Reduced blood-stage malaria growth and immune correlates in humans following RH5 vaccination

Angela M Minassian, Sarah E Silk, Jordan R Barrett, Carolyn M Nielsen, Kazutoyo Miura, Ababacar Diouf, Carolin Loos, Jonathan K Fallon, Ashlin R Michell, Michael T White, Nick J Edwards, Ian D Poulton, Celia H Mitton, Ruth O Payne, Michael Marks, Hector Maxwell-Scott, Antonio Querol-Rubiera, Karen Bisnauthsing, Rahul Batra, Tatiana Ogrina, Nathan J Brendish, Yrene Themistocleous, Thomas A Rawlinson, Katherine J Ellis, Doris Quinkert, Megan Baker, Raquel Lopez Ramon, Fernando Ramos Lopez, Lea Barfod, Pedro M Folegatti, Daniel Silman, Mehreen Datoo, Iona J Taylor, Jing Jin, David Pulido, Alexander D Douglas, Willem A de Jongh, Robert Smith, Eleanor Berrie, Amy R Noe, Carter L Diggs, Lorraine A Soisson, Rebecca Ashfield, Saul N Faust, Anna L Goodman, Alison M Lawrie, Fay L Nugent, Galit Alter, Carole A Long, Simon J Draper, Angela M Minassian, Sarah E Silk, Jordan R Barrett, Carolyn M Nielsen, Kazutoyo Miura, Ababacar Diouf, Carolin Loos, Jonathan K Fallon, Ashlin R Michell, Michael T White, Nick J Edwards, Ian D Poulton, Celia H Mitton, Ruth O Payne, Michael Marks, Hector Maxwell-Scott, Antonio Querol-Rubiera, Karen Bisnauthsing, Rahul Batra, Tatiana Ogrina, Nathan J Brendish, Yrene Themistocleous, Thomas A Rawlinson, Katherine J Ellis, Doris Quinkert, Megan Baker, Raquel Lopez Ramon, Fernando Ramos Lopez, Lea Barfod, Pedro M Folegatti, Daniel Silman, Mehreen Datoo, Iona J Taylor, Jing Jin, David Pulido, Alexander D Douglas, Willem A de Jongh, Robert Smith, Eleanor Berrie, Amy R Noe, Carter L Diggs, Lorraine A Soisson, Rebecca Ashfield, Saul N Faust, Anna L Goodman, Alison M Lawrie, Fay L Nugent, Galit Alter, Carole A Long, Simon J Draper

Abstract

Background: Development of an effective vaccine against the pathogenic blood-stage infection of human malaria has proved challenging, and no candidate vaccine has affected blood-stage parasitemia following controlled human malaria infection (CHMI) with blood-stage Plasmodium falciparum.

Methods: We undertook a phase I/IIa clinical trial in healthy adults in the United Kingdom of the RH5.1 recombinant protein vaccine, targeting the P. falciparum reticulocyte-binding protein homolog 5 (RH5), formulated in AS01B adjuvant. We assessed safety, immunogenicity, and efficacy against blood-stage CHMI. Trial registered at ClinicalTrials.gov, NCT02927145.

Findings: The RH5.1/AS01B formulation was administered using a range of RH5.1 protein vaccine doses (2, 10, and 50 μg) and was found to be safe and well tolerated. A regimen using a delayed and fractional third dose, in contrast to three doses given at monthly intervals, led to significantly improved antibody response longevity over ∼2 years of follow-up. Following primary and secondary CHMI of vaccinees with blood-stage P. falciparum, a significant reduction in parasite growth rate was observed, defining a milestone for the blood-stage malaria vaccine field. We show that growth inhibition activity measured in vitro using purified immunoglobulin G (IgG) antibody strongly correlates with in vivo reduction of the parasite growth rate and also identify other antibody feature sets by systems serology, including the plasma anti-RH5 IgA1 response, that are associated with challenge outcome.

Conclusions: Our data provide a new framework to guide rational design and delivery of next-generation vaccines to protect against malaria disease.

Funding: This study was supported by USAID, UK MRC, Wellcome Trust, NIAID, and the NIHR Oxford-BRC.

Keywords: CHMI; Plasmodium falciparum; RH5; blood-stage; clinical trial; malaria; systems serology; vaccine.

Conflict of interest statement

A.D.D. and S.J.D. are named inventors on patent applications relating to RH5 and/or other malaria vaccines and immunization regimens. W.A.d.J. is an employee of and shareholder in ExpreS2ion Biotechnologies, which has developed and is marketing the ExpreS2 cell expression platform. A.R.N. is an employee of Leidos, Inc., which holds the MVDP prime contract (AID-OAA-C-15-00071). A.M.M. has an immediate family member who is an inventor on patents relating to RH5 and/or other malaria vaccines and immunization regimens.

© 2021 The Author(s).

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Antibody immunogenicity of RH5.1/AS01B (A) Timing of immunizations and follow-up in groups 1–4. All antigen doses were formulated in 0.5 mL AS01B. (B and C) Median and individual anti-RH5_FL serum total IgG responses 14 days after two vaccinations (Vacs; day 42, B) and after three Vacs (day 70 or day 196, C). Both datasets were analyzed separately by Kruskal-Wallis test with Dunn’s multiple comparisons test; ∗∗p In vitro GIA of purified IgG assessed at 10 mg/mL against 3D7 clone P. falciparum parasites. Individual data and medians are shown for each group at the stated time-point; pooled sera were used for each group at baseline (day 0). Historical data for VV-RH5 were included as before. (E) Dilution series of purified IgG for all group 1–4 samples starting from 10 mg/mL. (F) Relationship between GIA data from the dilution series shown in (E) and concentration of anti-RH5_FL purified IgG used in the assay as measured by ELISA. A non-linear regression curve is shown for all samples combined (solid line, r2 = 0.96, n = 279). The EC50 (concentration of anti-RH5_FL polyclonal IgG that gives 50% GIA, dashed line) was calculated.
Figure 2
Figure 2
Assessment of the DFx regimen (A) Median anti-RH5_FL serum total IgG responses for groups 1–4 over time. Individual responses are shown in Figure S4B. (B) Median and individual anti-RH5_FL serum total IgG responses at the time of the late bleed. Statistical analysis was performed using a Mann-Whitney test, ∗∗∗∗p 50). Individual responses over time are shown in Figure S4C. Historical data for the VV-RH5 vaccine are shown for comparison. Kruskal-Wallis test with Dunn’s multiple comparison test, ∗∗∗p < 0.001 for group 3 versus groups 1, 2, and 4. (D) Estimated proportion of antibodies generated from LLPCs; for group 3, it was possible to provide separate estimates of the proportion following the first and second doses (purple) and following the third dose (pink). The long-lived response following the third dose in group 3 is significantly greater than in other groups; ∗∗∗∗p −6 in all cases by one-sided t test. (E) Anti-RH5_FL antibody levels at peak and 1 and 4 years following the third vaccine dose. Peak antibody levels are based on the maximum measured values (day 70 or day 84 for groups 1, 2, and 4 and on day 196 or day 210 for group 3). Antibody levels at 1 and 4 years are based on model estimates and are presented with 95% CI.
Figure 3
Figure 3
Results of primary and secondary blood-stage CHMI (A) Timing of immunizations, CHMIs, and follow-up in groups 5–9. All antigen doses were formulated in 0.5 mL AS01B. (B) qPCR data for the VAC063A phase IIa study; group 5 (n = 14) and group 6 (n = 15). Median parasitemia is shown over time for each group. The lower limit of quantification is indicated by the dotted line at 20 p/mL; values below this level are plotted for information only. Time = days after blood-stage CHMI. (C) Primary efficacy endpoint analysis of PMR, showing each individual plus the mean. Both datasets are normally distributed (D’Agostino-Pearson test); ∗p = 0.031 using two-tailed t test with Welch’s correction for non-equal variances (F test; p = 0.008). (D) Kaplan-Meier plot of time to diagnosis in days for the VAC063A study. Median time to patent parasitemia was 9.5 days for control individuals and 10.5 days for vaccinees. Secondary pre-specified efficacy analysis in the protocol compared time to diagnosis between the groups; p = 0.01, Mann-Whitney test. (E) Post hoc analysis combining the VAC063A dataset with the AMA1/AS01B trial (VAC054) data. Mean PMR ± 95% CI is shown for control individuals (n = 15 from VAC063A and n = 15 from VAC054), AMA1 vaccinees (n = 12 from VAC054), and RH5 vaccinees (n = 14 from VAC063A). ∗p < 0.05 for RH5 versus AMA1 and control individuals, using one-way ANOVA with Bonferroni correction for multiple comparisons. (F) qPCR data for the VAC063B phase IIa study shown as in (B); group 7 (n = 9), group 8 (n = 8), and group 9 (n = 6). (G) Secondary efficacy endpoint analysis of PMR, showing each individual, plus the median. ∗p = 0.022; Kruskal-Wallis test with Dunn’s multiple comparison test.
Figure 4
Figure 4
Antibody responses after CHMI and the fourth booster immunization (A) Median and individual anti-RH5_FL serum total IgG responses shown for groups 5 and 7 over time (note that a subset of group 5 vaccinees became group 7; Figure S6). Gray shading indicates periods of CHMI, and arrows indicate Vacs. The legend indicates the sequence of 10-μg RH5.1/AS01B Vacs as well as the first and second CHMIs, as relevant to each group. (B–E) Individual and median responses are shown for the indicated groups and time points. dC, day of challenge. ∗p 

Figure 5

Analysis of in vitro GIA…

Figure 5

Analysis of in vitro GIA versus IVGI (A) In vitro GIA of purified…

Figure 5
Analysis of in vitro GIA versus IVGI (A) In vitro GIA of purified IgG assessed at 10 mg/mL against 3D7 clone P. falciparum parasites. Individual data and medians are shown for each group (Figure 3A) at the stated time points. (B) Relationship between GIA and concentration of anti-RH5_FL purified IgG used in the assay, as measured by ELISA. A non-linear regression curve is shown for all samples combined (solid line, r2 = 0.97, n = 180). The EC50 (dashed line) was calculated. (C) In vitro GIA as in (A), using purified IgG assessed at 2.5 mg/mL. Historical data from the AMA1/AS01B (VAC054) trial are included. The median percent IVGI observed in each group following blood-stage CHMI is indicated below the graph. The red dashed line at 60% GIA indicates the threshold level required for protection in Aotus monkeys., ∗p < 0.05, ∗∗p < 0.01; Kruskal-Wallis test with Dunn’s multiple comparisons test. (D) Correlation of % IVGI observed in each individual following blood-stage CHMI versus their individual in vitro GIA measured at dC−1 using 2.5 mg/mL purified IgG. Spearman’s rank correlation coefficient and p value are shown; n = 35. Colored symbols are the same as those used in (C) for AMA1, group 5 (G5), and G7.

Figure 6

Systems serology analysis of CHMI…

Figure 6

Systems serology analysis of CHMI outcome in RH5 vaccinees (A) Correlation heatmap showing…

Figure 6
Systems serology analysis of CHMI outcome in RH5 vaccinees (A) Correlation heatmap showing the Spearman rank correlation coefficients (rS) between Fc functions and titers as well as the rank correlation of the features to the CHMI readouts of DOD and IVGI. ∗q < 0.1, ∗∗q < 0.01 (Benjamini-Hochberg procedure for multiple testing correction; the correction was done within the groups of comparison; 54 comparisons for Fc functions/titer, 40 for features/DOD, and 40 for features/IVGI). (B) Scatterplot to show the relationship between ADNP score and IVGI. Each color corresponds to one volunteer, and the shape indicates the group: G5 (n = 13) or G7 (n = 7). (C) Prediction for the random forest regression model plotted against the data for DOD. The model was obtained using leave-one-volunteer-out cross-validation. The Pearson correlation coefficient (rP) and p value are shown. (D) The antibody features in the predictive model for DOD are ranked according to how often they were chosen for 100 repetitions of recursive feature elimination (RFE) in the leave-one-volunteer-out cross-validation. (E) Graphs showing DOD versus systems serology assay data for the seven antibody features that were chosen in more than 10% of the elimination procedures (D). (F) The co-correlates network shows the pairwise correlation of features. The nodes correspond to features and the edges to Spearman rank correlations between the features. Only significant correlations (Benjamini-Hochberg q 
All figures (7)
Comment in
Similar articles
Cited by
References
    1. World Health Organization . 2019. World Malaria Report (2019)https://www.who.int/publications/i/item/9789241565721
    1. Moorthy V.S., Newman R.D., Okwo-Bele J.M. Malaria vaccine technology roadmap. Lancet. 2013;382:1700–1701. - PubMed
    1. Draper S.J., Sack B.K., King C.R., Nielsen C.M., Rayner J.C., Higgins M.K., Long C.A., Seder R.A. Malaria Vaccines: Recent Advances and New Horizons. Cell Host Microbe. 2018;24:43–56. - PMC - PubMed
    1. Takala S.L., Coulibaly D., Thera M.A., Batchelor A.H., Cummings M.P., Escalante A.A., Ouattara A., Traoré K., Niangaly A., Djimdé A.A. Extreme polymorphism in a vaccine antigen and risk of clinical malaria: implications for vaccine development. Sci. Transl. Med. 2009;1:2ra5. - PMC - PubMed
    1. Wright G.J., Rayner J.C. Plasmodium falciparum erythrocyte invasion: combining function with immune evasion. PLoS Pathog. 2014;10:e1003943. - PMC - PubMed
Show all 77 references
Publication types
Associated data
Related information
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 5
Figure 5
Analysis of in vitro GIA versus IVGI (A) In vitro GIA of purified IgG assessed at 10 mg/mL against 3D7 clone P. falciparum parasites. Individual data and medians are shown for each group (Figure 3A) at the stated time points. (B) Relationship between GIA and concentration of anti-RH5_FL purified IgG used in the assay, as measured by ELISA. A non-linear regression curve is shown for all samples combined (solid line, r2 = 0.97, n = 180). The EC50 (dashed line) was calculated. (C) In vitro GIA as in (A), using purified IgG assessed at 2.5 mg/mL. Historical data from the AMA1/AS01B (VAC054) trial are included. The median percent IVGI observed in each group following blood-stage CHMI is indicated below the graph. The red dashed line at 60% GIA indicates the threshold level required for protection in Aotus monkeys., ∗p < 0.05, ∗∗p < 0.01; Kruskal-Wallis test with Dunn’s multiple comparisons test. (D) Correlation of % IVGI observed in each individual following blood-stage CHMI versus their individual in vitro GIA measured at dC−1 using 2.5 mg/mL purified IgG. Spearman’s rank correlation coefficient and p value are shown; n = 35. Colored symbols are the same as those used in (C) for AMA1, group 5 (G5), and G7.
Figure 6
Figure 6
Systems serology analysis of CHMI outcome in RH5 vaccinees (A) Correlation heatmap showing the Spearman rank correlation coefficients (rS) between Fc functions and titers as well as the rank correlation of the features to the CHMI readouts of DOD and IVGI. ∗q < 0.1, ∗∗q < 0.01 (Benjamini-Hochberg procedure for multiple testing correction; the correction was done within the groups of comparison; 54 comparisons for Fc functions/titer, 40 for features/DOD, and 40 for features/IVGI). (B) Scatterplot to show the relationship between ADNP score and IVGI. Each color corresponds to one volunteer, and the shape indicates the group: G5 (n = 13) or G7 (n = 7). (C) Prediction for the random forest regression model plotted against the data for DOD. The model was obtained using leave-one-volunteer-out cross-validation. The Pearson correlation coefficient (rP) and p value are shown. (D) The antibody features in the predictive model for DOD are ranked according to how often they were chosen for 100 repetitions of recursive feature elimination (RFE) in the leave-one-volunteer-out cross-validation. (E) Graphs showing DOD versus systems serology assay data for the seven antibody features that were chosen in more than 10% of the elimination procedures (D). (F) The co-correlates network shows the pairwise correlation of features. The nodes correspond to features and the edges to Spearman rank correlations between the features. Only significant correlations (Benjamini-Hochberg q 
All figures (7)

References

    1. World Health Organization . 2019. World Malaria Report (2019)
    1. Moorthy V.S., Newman R.D., Okwo-Bele J.M. Malaria vaccine technology roadmap. Lancet. 2013;382:1700–1701.
    1. Draper S.J., Sack B.K., King C.R., Nielsen C.M., Rayner J.C., Higgins M.K., Long C.A., Seder R.A. Malaria Vaccines: Recent Advances and New Horizons. Cell Host Microbe. 2018;24:43–56.
    1. Takala S.L., Coulibaly D., Thera M.A., Batchelor A.H., Cummings M.P., Escalante A.A., Ouattara A., Traoré K., Niangaly A., Djimdé A.A. Extreme polymorphism in a vaccine antigen and risk of clinical malaria: implications for vaccine development. Sci. Transl. Med. 2009;1:2ra5.
    1. Wright G.J., Rayner J.C. Plasmodium falciparum erythrocyte invasion: combining function with immune evasion. PLoS Pathog. 2014;10:e1003943.
    1. Ragotte R.J., Higgins M.K., Draper S.J. The RH5-CyRPA-Ripr Complex as a Malaria Vaccine Target. Trends Parasitol. 2020;36:545–559.
    1. Crosnier C., Bustamante L.Y., Bartholdson S.J., Bei A.K., Theron M., Uchikawa M., Mboup S., Ndir O., Kwiatkowski D.P., Duraisingh M.T. Basigin is a receptor essential for erythrocyte invasion by Plasmodium falciparum. Nature. 2011;480:534–537.
    1. Galaway F., Yu R., Constantinou A., Prugnolle F., Wright G.J. Resurrection of the ancestral RH5 invasion ligand provides a molecular explanation for the origin of P. falciparum malaria in humans. PLoS Biol. 2019;17:e3000490.
    1. Douglas A.D., Williams A.R., Illingworth J.J., Kamuyu G., Biswas S., Goodman A.L., Wyllie D.H., Crosnier C., Miura K., Wright G.J. The blood-stage malaria antigen PfRH5 is susceptible to vaccine-inducible cross-strain neutralizing antibody. Nat. Commun. 2011;2:601.
    1. Osier F.H., Mackinnon M.J., Crosnier C., Fegan G., Kamuyu G., Wanaguru M., Ogada E., McDade B., Rayner J.C., Wright G.J., Marsh K. New antigens for a multicomponent blood-stage malaria vaccine. Sci. Transl. Med. 2014;6:247ra102.
    1. Payne R.O., Silk S.E., Elias S.C., Miura K., Diouf A., Galaway F., de Graaf H., Brendish N.J., Poulton I.D., Griffiths O.J. Human vaccination against RH5 induces neutralizing antimalarial antibodies that inhibit RH5 invasion complex interactions. JCI Insight. 2017;2:96381.
    1. Tran T.M., Ongoiba A., Coursen J., Crosnier C., Diouf A., Huang C.Y., Li S., Doumbo S., Doumtabe D., Kone Y. Naturally acquired antibodies specific for Plasmodium falciparum reticulocyte-binding protein homologue 5 inhibit parasite growth and predict protection from malaria. J. Infect. Dis. 2014;209:789–798.
    1. Williams A.R., Douglas A.D., Miura K., Illingworth J.J., Choudhary P., Murungi L.M., Furze J.M., Diouf A., Miotto O., Crosnier C. Enhancing blockade of Plasmodium falciparum erythrocyte invasion: assessing combinations of antibodies against PfRH5 and other merozoite antigens. PLoS Pathog. 2012;8:e1002991.
    1. Bustamante L.Y., Bartholdson S.J., Crosnier C., Campos M.G., Wanaguru M., Nguon C., Kwiatkowski D.P., Wright G.J., Rayner J.C. A full-length recombinant Plasmodium falciparum PfRH5 protein induces inhibitory antibodies that are effective across common PfRH5 genetic variants. Vaccine. 2013;31:373–379.
    1. Reddy K.S., Pandey A.K., Singh H., Sahar T., Emmanuel A., Chitnis C.E., Chauhan V.S., Gaur D. Bacterially expressed full-length recombinant Plasmodium falciparum RH5 protein binds erythrocytes and elicits potent strain-transcending parasite-neutralizing antibodies. Infect. Immun. 2014;82:152–164.
    1. Miura K., Zhou H., Diouf A., Moretz S.E., Fay M.P., Miller L.H., Martin L.B., Pierce M.A., Ellis R.D., Mullen G.E., Long C.A. Anti-apical-membrane-antigen-1 antibody is more effective than anti-42-kilodalton-merozoite-surface-protein-1 antibody in inhibiting plasmodium falciparum growth, as determined by the in vitro growth inhibition assay. Clin. Vaccine Immunol. 2009;16:963–968.
    1. Douglas A.D., Baldeviano G.C., Lucas C.M., Lugo-Roman L.A., Crosnier C., Bartholdson S.J., Diouf A., Miura K., Lambert L.E., Ventocilla J.A. A PfRH5-based vaccine is efficacious against heterologous strain blood-stage Plasmodium falciparum infection in aotus monkeys. Cell Host Microbe. 2015;17:130–139.
    1. Payne R.O., Milne K.H., Elias S.C., Edwards N.J., Douglas A.D., Brown R.E., Silk S.E., Biswas S., Miura K., Roberts R. Demonstration of the Blood-Stage Plasmodium falciparum Controlled Human Malaria Infection Model to Assess Efficacy of the P. falciparum Apical Membrane Antigen 1 Vaccine, FMP2.1/AS01. J. Infect. Dis. 2016;213:1743–1751.
    1. Hjerrild K.A., Jin J., Wright K.E., Brown R.E., Marshall J.M., Labbé G.M., Silk S.E., Cherry C.J., Clemmensen S.B., Jørgensen T. Production of full-length soluble Plasmodium falciparum RH5 protein vaccine using a Drosophila melanogaster Schneider 2 stable cell line system. Sci. Rep. 2016;6:30357.
    1. Jin J., Hjerrild K.A., Silk S.E., Brown R.E., Labbé G.M., Marshall J.M., Wright K.E., Bezemer S., Clemmensen S.B., Biswas S. Accelerating the clinical development of protein-based vaccines for malaria by efficient purification using a four amino acid C-terminal ‘C-tag’. Int. J. Parasitol. 2017;47:435–446.
    1. Jin J., Tarrant R.D., Bolam E.J., Angell-Manning P., Soegaard M., Pattinson D.J., Dulal P., Silk S.E., Marshall J.M., Dabbs R.A. Production, quality control, stability, and potency of cGMP-produced Plasmodium falciparum RH5.1 protein vaccine expressed in Drosophila S2 cells. NPJ Vaccines. 2018;3:32.
    1. Rampling T., Ewer K.J., Bowyer G., Bliss C.M., Edwards N.J., Wright D., Payne R.O., Venkatraman N., de Barra E., Snudden C.M. Safety and High Level Efficacy of the Combination Malaria Vaccine Regimen of RTS,S/AS01B With Chimpanzee Adenovirus 63 and Modified Vaccinia Ankara Vectored Vaccines Expressing ME-TRAP. J. Infect. Dis. 2016;214:772–781.
    1. Mullen G.E., Ellis R.D., Miura K., Malkin E., Nolan C., Hay M., Fay M.P., Saul A., Zhu D., Rausch K. Phase 1 trial of AMA1-C1/Alhydrogel plus CPG 7909: an asexual blood-stage vaccine for Plasmodium falciparum malaria. PLoS ONE. 2008;3:e2940.
    1. Hodgson S.H., Choudhary P., Elias S.C., Milne K.H., Rampling T.W., Biswas S., Poulton I.D., Miura K., Douglas A.D., Alanine D.G. Combining viral vectored and protein-in-adjuvant vaccines against the blood-stage malaria antigen AMA1: report on a phase 1a clinical trial. Mol. Ther. 2014;22:2142–2154.
    1. Duncan C.J., Sheehy S.H., Ewer K.J., Douglas A.D., Collins K.A., Halstead F.D., Elias S.C., Lillie P.J., Rausch K., Aebig J. Impact on malaria parasite multiplication rates in infected volunteers of the protein-in-adjuvant vaccine AMA1-C1/Alhydrogel+CPG 7909. PLoS ONE. 2011;6:e22271.
    1. Koutsakos M., Nguyen T.H.O., Kedzierska K. With a Little Help from T Follicular Helper Friends: Humoral Immunity to Influenza Vaccination. J. Immunol. 2019;202:360–367.
    1. Douglas A.D., Edwards N.J., Duncan C.J., Thompson F.M., Sheehy S.H., O’Hara G.A., Anagnostou N., Walther M., Webster D.P., Dunachie S.J. Comparison of modeling methods to determine liver-to-blood inocula and parasite multiplication rates during controlled human malaria infection. J. Infect. Dis. 2013;208:340–345.
    1. Villasis E., Lopez-Perez M., Torres K., Gamboa D., Neyra V., Bendezu J., Tricoche N., Lobo C., Vinetz J.M., Lustigman S. Anti-Plasmodium falciparum invasion ligand antibodies in a low malaria transmission region, Loreto, Peru. Malar. J. 2012;11:361.
    1. Hodgson S.H., Llewellyn D., Silk S.E., Milne K.H., Elias S.C., Miura K., Kamuyu G., Juma E.A., Magiri C., Muia A. Changes in Serological Immunology Measures in UK and Kenyan Adults Post-controlled Human Malaria Infection. Front. Microbiol. 2016;7:1604.
    1. Singh S., Miura K., Zhou H., Muratova O., Keegan B., Miles A., Martin L.B., Saul A.J., Miller L.H., Long C.A. Immunity to recombinant plasmodium falciparum merozoite surface protein 1 (MSP1): protection in Aotus nancymai monkeys strongly correlates with anti-MSP1 antibody titer and in vitro parasite-inhibitory activity. Infect. Immun. 2006;74:4573–4580.
    1. Mahdi Abdel Hamid M., Remarque E.J., van Duivenvoorde L.M., van der Werff N., Walraven V., Faber B.W., Kocken C.H., Thomas A.W. Vaccination with Plasmodium knowlesi AMA1 formulated in the novel adjuvant co-vaccine HT™ protects against blood-stage challenge in rhesus macaques. PLoS ONE. 2011;6:e20547.
    1. Chung A.W., Kumar M.P., Arnold K.B., Yu W.H., Schoen M.K., Dunphy L.J., Suscovich T.J., Frahm N., Linde C., Mahan A.E. Dissecting Polyclonal Vaccine-Induced Humoral Immunity against HIV Using Systems Serology. Cell. 2015;163:988–998.
    1. Chung A.W., Alter G. Systems serology: profiling vaccine induced humoral immunity against HIV. Retrovirology. 2017;14:57.
    1. Jennewein M.F., Alter G. The Immunoregulatory Roles of Antibody Glycosylation. Trends Immunol. 2017;38:358–372.
    1. Sagara I., Dicko A., Ellis R.D., Fay M.P., Diawara S.I., Assadou M.H., Sissoko M.S., Kone M., Diallo A.I., Saye R. A randomized controlled phase 2 trial of the blood stage AMA1-C1/Alhydrogel malaria vaccine in children in Mali. Vaccine. 2009;27:3090–3098.
    1. Ogutu B.R., Apollo O.J., McKinney D., Okoth W., Siangla J., Dubovsky F., Tucker K., Waitumbi J.N., Diggs C., Wittes J., MSP-1 Malaria Vaccine Working Group 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. Thera M.A., Doumbo O.K., Coulibaly D., Laurens M.B., Ouattara A., Kone A.K., Guindo A.B., Traore K., Traore I., Kouriba B. A field trial to assess a blood-stage malaria vaccine. N. Engl. J. Med. 2011;365:1004–1013.
    1. Sirima S.B., Mordmüller B., Milligan P., Ngoa U.A., Kironde F., Atuguba F., Tiono A.B., Issifou S., Kaddumukasa M., Bangre O., GMZ2 Trial Study Group A phase 2b randomized, controlled trial of the efficacy of the GMZ2 malaria vaccine in African children. Vaccine. 2016;34:4536–4542.
    1. Dejon-Agobe J.C., Ateba-Ngoa U., Lalremruata A., Homoet A., Engelhorn J., Nouatin O.P., Edoa J.R., Fernandes J.F., Esen M., Mouwenda Y.D. Controlled Human Malaria Infection of Healthy Adults With Lifelong Malaria Exposure to Assess Safety, Immunogenicity, and Efficacy of the Asexual Blood Stage Malaria Vaccine Candidate GMZ2. Clin. Infect. Dis. 2019;69:1377–1384.
    1. Sheehy S.H., Duncan C.J., Elias S.C., Choudhary P., Biswas S., Halstead F.D., Collins K.A., Edwards N.J., Douglas A.D., Anagnostou N.A. 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. Spring M.D., Cummings J.F., Ockenhouse C.F., Dutta S., Reidler R., Angov E., Bergmann-Leitner E., Stewart V.A., Bittner S., Juompan L. Phase 1/2a study of the malaria vaccine candidate apical membrane antigen-1 (AMA-1) administered in adjuvant system AS01B or AS02A. PLoS ONE. 2009;4:e5254.
    1. Lawrence G., Cheng Q.Q., Reed C., Taylor D., Stowers A., Cloonan N., Rzepczyk C., Smillie A., Anderson K., Pombo D. Effect of vaccination with 3 recombinant asexual-stage malaria antigens on initial growth rates of Plasmodium falciparum in non-immune volunteers. Vaccine. 2000;18:1925–1931.
    1. Douglas A.D., Andrews L., Draper S.J., Bojang K., Milligan P., Gilbert S.C., Imoukhuede E.B., Hill A.V. Substantially reduced pre-patent parasite multiplication rates are associated with naturally acquired immunity to Plasmodium falciparum. J. Infect. Dis. 2011;203:1337–1340.
    1. Hodgson S.H., Juma E., Salim A., Magiri C., Kimani D., Njenga D., Muia A., Cole A.O., Ogwang C., Awuondo K. Evaluating controlled human malaria infection in Kenyan adults with varying degrees of prior exposure to Plasmodium falciparum using sporozoites administered by intramuscular injection. Front. Microbiol. 2014;5:686.
    1. Achan J., Reuling I., Yap X.Z., Dabira E., Ahmad A., Cox M., Nwakanma D., Tetteh K., Wu L., Bastiaens G.J.H. Serologic markers of previous malaria exposure and functional antibodies inhibiting parasite growth are associated with parasite kinetics following a Plasmodium falciparum controlled human infection. Clin. Infect. Dis. 2020;70:2544–2552.
    1. Douglas A.D., Baldeviano G.C., Jin J., Miura K., Diouf A., Zenonos Z.A., Ventocilla J.A., Silk S.E., Marshall J.M., Alanine D.G.W. A defined mechanistic correlate of protection against Plasmodium falciparum malaria in non-human primates. Nat. Commun. 2019;10:1953.
    1. Foquet L., Schafer C., Minkah N.K., Alanine D.G.W., Flannery E.L., Steel R.W.J., Sack B.K., Camargo N., Fishbaugher M., Betz W. Plasmodium falciparum Liver Stage Infection and Transition to Stable Blood Stage Infection in Liver-Humanized and Blood-Humanized FRGN KO Mice Enables Testing of Blood Stage Inhibitory Antibodies (Reticulocyte-Binding Protein Homolog 5) In Vivo. Front. Immunol. 2018;9:524.
    1. Joos C., Marrama L., Polson H.E., Corre S., Diatta A.M., Diouf B., Trape J.F., Tall A., Longacre S., Perraut R. Clinical protection from falciparum malaria correlates with neutrophil respiratory bursts induced by merozoites opsonized with human serum antibodies. PLoS ONE. 2010;5:e9871.
    1. Bliss C.M., Drammeh A., Bowyer G., Sanou G.S., Jagne Y.J., Ouedraogo O., Edwards N.J., Tarama C., Ouedraogo N., Ouedraogo M. Viral Vector Malaria Vaccines Induce High-Level T Cell and Antibody Responses in West African Children and Infants. Mol. Ther. 2017;25:547–559.
    1. Rts S.C.T.P., RTS,S Clinical Trials Partnership Efficacy and safety of the RTS,S/AS01 malaria vaccine during 18 months after vaccination: a phase 3 randomized, controlled trial in children and young infants at 11 African sites. PLoS Med. 2014;11:e1001685.
    1. Leroux-Roels G., Leroux-Roels I., Clement F., Ofori-Anyinam O., Lievens M., Jongert E., Moris P., Ballou W.R., Cohen J. Evaluation of the immune response to RTS,S/AS01 and RTS,S/AS02 adjuvanted vaccines: randomized, double-blind study in malaria-naïve adults. Hum. Vaccin. Immunother. 2014;10:2211–2219.
    1. Azasi Y., Gallagher S.K., Diouf A., Dabbs R.A., Jin J., Mian S.Y., Narum D.L., Long C.A., Gaur D., Draper S.J. Bliss’ and Loewe’s additive and synergistic effects in Plasmodium falciparum growth inhibition by AMA1-RON2L, RH5, RIPR and CyRPA antibody combinations. Sci. Rep. 2020;10:11802.
    1. Alanine D.G.W., Quinkert D., Kumarasingha R., Mehmood S., Donnellan F.R., Minkah N.K., Dadonaite B., Diouf A., Galaway F., Silk S.E. Human Antibodies that Slow Erythrocyte Invasion Potentiate Malaria-Neutralizing Antibodies. Cell. 2019;178:216–228.e21.
    1. Campeotto I., Goldenzweig A., Davey J., Barfod L., Marshall J.M., Silk S.E., Wright K.E., Draper S.J., Higgins M.K., Fleishman S.J. One-step design of a stable variant of the malaria invasion protein RH5 for use as a vaccine immunogen. Proc. Natl. Acad. Sci. USA. 2017;114:998–1002.
    1. Marini A., Zhou Y., Li Y., Taylor I.J., Leneghan D.B., Jin J., Zaric M., Mekhaiel D., Long C.A., Miura K., Biswas S. A Universal Plug-and-Display Vaccine Carrier Based on HBsAg VLP to Maximize Effective Antibody Response. Front. Immunol. 2019;10:2931.
    1. Pérez-Mazliah D., Ndungu F.M., Aye R., Langhorne J. B-cell memory in malaria: Myths and realities. Immunol. Rev. 2020;293:57–69.
    1. Regules J.A., Cicatelli S.B., Bennett J.W., Paolino K.M., Twomey P.S., Moon J.E., Kathcart A.K., Hauns K.D., Komisar J.L., Qabar A.N. Fractional Third and Fourth Dose of RTS,S/AS01 Malaria Candidate Vaccine: A Phase 2a Controlled Human Malaria Parasite Infection and Immunogenicity Study. J. Infect. Dis. 2016;214:762–771.
    1. Chandramohan D., Dicko A., Zongo I., Sagara I., Cairns M., Kuepfer I., Diarra M., Tapily A., Issiaka D., Sanogo K. Seasonal malaria vaccination: protocol of a phase 3 trial of seasonal vaccination with the RTS,S/AS01E vaccine, seasonal malaria chemoprevention and the combination of vaccination and chemoprevention. BMJ Open. 2020;10:e035433.
    1. Pallikkuth S., Chaudhury S., Lu P., Pan L., Jongert E., Wille-Reece U., Pahwa S. A delayed fractionated dose RTS,S AS01 vaccine regimen mediates protection via improved T follicular helper and B cell responses. eLife. 2020;9:e51889.
    1. Cheng Q., Lawrence G., Reed C., Stowers A., Ranford-Cartwright L., Creasey A., Carter R., Saul A. Measurement of Plasmodium falciparum growth rates in vivo: a test of malaria vaccines. Am. J. Trop. Med. Hyg. 1997;57:495–500.
    1. Stoute J.A., Slaoui M., Heppner D.G., Momin P., Kester K.E., Desmons P., Wellde B.T., Garçon N., Krzych U., Marchand M. A preliminary evaluation of a recombinant circumsporozoite protein vaccine against Plasmodium falciparum malaria. RTS,S Malaria Vaccine Evaluation Group. N. Engl. J. Med. 1997;336:86–91.
    1. Joint United Kingdom (UK) Blood Transfusion and Tissue Transplantation Services Professional Advisory Committee . 2018. Whole Blood and Components; Donor Selection Guidelines.
    1. Engwerda C.R., Minigo G., Amante F.H., McCarthy J.S. Experimentally induced blood stage malaria infection as a tool for clinical research. Trends Parasitol. 2012;28:515–521.
    1. Duncan C.J., Draper S.J. Controlled human blood stage malaria infection: current status and potential applications. Am. J. Trop. Med. Hyg. 2012;86:561–565.
    1. Sheehy S.H., Duncan C.J., Elias S.C., Biswas S., Collins K.A., O’Hara G.A., Halstead F.D., Ewer K.J., Mahungu T., Spencer A.J. Phase Ia clinical evaluation of the safety and immunogenicity of the Plasmodium falciparum blood-stage antigen AMA1 in ChAd63 and MVA vaccine vectors. PLoS ONE. 2012;7:e31208.
    1. Nielsen C.M., Ogbe A., Pedroza-Pacheco I., Doeleman S.E., Chen Y., Silk S.E., Barrett J.R., Elias S.C., Miura K., Diouf A. Protein/AS01B vaccination elicits stronger, more Th2-skewed antigen-specific human T follicular helper cell responses than heterologous viral vectors. Cell Rep Med. 2021;2:100207.
    1. Malkin E.M., Diemert D.J., McArthur J.H., Perreault J.R., Miles A.P., Giersing B.K., Mullen G.E., Orcutt A., Muratova O., Awkal M. Phase 1 clinical trial of apical membrane antigen 1: an asexual blood-stage vaccine for Plasmodium falciparum malaria. Infect. Immun. 2005;73:3677–3685.
    1. Ackerman M.E., Moldt B., Wyatt R.T., Dugast A.S., McAndrew E., Tsoukas S., Jost S., Berger C.T., Sciaranghella G., Liu Q. A robust, high-throughput assay to determine the phagocytic activity of clinical antibody samples. J. Immunol. Methods. 2011;366:8–19.
    1. Karsten C.B., Mehta N., Shin S.A., Diefenbach T.J., Slein M.D., Karpinski W., Irvine E.B., Broge T., Suscovich T.J., Alter G. A versatile high-throughput assay to characterize antibody-mediated neutrophil phagocytosis. J. Immunol. Methods. 2019;471:46–56.
    1. Fischinger S., Fallon J.K., Michell A.R., Broge T., Suscovich T.J., Streeck H., Alter G. A high-throughput, bead-based, antigen-specific assay to assess the ability of antibodies to induce complement activation. J. Immunol. Methods. 2019;473:112630.
    1. Jegaskanda S., Job E.R., Kramski M., Laurie K., Isitman G., de Rose R., Winnall W.R., Stratov I., Brooks A.G., Reading P.C., Kent S.J. Cross-reactive influenza-specific antibody-dependent cellular cytotoxicity antibodies in the absence of neutralizing antibodies. J. Immunol. 2013;190:1837–1848.
    1. Lu L.L., Chung A.W., Rosebrock T.R., Ghebremichael M., Yu W.H., Grace P.S., Schoen M.K., Tafesse F., Martin C., Leung V. A Functional Role for Antibodies in Tuberculosis. Cell. 2016;167:433–443.e14.
    1. Brown E.P., Licht A.F., Dugast A.S., Choi I., Bailey-Kellogg C., Alter G., Ackerman M.E. High-throughput, multiplexed IgG subclassing of antigen-specific antibodies from clinical samples. J. Immunol. Methods. 2012;386:117–123.
    1. Brown E.P., Dowell K.G., Boesch A.W., Normandin E., Mahan A.E., Chu T., Barouch D.H., Bailey-Kellogg C., Alter G., Ackerman M.E. Multiplexed Fc array for evaluation of antigen-specific antibody effector profiles. J. Immunol. Methods. 2017;443:33–44.
    1. Brown E.P., Weiner J.A., Lin S., Natarajan H., Normandin E., Barouch D.H., Alter G., Sarzotti-Kelsoe M., Ackerman M.E. Optimization and qualification of an Fc Array assay for assessments of antibodies against HIV-1/SIV. J. Immunol. Methods. 2018;455:24–33.
    1. Mahan A.E., Tedesco J., Dionne K., Baruah K., Cheng H.D., De Jager P.L., Barouch D.H., Suscovich T., Ackerman M., Crispin M., Alter G. A method for high-throughput, sensitive analysis of IgG Fc and Fab glycosylation by capillary electrophoresis. J. Immunol. Methods. 2015;417:34–44.
    1. Brown E.P., Normandin E., Osei-Owusu N.Y., Mahan A.E., Chan Y.N., Lai J.I., Vaccari M., Rao M., Franchini G., Alter G., Ackerman M.E. Microscale purification of antigen-specific antibodies. J. Immunol. Methods. 2015;425:27–36.

Source: PubMed

3
구독하다