Immunization with full-length Plasmodium falciparum merozoite surface protein 1 is safe and elicits functional cytophilic antibodies in a randomized first-in-human trial

Antje Blank, Kristin Fürle, Anja Jäschke, Gerd Mikus, Monika Lehmann, Johannes Hüsing, Kirsten Heiss, Thomas Giese, Darrick Carter, Ernst Böhnlein, Michael Lanzer, Walter E Haefeli, Hermann Bujard, Antje Blank, Kristin Fürle, Anja Jäschke, Gerd Mikus, Monika Lehmann, Johannes Hüsing, Kirsten Heiss, Thomas Giese, Darrick Carter, Ernst Böhnlein, Michael Lanzer, Walter E Haefeli, Hermann Bujard

Abstract

A vaccine remains a priority in the global fight against malaria. Here, we report on a single-center, randomized, double-blind, placebo and adjuvant-controlled, dose escalation phase 1a safety and immunogenicity clinical trial of full-length Plasmodium falciparum merozoite surface protein 1 (MSP1) in combination with GLA-SE adjuvant. Thirty-two healthy volunteers were vaccinated at least three times with MSP1 plus adjuvant, adjuvant alone, or placebo (24:4:4) to evaluate the safety and immunogenicity. MSP1 was safe, well tolerated and immunogenic, with all vaccinees sero-converting independent of the dose. The MSP1-specific IgG and IgM titers persisted above levels found in malaria semi-immune humans for at least 6 months after the last immunization. The antibodies were variant- and strain-transcending and stimulated respiratory activity in granulocytes. Furthermore, full-length MSP1 induced memory T-cells. Our findings encourage challenge studies as the next step to evaluate the efficacy of full-length MSP1 as a vaccine candidate against falciparum malaria (EudraCT 2016-002463-33).

Keywords: Malaria; Protein vaccines.

Conflict of interest statement

Competing interestsH.B., E.B. and M.L. are shareholders of Sumaya GmbH & Co. KG. The other authors declare no competing interests.

© The Author(s) 2020.

Figures

Fig. 1. Immunization scheme and study design.
Fig. 1. Immunization scheme and study design.
a Volunteers were immunized with SumayaVAC-1 (full length MSP1 plus GLA-SE as adjuvant, 500 µl volume) on days 0, 29, 57, and optionally after unblinding of the cohort on day 182. Blood samples were taken for serological analysis on the days indicated. The safety follow-up was scheduled 6 months after the last vaccination. b 32 healthy volunteers were recruited in two consecutive cohorts. Sixteen volunteers each were randomly assigned within a cohort. The number of volunteers receiving three or four vaccinations is indicated for each cohort.
Fig. 2. IgG and IgM antibody titers…
Fig. 2. IgG and IgM antibody titers against MSP1-D after immunization with SumayaVac-1.
Blood samples were collected from the vaccines on the days indicated and the a IgG-specific and b IgM-specific MSP1-D antibody titers were determined by ELISA. The data points represent the geometric means ± 95% confidence interval of each cohort. Crossed circles indicate samples from single individuals. Red arrows indicate the days of immunization. Dashed lines indicate reference titers obtained from pooled sera from semi-immune individuals from Nouna, Burkina Faso (orange line, n = 11) and from Kisumu, Kenya (black line, NIBSC code 10/198). Controls comprise the placebo and the GLA-SE vaccinees. The pie chart shows the average percentile distribution of IgG subclasses after three immunizations with SumayaVac-1 (day 85, n = 24).
Fig. 3. IgG responses against MSP1-D sub-fragments…
Fig. 3. IgG responses against MSP1-D sub-fragments and cross-reactivity against MSP1-F.
a Structural organization of MSP1. MSP1 is synthesized as a precursor protein of 196 kDa. During merozoite maturation, MSP1 is cleaved by PfSub1 into four major MSP1 fragments, termed p83, p30, p38, and p42. The cleavage sites are indicated by arrowheads. Conserved, dimorphic and oligomorphic domains of MSP1 are indicated. b The net α-MSP1-D titers of the vaccinees on day 85 are shown for the four major MSP1 fragments. Each data point represents the result (performed in duplicates) from one individual. Black marks indicate the mean value of the corresponding cohort. c Antibody titers against MSP1-D and MSP1-F after three immunizations on day 85.
Fig. 4. Recognition of native MSP1-D by…
Fig. 4. Recognition of native MSP1-D by induced antibodies.
a Representative indirect immuno-fluorescence assay (IFA) showing reactivity of serum from an individual immunized 3× with SumayaVac-1 with developing merozoites during schizogony and free mature merozoites. Trophozoites served as a negative control. Trophozoite and schizont samples were fixed for analysis, whereas merozoites were unfixed. Scale bar, 5 µm. b Correlation between the α-merozoite titer and the α-MSP1-D IgG titer for all volunteers vaccinated 3x with SumayaVac-1 (day 85, n = 24). Each data point represents the results from one vaccinee. Titers were determined by quantitative ELISAs, whereby one set of plates was coated with purified full-length MSP1 and the other with purified P. falciparum merozoites. The correlation between two parameters was assessed using Pearson product moment correlation (correlation coefficient, 0.624; p = 0.0011).
Fig. 5. Effect of induced α-MSP1 antibodies…
Fig. 5. Effect of induced α-MSP1 antibodies on parasite growth inhibition, complement activation, and invasion inhibition.
a Growth inhibition assay (GIA). The P. falciparum strain 3D7 was cultured for one cycle in the presence of total IgGs from volunteers vaccinated 3x with SumayaVac-1 (day 85; n = 14, circles). Two IgG preparations each from MSP1-immunized rabbits (triangles) and semi-immune adults from Burkina Faso (squares) served as positive controls, and pooled IgGs from volunteers collected on day 0 (inverted triangles) as negative controls. The concentrations of purified total IgG antibodies used in the assay are indicated. Inset: GIA activity in the presence of active complement. b Sera from volunteers vaccinated 3x with SumayaVac-1 (day 85; n = 24) fix complement in the presence of purified MSP1, as determined using a C1q-specific ELISA. The readout is the absorbance at a wavelength of 492 nm. A serum pool from semi-immune adults from Burkina Faso (n = 11) served as a positive control and the sera from the placebo and GLA-SE vacinees as negative control. Each data point represents the result (performed in duplicates) from one individual. A box plot analysis is overlaid over the individual data points with the median (black line), mean (red lines), and 25% and 75% quartile ranges being shown. The error bars above and below the box indicate the 90th and 10th percentiles. Filled black circles indicated outliers. Statistical significance was assessed using one way ANOVA. c Direct invasion assay (IIA). Purified merozoites (3D7) were incubated with erythrocytes in the presence of 3 mg ml−1 purified IgG for 30 min before the number of invaded erythrocyte was determined and the parasitemia calculated. IgG preparations from rabbits immunized with apical membrane antigen 1 (AMA-1) and a semi-immune adult from Burkina Faso served as positive controls and an IgG pool from the placebo cohort as negative control. Each data point represents the result (performed in duplicates) from one individual. Box plot analysis as described above.
Fig. 6. Mapping of linear B-cell epitopes…
Fig. 6. Mapping of linear B-cell epitopes across MSP1.
Sera from vaccinees (1:1000), two malaria semi-immune adults from Burkina Faso (1:1000), and two MSP1 immunized rabbits (1:10,000) were applied to a custom-made MSP1 peptide microarray chip consisting of 1706 oligomeric peptides of 15 amino acids with a peptide-to-peptide overlap of 14 amino acids. Each peptide was printed in duplicate on the chip. As secondary antibodies the appropriate DyLight680 conjugated anti-IgG (Fc) antibodies were used. The fluorescence intensity landscapes across MSP1 are shown for each sample. Relevant epitopes have been highlighted. Controls comprise the placebo and the GLA-SE vaccinees. For orientation, the structural organization of MSP1 is shown below the fluorescence profiles. Full details of epitopes and fluorescence intensities are provided in the Supplementary Table 2.
Fig. 7. Association of GIA activity with…
Fig. 7. Association of GIA activity with MSP1 epitopes.
Major epitopes putatively associated with GIA activity as defined by LOD scores > 2.5 (p = 0.001) for at least three consecutive peptides are highlighted. The GIA activity underpinning the correlation are depicted in Fig. 5a, with sera from MSP1-immunized rabbits and semi-immune adults from Burkina Faso showing GIA activity, whereas sera from vaccinees being GIA inactive. Epitopes that map to MSP1 processing sites, are indicated in blue, with the cleavage site being pointed out by an arrow. Note that three processing sites were detected between the p38/p42 junction: the canonical cleavage site (TGEAISV) and two adjacent alternative sites. Another epitope includes a lysine (highlighted in red) that is posttranslationally modified by acetylation. The correlation was determined using a Pearson function followed by a Bonferroni correction and conversion of the resulting p-values into a LOD score (logarithm of odds). The confidence line as defined by p < 0.001 is indicated. For orientation, the structural organization of MSP1 is shown below the graph. Full details of epitopes and LOD scores are provided in the Supplementary Table 2.
Fig. 8. Activation of neutrophils by IgG…
Fig. 8. Activation of neutrophils by IgG from volunteers immunized with SumayaVac-1.
P. falciparum merozoites from the a 3D7 strain expressing MSP1-D or the b FCB1 strain expressing MSP1-F and neutrophils were incubated in the presence of purified IgG from volunteers immunized three times with SumayaVac-1 (collected at days 0 and 85). Opsonized merozoites activated neutrophils to produce a respiratory burst, which was quantitatively expressed in terms of an ADRB index. Each data point represents the mean ADRB Index of at least two independent experiments each performed in duplicates from one volunteer. Black marks indicate the geometric mean values of the corresponding cohort. Statistical significance was assessed using paired t-test (between day 0 and day 85) and the Kruskal–Wallis one-way ANOVA on ranks (between different dosing groups). ADRB activities for purified IgG from semi-immune adults from Nouna, Burkina Faso (n = 11), and from Kenya (pooled IgG) are indicated as reference. c Correlation between the net ADRB index (corrected for the background activity at day 0) and the α-MSP1-D titer (at day 85) (correlation coefficient, 0.545; p = 0.006, according to Spearman rank order correlation). Each data point represents the results from one volunteer (n = 24).
Fig. 9. MSP1-specific recall T-cell response.
Fig. 9. MSP1-specific recall T-cell response.
PBMCs from six volunteers immunized three times with SumayaVac-1 were exposed to purified MSP1-D (5 µg ml−1 for 24 h) and the recall T-cell response as defined by IFN-γ production was measured in the cultured ELISpot assay. a Representative images of ELISpot wells are shown for different vaccinees. b The net mean spot-forming units (SFU) per 1 million PBMCs were calculated and analyzed as a function of time post the first immunization. Each data point represents the mean from three determinations ± SD. The red arrows indicate the days of immunization with SumayaVac-1. Significance was assessed using Kruskal–Wallis one way ANOVA on ranks. c Stimulation of PBMCs by peptides corresponding to HLA-A0201-restricted MSP1 CD8+ T-cell epitopes. PBMCs from the same vaccinees as above were analyzed (same color code as above), in addition to PBMCs from three semi-immune adults from Burkina Faso (B.F.). The following peptides were used: 291, GLHHLITEL; 374, SLLTELQQV; 437, VIYLKPLAGV; 674, KLKEFIPKV.,, Statistical significance was assessed using the two tailed t-test.

References

    1. World Health Organization. World Malaria Report 2018 (WHO Press, Geneva 2018).
    1. Ashley EA, Pyae Phyo A, Woodrow CJ. Malaria. Lancet. 2018;391:1608–1621.
    1. Visser BJ, van Vugt M, Grobusch MP. Malaria: an update on current chemotherapy. Expert Opin. Pharmacother. 2014;15:2219–2254.
    1. Moorthy VS, Newman RD, Okwo-Bele JM. Malaria vaccine technology roadmap. Lancet. 2013;382:1700–1701.
    1. World Health Organization. Global Technical Strategy for Malaria 2016–2030 (WHO Press, Geneva, 2016).
    1. Marsh K, Kinyanjui S. Immune effector mechanisms in malaria. Parasite Immunol. 2006;28:51–60.
    1. Miura K. Progress and prospects for blood-stage malaria vaccines. Expert Rev. Vaccines. 2016;15:765–781.
    1. Wilson KL, Flanagan KL, Prakash MD, Plebanski M. Malaria vaccines in the eradication era: current status and future perspectives. Expert Rev. Vaccines. 2019;18:133–151.
    1. Witte D, et al. Safety and immunogenicity of seven dosing regimens of the candidate RTS,S/AS01E malaria vaccine integrated within an expanded program on immunization regimen: a phase II, single-center, open, controlled trial in infants in Malawi. Pediatr. Infect. Dis. J. 2018;37:483–491.
    1. Rampling T, et al. Safety and efficacy of novel malaria vaccine regimens of RTS,S/AS01B alone, or with concomitant ChAd63-MVA-vectored vaccines expressing ME-TRAP. NPJ Vaccines. 2018;3:49.
    1. Dimala CA, Kika BT, Kadia BM, Blencowe H. Current challenges and proposed solutions to the effective implementation of the RTS, S/AS01 Malaria Vaccine Program in sub-Saharan Africa: a systematic review. PLoS ONE. 2018;13:e0209744.
    1. Rts SCTP. Efficacy and safety of RTS,S/AS01 malaria vaccine with or without a booster dose in infants and children in Africa: final results of a phase 3, individually randomised, controlled trial. Lancet. 2015;386:31–45.
    1. World Health Organization. Malaria vaccine: WHO position paper—January 2016. Weekly Epidemiol Rec91, 33–52 (2016).
    1. Kurtovic L, et al. Induction and decay of functional complement-fixing antibodies by the RTS,S malaria vaccine in children, and a negative impact of malaria exposure. BMC Med. 2019;17:45.
    1. Child MA, Epp C, Bujard H, Blackman MJ. Regulated maturation of malaria merozoite surface protein-1 is essential for parasite growth. Mol. Microbiol. 2010;78:187–202.
    1. Das S, et al. Processing of Plasmodium falciparum merozoite surface protein MSP1 activates a spectrin-binding function enabling parasite egress from RBCs. Cell Host Microbe. 2015;18:433–444.
    1. Lin CS, et al. Multiple Plasmodium falciparum merozoite surface protein 1 complexes mediate merozoite binding to human erythrocytes. J. Biol. Chem. 2016;291:7703–7715.
    1. Lin CS, et al. The merozoite surface protein 1 complex is a platform for binding to human erythrocytes by Plasmodium falciparum. J. Biol. Chem. 2014;289:25655–25669.
    1. Baldwin MR, Li X, Hanada T, Liu SC, Chishti AH. Merozoite surface protein 1 recognition of host glycophorin A mediates malaria parasite invasion of red blood cells. Blood. 2015;125:2704–2711.
    1. Boyle MJ, Richards JS, Gilson PR, Chai W, Beeson JG. Interactions with heparin-like molecules during erythrocyte invasion by Plasmodium falciparum merozoites. Blood. 2010;115:4559–4568.
    1. Zhang Y, et al. Proteomic analysis of Plasmodium falciparum schizonts reveals heparin-binding merozoite proteins. J. Proteome Res. 2013;12:2185–2193.
    1. Goel VK, Li X, Chen H, Liu S-C, Chishti AH, Oh SS. Band 3 is a host receptor binding merozoite surface protein 1 during the Plasmodium falciparum invasion of erythrocytes. Proc. Natl Acad. Sci. USA. 2003;100:5164–5169.
    1. Blackman MJ, Heidrich HG, Donachie S, Mcbride JS, Holder AA. A single fragment of a malaria merozoite surface protein remains on the parasite during red-cell invasion and is the target of invasion-inhibiting antibodies. J. Exp. Med. 1990;172:379–382.
    1. Suhrbier A, Holder AA, Wiser MF, Nicholas J, Sinden RE. Expression of the precursor of the major merozoite surface antigens during the hepatic stage of malaria. Am. J. Trop. Med. Hyg. 1989;40:351–355.
    1. Szarfman A, Walliker D, McBride JS, Lyon JA, Quakyi IA, Carter R. Allelic forms of gp195, a major blood-stage antigen of Plasmodium falciparum, are expressed in liver stages. J. Exp. Med. 1988;167:231–236.
    1. Fowkes FJ, Richards JS, Simpson JA, Beeson JG. The relationship between anti-merozoite antibodies and incidence of Plasmodium falciparum malaria: A systematic review and meta-analysis. PLoS Med. 2010;7:e1000218.
    1. Bowman NM, et al. Longevity of genotype-specific immune responses to Plasmodium falciparum merozoite surface protein 1 in Kenyan children from regions of different malaria transmission intensity. Am. J. Trop. Med. Hyg. 2016;95:580–587.
    1. Milet J, et al. Genome-wide association study of antibody responses to Plasmodium falciparum candidate vaccine antigens. Genes Immun. 2016;17:110–117.
    1. Crompton PD, et al. A prospective analysis of the Ab response to Plasmodium falciparum before and after a malaria season by protein microarray. Proc. Natl Acad. Sci. USA. 2010;107:6958–6963.
    1. Dent AE, et al. Plasmodium falciparum protein microarray antibody profiles correlate with protection from symptomatic malaria in Kenya. J. Infect. Dis. 2015;212:1429–1438.
    1. Woehlbier U, Epp C, Hackett F, Blackman MJ, Bujard H. Antibodies against multiple merozoite surface antigens of the human malaria parasite Plasmodium falciparum inhibit parasite maturation and red blood cell invasion. Malar. J. 2010;9:77.
    1. Woehlbier U, et al. Analysis of antibodies directed against merozoite surface protein 1 of the human malaria parasite Plasmodium falciparum. Infect. Immun. 2006;74:1313–1322.
    1. Egan AF, Burghaus P, Druilhe P, Holder AA, Riley EM. Human antibodies to the 19kDa C-terminal fragment of Plasmodium falciparum merozoite surface protein 1 inhibit parasite growth in vitro. Parasite Immunol. 1999;21:133–139.
    1. O’Donnell RA, et al. Antibodies against merozoite surface protein (MSP)-1(19) are a major component of the invasion-inhibitory response in individuals immune to malaria. J. Exp. Med. 2001;193:1403–1412.
    1. Boyle MJ, et al. Human antibodies fix complement to inhibit Plasmodium falciparum invasion of erythrocytes and are associated with protection against malaria. Immunity. 2015;42:580–590.
    1. Joos C, et al. Antibodies to Plasmodium falciparum merozoite surface protein-1p19 malaria vaccine candidate induce antibody-dependent respiratory burst in human neutrophils. Malar. J. 2015;14:409.
    1. Galamo CD, Jafarshad A, Blanc C, Druilhe P. Anti-MSP1 block 2 antibodies are effective at parasite killing in an allele-specific manner by monocyte-mediated antibody-dependent cellular inhibition. J. Infect. Dis. 2009;199:1151–1154.
    1. Draper SJ, et al. Recombinant viral vaccines expressing merozoite surface protein-1 induce antibody- and T cell-mediated multistage protection against malaria. Cell Host Microbe. 2009;5:95–105.
    1. Krzych U, et al. T lymphocytes from volunteers immunized with irradiated Plasmodium falciparum sporozoites recognize liver and blood stage malaria antigens. J. Immunol. 1995;155:4072–4077.
    1. Carralot JP, Lemmel C, Stevanovic S, Pascolo S. Mass spectrometric identification of an HLA-A*0201 epitope from Plasmodium falciparum MSP-1. Int. Immunol. 2008;20:1451–1456.
    1. Daly TM, Long CA. A recombinant 15-kilodalton carboxyl-terminal fragment of Plasmodium yoelii yoelii 17XL merozoite surface protein 1 induces a protective immune response in mice. Infect. Immun. 1993;61:2462–2467.
    1. Renia L, Ling IT, Marussig M, Miltgen F, Holder AA, Mazier D. Immunization with a recombinant C-terminal fragment of Plasmodium yoelii merozoite surface protein 1 protects mice against homologous but not heterologous P. yoelii sporozoite challenge. Infect. Immun. 1997;65:4419–4423.
    1. Perrin LH, Merkli B, Loche M, Chizzolini C, Smart J, Richle R. Antimalarial immunity in Saimiri monkeys. Immunization with surface components of asexual blood stages. J. Exp. Med. 1984;160:441–451.
    1. Siddiqui WA, et al. Merozoite surface coat precursor protein completely protects Aotus monkeys against Plasmodium falciparum malaria. Proc. Natl Acad. Sci. USA. 1987;84:3014–3018.
    1. Etlinger HM, Caspers P, Matile H, Schoenfeld HJ, Stueber D, Takacs B. Ability of recombinant or native proteins to protect monkeys against heterologous challenge with Plasmodium falciparum. Infect. Immun. 1991;59:3498–3503.
    1. Ogutu BR, et al. Blood stage malaria vaccine eliciting high antigen-specific antibody concentrations confers no protection to young children in Western Kenya. PLoS ONE. 2009;4:e4708.
    1. Sheehy SH, et al. ChAd63-MVA-vectored blood-stage malaria vaccines targeting MSP1 and AMA1: assessment of efficacy against mosquito bite challenge in humans. Mol. Ther. 2012;20:2355–2368.
    1. Jäschke, A., Coulibaly, B., Remarque, E. J., Bujard, H. & Epp, C. Merozoite surface protein 1 from Plasmodium falciparum is a major target of opsonizing antibodies in individuals with acquired immunity against malaria. Clin. Vaccine Immunol.24, e00155–17 (2017).
    1. Crisanti A, Fruh K, Muller HM, Bujard H. The T cell reactivity against the major merozoite protein of Plasmodium falciparum. Immunol. Lett. 1990;25:143–148.
    1. Pusic KM, Hashimoto CN, Lehrer A, Aniya C, Clements DE, Hui GS. T cell epitope regions of the P. falciparum MSP1-33 critically influence immune responses and in vitro efficacy of MSP1-42 vaccines. PLoS ONE. 2011;6:e24782.
    1. Lee EA, et al. Identification of frequently recognized dimorphic T-cell epitopes in Plasmodium falciparum merozoite surface protein-1 in West and East Africans: lack of correlation of immune recognition and allelic prevalence. Am. J. Trop. Med. Hyg. 2001;64:194–203.
    1. Kim Y, et al. Immune epitope database analysis resource. Nucleic Acids Res. 2012;40:W525–530.
    1. Bujard H. Recombinant malaria vaccine. Patent EP, 07 000 710.9 (2007).
    1. Coler RN, et al. The TLR-4 agonist adjuvant, GLA-SE, improves magnitude and quality of immune responses elicited by the ID93 tuberculosis vaccine: first-in-human trial. NPJ Vaccines. 2018;3:34.
    1. Coler RN, et al. From mouse to man: safety, immunogenicity and efficacy of a candidate leishmaniasis vaccine LEISH-F3+GLA-SE. Clin. Transl. Immunol. 2015;4:e35.
    1. Windish HP, et al. Protection of mice from Mycobacterium tuberculosis by ID87/GLA-SE, a novel tuberculosis subunit vaccine candidate. Vaccine. 2011;29:7842–7848.
    1. Coler RN, et al. Development and characterization of synthetic glucopyranosyl lipid adjuvant system as a vaccine adjuvant. PLoS ONE. 2011;6:e16333.
    1. Stephens R, Langhorne J. Priming of CD4+ T cells and development of CD4+ T cell memory; lessons for malaria. Parasite Immunol. 2006;28:25–30.
    1. Conway DJ, Greenwood BM, McBride JS. Longitudinal study of Plasmodium falciparum polymorphic antigens in a malaria-endemic population. Infect. Immun. 1992;60:1122–1127.
    1. Babiker HA, Creasey AM, Fenton B, Bayoumi RA, Arnot DE, Walliker D. Genetic diversity of Plasmodium falciparum in a village in eastern Sudan. 1. Diversity of enzymes, 2D-PAGE proteins and antigens. Trans. R. Soc. Trop. Med. Hyg. 1991;85:572–577.
    1. Boyle MJ, et al. Isolation of viable Plasmodium falciparum merozoites to define erythrocyte invasion events and advance vaccine and drug development. Proc. Natl Acad. Sci. USA. 2010;107:14378–14383.
    1. Joos C, et al. Clinical protection from falciparum malaria correlates with neutrophil respiratory bursts induced by merozoites opsonized with human serum antibodies. PLoS ONE. 2010;5:e9871.
    1. Murungi LM, et al. Cord blood IgG and the risk of severe Plasmodium falciparum malaria in the first year of life. Int. J. Parasitol. 2017;47:153–162.
    1. Murungi LM, et al. Targets and mechanisms associated with protection from severe Plasmodium falciparum malaria in Kenyan children. Infect. Immun. 2016;84:950–963.
    1. Todryk SM, et al. The relationship between human effector and memory T cells measured by ex vivo and cultured ELISPOT following recent and distal priming. Immunology. 2009;128:83–91.
    1. Reece WH, et al. A CD4(+) T-cell immune response to a conserved epitope in the circumsporozoite protein correlates with protection from natural Plasmodium falciparum infection and disease. Nat. Med. 2004;10:406–410.
    1. Smith-Garvin JE, Koretzky GA, Jordan MS. T cell activation. Annu. Rev. Immunol. 2009;27:591–619.
    1. Idler I. Untersuchungen zur zellulären und humoralen Immunantwort gegen das Merozoitenoberflächenprotein-1 des Malariaerregers Plasmodium falciparum. PhD thesis, Heidelberg University, Heidelberg, Germany, 2004.
    1. Jäschke, A. Analysis of the human immune response against the Merozoite Surface Protein 1 (MSP-1) from Plasmodium falciparum—a malaria vaccine candidate. PhD thesis, Heidelberg University, Heidelberg, Germany, 2017.
    1. Herrera S, et al. Immunization of Aotus monkeys with Plasmodium falciparum blood-stage recombinant proteins. Proc. Natl Acad. Sci. USA. 1990;87:4017–4021.
    1. Kumar S, et al. Immunogenicity and in vivo efficacy of recombinant Plasmodium falciparum merozoite surface protein-1 in Aotus monkeys. Mol. Med. 1995;1:325–332.
    1. Cavanagh DR, et al. Antibody responses to a novel Plasmodium falciparum merozoite surface protein vaccine correlate with protection against experimental malaria infection in Aotus monkeys. PLoS ONE. 2014;9:e83704.
    1. Cockburn IA, Seder RA. Malaria prevention: from immunological concepts to effective vaccines and protective antibodies. Nat. Immunol. 2018;19:1199–1211.
    1. White MT, et al. Dynamics of the antibody response to Plasmodium falciparum infection in African children. J. Infect. Dis. 2014;210:1115–1122.
    1. Kinyanjui SM, Conway DJ, Lanar DE, Marsh K. IgG antibody responses to Plasmodium falciparum merozoite antigens in Kenyan children have a short half-life. Malar. J. 2007;6:82.
    1. Krishnamurty AT, et al. Somatically hypermutated Plasmodium-specific IgM(+) memory B cells are rapid, plastic, early responders upon malaria rechallenge. Immunity. 2016;45:402–414.
    1. Zenklusen I, et al. Immunization of malaria-preexposed volunteers with PfSPZ vaccine elicits long-lived IgM invasion-inhibitory and complement-fixing antibodies. J. Infect. Dis. 2018;217:1569–1578.
    1. Duncan CJ, Hill AV, Ellis RD. Can growth inhibition assays (GIA) predict blood-stage malaria vaccine efficacy? Hum. Vaccin. Immunother. 2012;8:706–714.
    1. Corran PH, et al. The fine specificity, but not the invasion inhibitory activity, of 19-kilodalton merozoite surface protein 1-specific antibodies is associated with resistance to malarial parasitemia in a cross-sectional survey in The Gambia. Infect. Immun. 2004;72:6185–6189.
    1. Perraut R, et al. Antibodies to the conserved C-terminal domain of the Plasmodium falciparum merozoite surface protein 1 and to the merozoite extract and their relationship with in vitro inhibitory antibodies and protection against clinical malaria in a Senegalese village. J. Infect. Dis. 2005;191:264–271.
    1. Reiling L, et al. Targets of complement-fixing antibodies in protective immunity against malaria in children. Nat. Commun. 2019;10:610.
    1. Murungi LM, et al. A threshold concentration of anti-merozoite antibodies is required for protection from clinical episodes of malaria. Vaccine. 2013;31:3936–3942.
    1. Blackman MJ, Scott-Finnigan TJ, Shai S, Holder AA. Antibodies inhibit the protease-mediated processing of a malaria merozoite surface protein. J. Exp. Med. 1994;180:389–393.
    1. Kadekoppala M, Holder AA. Merozoite surface proteins of the malaria parasite: the MSP1 complex and the MSP7 family. Int. J. Parasitol. 2010;40:1155–1161.
    1. Kapelski S, Klockenbring T, Fischer R, Barth S, Fendel R. Assessment of the neutrophilic antibody-dependent respiratory burst (ADRB) response to Plasmodium falciparum. J. Leukoc. Biol. 2014;96:1131–1142.
    1. Greve B, Lehman LG, Lell B, Luckner D, Schmidt-Ott R, Kremsner PG. High oxygen radical production is associated with fast parasite clearance in children with Plasmodium falciparum malaria. J. Infect. Dis. 1999;179:1584–1586.
    1. Osier FH, et al. Opsonic phagocytosis of Plasmodium falciparum merozoites: mechanism in human immunity and a correlate of protection against malaria. BMC Med. 2014;12:108.
    1. Hill DL, et al. Opsonising antibodies to P. falciparum merozoites associated with immunity to clinical malaria. PLoS ONE. 2013;8:e74627.
    1. Hill DL, et al. Merozoite antigens of Plasmodium falciparum elicit strain-transcending opsonizing immunity. Infect. Immun. 2016;84:2175–2184.
    1. Tiendrebeogo RW, et al. Antibody-dependent cellular inhibition is associated with reduced risk against febrile malaria in a longitudinal cohort study involving Ghanaian children. Open Forum Infect. Dis. 2015;2:ofv044.
    1. Chiu CY, et al. Antibodies to the Plasmodium falciparum proteins MSPDBL1 and MSPDBL2 opsonize merozoites, inhibit parasite growth, and predict protection from clinical malaria. J. Infect. Dis. 2015;212:406–415.
    1. Arora, G. et al. NK cells inhibit Plasmodium falciparum growth in red blood cells via antibody-dependent cellular cytotoxicity. Elife7, e36806 (2018).
    1. Bouharoun-Tayoun H, Oeuvray C, Lunel F, Druilhe P. Mechanisms underlying the monocyte-mediated antibody-dependent killing of Plasmodium falciparum asexual blood stages. J. Exp. Med. 1995;182:409–418.
    1. Bouharoun-Tayoun H, Attanath P, Sabchareon A, Chongsuphajaisiddhi T, Druilhe P. Antibodies that protect humans against Plasmodium falciparum blood stages do not on their own inhibit parasite growth and invasion in vitro, but act in cooperation with monocytes. J. Exp. Med. 1990;172:1633–1641.
    1. Soon MSF, Haque A. Recent insights into CD4(+) Th cell differentiation in malaria. J. Immunol. 2018;200:1965–1975.
    1. Wolf AS, Sherratt S, Riley EM. NK cells: uncertain allies against malaria. Front. Immunol. 2017;8:212.
    1. Food and Drug Administration. Guidance for Industry: Submitting Study Datasets for Vaccines to the Office of Vaccines Research and Review, 2018 (U.S. Department of Health and Human Services, accessed 27 August 2019); .
    1. Committee for Medicinal Products for Human Use. Guideline on the Clinical Evaluation of New Vaccines, 2007 (European Medicines Agency, Accessed 27 August 2019); .
    1. Kauth CW, Epp C, Bujard H, Lutz R. The merozoite surface protein 1 complex of human malaria parasite Plasmodium falciparum: interactions and arrangements of subunits. J. Biol. Chem. 2003;278:22257–22264.
    1. Kauth CW, et al. Interactions between merozoite surface proteins 1, 6, and 7 of the malaria parasite Plasmodium falciparum. J. Biol. Chem. 2006;281:31517–31527.
    1. Feng G, et al. Human immunization with a polymorphic malaria vaccine candidate induced antibodies to conserved epitopes that promote functional antibodies to multiple parasite strains. J. Infect. Dis. 2018;218:35–43.
    1. Lambros C, Vanderberg JP. Synchronization of Plasmodium falciparum erythrocytic stages in culture. J. Parasitol. 1979;65:418–420.
    1. Haley CS, Knott SA. A simple regression method for mapping quantitative trait loci in line crosses using flanking markers. Heredity. 1992;69:315–324.
    1. Bryan, D., Silva, N., Rigsby, P., Dougall, T. & Ho, M. M. The establishment of a WHO Reference Reagent for anti-malaria (Plasmodium falciparum) human serum. Malar J.16, 314 (2017).
    1. Dutta S, Haynes JD, Moch JK, Barbosa A, Lanar DE. Invasion-inhibitory antibodies inhibit proteolytic processing of apical membrane antigen 1 of Plasmodium falciparum merozoites. Proc. Natl Acad. Sci. USA. 2003;100:12295–12300.

Source: PubMed

3
S'abonner