binding DBL1x-2x region of the 3D7-VAR2CSA variant was selected15 and transitioned to current good manufacturing practice production.19 Preliminary safety and immunogenicity results of the PAMVAC study, assessing a placental malaria vaccine derived from the FCR3-VAR2CSA variant in volunteers who were malaria naive in Germany, reported that PAMVAC is safe and can induce functional antibodies against the homologous VAR2CSA-expressing strain.20

We aimed to assess the safety and immunogenicity of PRIMVAC in both women who were malaria naive and not pregnant in France and in women who were P falciparum-naturally exposed and nulligravid in Burkina Faso.

Section snippets

Study design and participants

We did a first-in-human, randomised, double-blind, placebo-controlled, phase 1A/1B dose escalation trial, evaluating the safety and immunogenicity of three vaccinations with progressively higher doses of PRIMVAC adjuvanted with Alhydrogel or glucopyranosyl lipid adjuvant in stable emulsion (GLA-SE). 68 healthy adult women who were not pregnant were enrolled in four sequential cohorts. The trial started with two cohorts in women who were malaria-naive in Cochin Hospital in France (phase 1A, with

Results

Between April 19, 2016, and July 5, 2016, 25 women were screened and 18 (72%) were included in cohorts A and B in France (figure 1). Six (33%) participants were first assigned to receive 20 μg of PRIMVAC in combination with either Alhydrogel (n=3) or GLA-SE (n=3). The 12 (67%) other participants were assigned to receive 50 μg of PRIMVAC/Alhydrogel (n=6) or 50 μg of PRIMVAC/GLA-SE (n=6). Between Nov 25, 2016, and July 13, 2017, 76 women were screened and 50 (66%) were included in cohorts C and D

Discussion

VAR2CSA-based placental malaria vaccines administered before first pregnancy stand as the main anti-disease strategy to reduce malaria morbidity and mortality during pregnancy.18 Here, we report the results of a phase 1A/1B clinical trial, assessing the safety and immunogenicity of the PRIMVAC placental malaria vaccine. PRIMVAC adjuvanted with either Alhydrogel or GLA-SE presented an acceptable safety profile, as only grade 1 and 2 adverse events related to the vaccine were reported. All women

Data sharing

All data requests should be submitted to the corresponding author for consideration and will be reviewed according to the trial sponsor's obligations. Access to data might be granted following review.

References (32)

  • M Fried et al.

    Maternal antibodies block malaria

    Nature

    (1998)
  • M Fried et al.

    Adherence of Plasmodium falciparum to chondroitin sulfate A in the human placenta

    Science

    (1996)
  • A Salanti 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)
  • A Srivastava et al.

    Full-length extracellular region of the var2CSA variant of PfEMP1 is required for specific, high-affinity binding to CSA

    Proc Natl Acad Sci USA

    (2010)
  • NK Viebig et al.

    A single member of the Plasmodium falciparum var multigene family determines cytoadhesion to the placental receptor chondroitin sulphate A

    EMBO Rep

    (2005)
  • M Avril et al.

    Antibodies to a full-length VAR2CSA immunogen are broadly strain-transcendent but do not cross-inhibit different placental-type parasite isolates

    PLoS One

    (2011)
  • Cited by (76)

    • Disulfide bond and crosslinking analyses reveal inter-domain interactions that contribute to the rigidity of placental malaria VAR2CSA structure and formation of CSA binding channel

      2023, International Journal of Biological Macromolecules
      Citation Excerpt :

      However, in these studies, significant portions of the VAR2CSA structure, including ID1, ID2, linker connecting ID2-DBL3X and DBL3X-DBL4ε have not been modeled due to insufficient isoelectric potential maps. These regions include parts of the minimal C4S-binding region, such as ID1, that are common component of the placental malaria vaccine candidates [34–38]. Moreover, the structural elements that contribute to the stability of compactly folded NTS-ID3 core structure, including the interactions between IDs and DBL domains have not been completely understood.

    • Current approaches to malaria vaccines

      2022, Current Opinion in Microbiology
    View all citing articles on Scopus

    Co-last authors

    View full text