Transcriptional correlates of malaria in RTS,S/AS01-vaccinated African children: a matched case-control study

Gemma Moncunill, Jason Carnes, William Chad Young, Lindsay Carpp, Stephen De Rosa, Joseph J Campo, Augusto Nhabomba, Maxmillian Mpina, Chenjerai Jairoce, Greg Finak, Paige Haas, Carl Muriel, Phu Van, Héctor Sanz, Sheetij Dutta, Benjamin Mordmüller, Selidji T Agnandji, Núria Díez-Padrisa, Nana Aba Williams, John J Aponte, Clarissa Valim, Daniel E Neafsey, Claudia Daubenberger, M Juliana McElrath, Carlota Dobaño, Ken Stuart, Raphael Gottardo, Gemma Moncunill, Jason Carnes, William Chad Young, Lindsay Carpp, Stephen De Rosa, Joseph J Campo, Augusto Nhabomba, Maxmillian Mpina, Chenjerai Jairoce, Greg Finak, Paige Haas, Carl Muriel, Phu Van, Héctor Sanz, Sheetij Dutta, Benjamin Mordmüller, Selidji T Agnandji, Núria Díez-Padrisa, Nana Aba Williams, John J Aponte, Clarissa Valim, Daniel E Neafsey, Claudia Daubenberger, M Juliana McElrath, Carlota Dobaño, Ken Stuart, Raphael Gottardo

Abstract

Background: In a phase 3 trial in African infants and children, the RTS,S/AS01 vaccine (GSK) showed moderate efficacy against clinical malaria. We sought to further understand RTS,S/AS01-induced immune responses associated with vaccine protection.

Methods: Applying the blood transcriptional module (BTM) framework, we characterized the transcriptomic response to RTS,S/AS01 vaccination in antigen-stimulated (and vehicle control) peripheral blood mononuclear cells sampled from a subset of trial participants at baseline and month 3 (1-month post-third dose). Using a matched case-control study design, we evaluated which of these 'RTS,S/AS01 signature BTMs' associated with malaria case status in RTS,S/AS01 vaccinees. Antigen-specific T-cell responses were analyzed by flow cytometry. We also performed a cross-study correlates analysis where we assessed the generalizability of our findings across three controlled human malaria infection studies of healthy, malaria-naive adult RTS,S/AS01 recipients.

Results: RTS,S/AS01 vaccination was associated with downregulation of B-cell and monocyte-related BTMs and upregulation of T-cell-related BTMs, as well as higher month 3 (vs. baseline) circumsporozoite protein-specific CD4+ T-cell responses. There were few RTS,S/AS01-associated BTMs whose month 3 levels correlated with malaria risk. In contrast, baseline levels of BTMs associated with dendritic cells and with monocytes (among others) correlated with malaria risk. The baseline dendritic cell- and monocyte-related BTM correlations with malaria risk appeared to generalize to healthy, malaria-naive adults.

Conclusions: A prevaccination transcriptomic signature associates with malaria in RTS,S/AS01-vaccinated African children, and elements of this signature may be broadly generalizable. The consistent presence of monocyte-related modules suggests that certain monocyte subsets may inhibit protective RTS,S/AS01-induced responses.

Funding: Funding was obtained from the NIH-NIAID (R01AI095789), NIH-NIAID (U19AI128914), PATH Malaria Vaccine Initiative (MVI), and Ministerio de Economía y Competitividad (Instituto de Salud Carlos III, PI11/00423 and PI14/01422). The RNA-seq project has been funded in whole or in part with Federal funds from the National Institute of Allergy and Infectious Diseases, National Institutes of Health, Department of Health and Human Services, under grant number U19AI110818 to the Broad Institute. This study was also supported by the Vaccine Statistical Support (Bill and Melinda Gates Foundation award INV-008576/OPP1154739 to R.G.). C.D. was the recipient of a Ramon y Cajal Contract from the Ministerio de Economía y Competitividad (RYC-2008-02631). G.M. was the recipient of a Sara Borrell-ISCIII fellowship (CD010/00156) and work was performed with the support of Department of Health, Catalan Government grant (SLT006/17/00109). This research is part of the ISGlobal's Program on the Molecular Mechanisms of Malaria which is partially supported by the Fundación Ramón Areces and we acknowledge support from the Spanish Ministry of Science and Innovation through the 'Centro de Excelencia Severo Ochoa 2019-2023' Program (CEX2018-000806-S), and support from the Generalitat de Catalunya through the CERCA Program.

Trial registration: ClinicalTrials.gov NCT00866619.

Keywords: Plasmodium falciparum; gene expression; human; immune cell responses; immunology; infectious disease; inflammation; malaria; microbiology; vaccine.

Conflict of interest statement

GM, JC, WC, LC, SD, AN, MM, CJ, GF, PH, CM, PV, HS, SD, BM, SA, ND, NW, JA, CV, DN, CD, MM, CD, KS, RG No competing interests declared, JC is an employee of Antigen Discovery Inc. The author declare that no other competing interests exist

Figures

Figure 1.. Schematic showing vaccination and sampling…
Figure 1.. Schematic showing vaccination and sampling schedule.
Participants received RTS,S/AS01 (or comparator) at months 0, 1, and 2; peripheral blood mononuclear cells (PBMCs) were collected for fresh stimulations and RNA-sequencing and for cryopreservation at months 0 and 3 (1-month postfinal primary vaccination dose). Stim, stimulation.
Figure 2.. Transcriptional responses and antigen-specific transcriptional…
Figure 2.. Transcriptional responses and antigen-specific transcriptional responses at 1-month postfinal dose associated with RTS,S/AS01 vaccination.
(A) Comparison 1: month 3 (M3) peripheral blood mononuclear cells (PBMC), RTS,S/AS01 vs. comparator; (B) Comparison 2: M3 PBMC vs. month 0 (M0) PBMC, RTS,S/AS01 recipients only. Cell color intensity represents the significance of the difference in the relevant comparison, expressed as signed log10 false discovery rate (FDR); blood transcriptional modules (BTMs) with significantly different expression (FDR ≤0.2) between the two compared groups are outlined in black. |FDR| < 0.2 (*), <0.05 (**), <0.01 (***). Red, higher expression in RTS,S/AS01 recipients vs. comparator recipients at M3 (Comparison 1) or higher expression in RTS,S/AS01 recipients at M3 vs. M0 (Comparison 2); blue, lower expression in RTS,S/AS01 recipients vs. comparator recipients at M3 (Comparison 1) or lower expression in RTS,S/AS01 recipients at M3 vs. M0 (Comparison 2). High-level BTM annotation groups are shown in the left-most color bar. Numbers of participants in each analysis are: (A) Vehicle: 348 (131 comparator, 217 RTS,S/AS01), CSP: 355 (135 comparator, 220 RTS,S/AS01), HBS: 353 (132 comparator, 221 RTS,S/AS01), and AMA1: 351 (132 comparator, 219 RTS,S/AS01). (B) Vehicle: 221, CSP: 224 (221 vehicle, 219 CSP), HBS: 225 (221 vehicle, 211 HBS), AMA1: 223 (221 vehicle, 195 AMA1). Numbers include participants not part of the case–control cohort, and thus exceed the numbers in Table 1. Each ‘vehicle’ column displays the vaccine effect in vehicle; each ‘stimulation’ column displays the vaccine effect for that stimulation compared to vehicle, that is adjusted for vehicle. Detailed equations are given in Methods.
Figure 3.. Associations of month 3 levels…
Figure 3.. Associations of month 3 levels of RTS,S/AS01 signature blood transcriptional modules (BTMs) with malaria case status in RTS,S/AS01 recipients.
Heatmap showing downselected signature BTMs (Comparison 1) with significantly different expression (false discovery rate [FDR] ≤0.2) in month 3 peripheral blood mononuclear cells (PBMC) from RTS,S/AS01 malaria cases vs. nonmalaria controls, in at least one stimulation condition. Cell color intensity represents the significance of the difference in the relevant comparison, expressed as signed log10 FDR; BTMs with significantly different expression in the comparison are outlined in black. |FDR| < 0.2 (*), <0.05 (**), <0.01 (***). Red, higher expression in RTS,S/AS01 cases vs. controls; blue, lower expression in RTS,S/AS01 cases vs. controls. High-level BTM annotation groups are shown in the left-most color bar. Numbers of participants in each analysis are: vehicle: 122, CSP: 123 (122 vehicle, 122 CSP), HBS: 123 (122 vehicle, 115 HBS), AMA1: 123 (122 vehicle, 97 AMA1). The ‘vehicle’ column displays the vaccine effect in vehicle; each ‘stimulation’ column displays the vaccine effect for that stimulation compared to vehicle, that is adjusted for vehicle. Detailed equations are given in Methods.
Figure 3—figure supplement 1.. Associations of month…
Figure 3—figure supplement 1.. Associations of month 3 levels of RTS,S/AS01 signature blood transcriptional modules (BTMs) with malaria case status in comparator recipients.
Heatmap showing which of the 68 downselected RTS,S/AS01 signature BTMs (Comparison 1) showed significantly different expression (false discovery rate [FDR] ≤ 0.2) in month 3 peripheral blood mononuclear cells (PBMC) between comparator cases vs. nonmalaria controls in vehicle-stimulated PBMC from comparator recipients. |FDR| 10 FDR. Red, higher expression in comparator cases vs. controls; blue, lower expression in comparator cases vs. controls. High-level BTM annotation groups are shown in the left-most color bar.
Figure 4.. RTS,S/AS01 vaccination elicits circumsporozoite protein…
Figure 4.. RTS,S/AS01 vaccination elicits circumsporozoite protein (CSP)-specific polyfunctional T-cell responses that do not correlate with clinical malaria risk.
Boxplots show (A) polyfunctionality score and (B) magnitude (% CD4+ T cells expressing IL2 or TNF-α or CD154) of CSP-specific CD4+ T-cell responses in RTS,S/AS01 recipients as assessed by intracellular cytokine staining of peripheral blood mononuclear cells (PBMC) collected at month 0 (M0) or at month 3 (M3). Each dot represents a single participant. Data plotted include all available months 0 and 3 samples, that is paired months 0–3 samples were not required for plotting. (C) Polyfunctionality score and (D) magnitude of CSP-specific CD4+ T-cell responses in RTS,S/AS01 vaccine recipients at month 3, stratified by case–control status. In panels A and B, p values were obtained using a mixed-effects model with participant as a random effect. In panels C and D, p values were obtained using a mixed-effects model with match_id as a random effect. Number of participants in each panel is: (A) 213 (73 M0 and 182 M3), (B) 194 (61 M0, 175 M3), (C) 37 cases and 145 controls, and (D) 36 cases and 139 controls.
Figure 5.. Correlations of month 3 transcriptional…
Figure 5.. Correlations of month 3 transcriptional and adaptive responses in RTS,S/AS01 vaccine recipients.
Heatmap showing correlations between month 3 levels of RTS,S/AS01 signature blood transcriptional modules (BTMs) in vehicle-treated peripheral blood mononuclear cells (PBMC) and month 3 antibody responses. Cell color intensity represents the strength of the correlation; BTM/response pairs with significant correlations (false discovery rate [FDR] ≤0.2) are outlined in black. Cell color represents correlation direction: red, positive correlation; blue, negative correlation. High-level BTM annotation groups are shown in the left-most color bar. Number of participants: 30–42.
Figure 5—figure supplement 1.. Correlations of transcriptional…
Figure 5—figure supplement 1.. Correlations of transcriptional and adaptive responses in comparator vaccine recipients.
Heatmap showing significant correlations between month 3 levels of RTS,S/AS01 signature blood transcriptional modules (BTMs) in vehicle-treated peripheral blood mononuclear cells (PBMC) and month 3 antibody responses in comparator recipients. Cell color intensity represents the strength of the correlation; BTM/response pairs with significant correlations (false discovery rate [FDR] ≤0.2) are outlined in black. Cell color represents correlation direction: red, positive correlation; blue, negative correlation. High-level BTM annotation groups are shown in the left-most color bar. No correlations with month 3 cellular responses were seen.
Figure 6.. Associations of ( A )…
Figure 6.. Associations of (A) month 3 or (B) month 0 levels of downselected blood transcriptional modules (BTMs) with malaria case status RTS,S/AS01 vaccine recipients across studies sharing a common months 0, 1, and 2 RTS,S/AS01 arm.
(A) Heatmap showing the difference in month 3 peripheral blood mononuclear cell (PBMC) BTM expression between RTS,S/AS01 cases vs. controls, in each of three controlled human malaria infection (CHMI) studies, of the seven BTMs whose month 3 levels in vehicle-stimulated PBMC associated with malaria case status in MAL067 (Figure 3). ‘Month 3’ = 21-day postfinal dose in MAL068 and MAL071, and 14-day postfinal dose in WRAIR 1032. BTMs with significantly different expression (false discovery rate [FDR] ≤0.2, with adjustment done across the five BTMs) are outlined in black. |FDR| < 0.2 (*), <0.05 (**), <0.01 (***). (B) Heatmap showing the 45 BTMs whose month 0 levels showed significantly different expression in MAL067 RTS,S/AS01 malaria cases vs. nonmalaria controls. These 45 BTMs were also examined as potential correlates of challenge outcome in each of the 3 CHMI studies. Significantly different expression is defined as FDR ≤0.2, with adjustment across the 45 BTMs. All data shown are from participants who received the same vaccine regimen: a dose of RTS,S/AS01 at months 0, 1, and 2. Cell color intensity represents the significance of the difference in the case vs. control comparison, expressed as signed log10 FDR; BTMs with significantly different expression (FDR ≤0.2) between the two compared groups are outlined in black. |FDR| < 0.2 (*), <0.05 (**), <0.01 (***). Red, higher expression in RTS,S/AS01 cases vs. controls; blue, lower expression in RTS,S/AS01 cases vs. controls. High-level BTM annotation groups are shown in the left-most color bar. Numbers of participants in each analysis are: (A) MAL067, 122; WRAIR 1032, 39; MAL068 RRR, 21; MAL071 RRR, 16. (B) MAL067, 37; WRAIR 1032, 39; MAL068 RRR, 21; MAL071 RRR, 16. Detailed equations are given in Methods.
Figure 6—figure supplement 1.. Association of month…
Figure 6—figure supplement 1.. Association of month 0 level of each gene in the ‘cell cycle and transcription (M4.0)’ module with malaria case status across studies sharing a common months 0, 1, and 2 RTS,S/AS01 arm.
Cell color intensity represents the strength of the difference in the case vs. control comparison, expressed as signed log10 p value; genes with significantly different expression (false discovery rate ≤0.2) between the two compared groups are outlined in black. Red, higher expression in RTS,S/AS01 cases vs. controls; blue, lower expression in RTS,S/AS01 cases vs. controls. The red-outlined portion of the heatmap in the lower right corresponds to an expanded view of the red-outlined portion of the heatmap on the left, for increased legibility of gene names.
Figure 6—figure supplement 2.. Association of month…
Figure 6—figure supplement 2.. Association of month 0 level of each gene in the ‘Resting dendritic cell surface signature (S10)’ module with malaria case status across studies sharing a common months 0, 1, and 2 RTS,S/AS01 arm.
Cell color intensity represents the strength of the difference in the case vs. control comparison, expressed as signed log10 p value; genes with significantly different expression (false discovery rate ≤0.2) between the two compared groups are outlined in black. Red, higher expression in RTS,S/AS01 cases vs. controls; blue, lower expression in RTS,S/AS01 cases vs. controls.
Figure 6—figure supplement 3.. Association of month…
Figure 6—figure supplement 3.. Association of month 0 level of each gene in the ‘DC surface signature (S5)’ module with malaria case status across studies sharing a common months 0, 1, and 2 RTS,S/AS01 arm.
Cell color intensity represents the strength of the difference in the case vs. control comparison, expressed as signed log10 p value; genes with significantly different expression (false discovery rate ≤0.2) between the two compared groups are outlined in black. Red, higher expression in RTS,S/AS01 cases vs. controls; blue, lower expression in RTS,S/AS01 cases vs. controls.
Figure 6—figure supplement 4.. Association of month…
Figure 6—figure supplement 4.. Association of month 0 level of each gene in the ‘Enriched in dendritic cells (M168)’ module with malaria case status across studies sharing a common months 0, 1, and 2 RTS,S/AS01 arm.
Cell color intensity represents the strength of the difference in the case vs. control comparison, expressed as signed log10 p value; genes with significantly different expression (false discovery rate ≤0.2) between the two compared groups are outlined in black. Red, higher expression in RTS,S/AS01 cases vs. controls; blue, lower expression in RTS,S/AS01 cases vs. controls.
Figure 6—figure supplement 5.. Association of month…
Figure 6—figure supplement 5.. Association of month 0 level of each gene in the ‘Myeloid cell enriched receptors and transporters (M4.3)’ module with malaria case status across studies sharing a common months 0, 1, and 2 RTS,S/AS01 arm.
Cell color intensity represents the strength of the difference in the case vs. control comparison, expressed as signed log10 p value; genes with significantly different expression (false discovery rate ≤0.2) between the two compared groups are outlined in black. Red, higher expression in RTS,S/AS01 cases vs. controls; blue, lower expression in RTS,S/AS01 cases vs. controls.
Figure 6—figure supplement 6.. Association of month…
Figure 6—figure supplement 6.. Association of month 0 level of each gene in the ‘Enriched in monocytes (II) (M11.0)’ module with malaria case status across studies sharing a common months 0, 1, and 2 RTS,S/AS01 arm.
Cell color intensity represents the strength of the difference in the case vs. control comparison, expressed as signed log10 p value; genes with significantly different expression (false discovery rate ≤0.2) between the two compared groups are outlined in black. Red, higher expression in RTS,S/AS01 cases vs. controls; blue, lower expression in RTS,S/AS01 cases vs. controls.
Figure 6—figure supplement 7.. Association of month…
Figure 6—figure supplement 7.. Association of month 0 level of each gene in the ‘Enriched in monocytes (I) (M4.15)’ module with malaria case status across studies sharing a common months 0, 1, and 2 RTS,S/AS01 arm.
Cell color intensity represents the strength of the difference in the case vs. control comparison, expressed as signed log10 p value; genes with significantly different expression (false discovery rate ≤0.2) between the two compared groups are outlined in black. Red, higher expression in RTS,S/AS01 cases vs. controls; blue, lower expression in RTS,S/AS01 cases vs. controls.
Figure 6—figure supplement 8.. Association of month…
Figure 6—figure supplement 8.. Association of month 0 level of each gene in the ‘Monocyte surface signature (S4)’ module with malaria case status across studies sharing a common months 0, 1, and 2 RTS,S/AS01 arm.
Cell color intensity represents the strength of the difference in the case vs. control comparison, expressed as signed log10 p value; genes with significantly different expression (false discovery rate ≤0.2) between the two compared groups are outlined in black. Red, higher expression in RTS,S/AS01 cases vs. controls; blue, lower expression in RTS,S/AS01 cases vs. controls.
Figure 6—figure supplement 9.. Correlations of month…
Figure 6—figure supplement 9.. Correlations of month 0 blood transcriptional module (BTM) expression in vehicle-treated peripheral blood mononuclear cells (PBMC) with month 3 T-cell responses in RTS,S/AS01 vaccine recipients.
Heatmap showing significant correlations between month 0 levels of RTS,S/AS01 signature BTMs and month 3 cellular responses (no significant correlations were seen with any month 3 antibody responses). Cell color intensity represents the strength of the correlation; BTM/response pairs with significant correlations (false discovery rate [FDR] ≤0.2) are outlined in black. Cell color represents correlation direction: red, positive correlation; blue, negative correlation. High-level BTM annotation groups are shown in the left-most color bar. No significant correlations were seen with month 3 antibody responses. PFS, polyfunctionality score.
Figure 6—figure supplement 10.. No significant differences…
Figure 6—figure supplement 10.. No significant differences in monocyte frequencies in cases vs. controls in RTS,S/AS01 vaccinees at either month 0 or 3.
(A) Monocyte frequency, (B) inflammatory monocyte frequency, and (C) inflammatory monocyte/lymphocyte ratio, stratified by case/control status, in RTS,S/AS01 recipients. Flow cytometry data were analyzed using the gating strategy defined in Moncunill et al., 2017a. ‘Inflammatory monocytes’ refers to ‘HLA-DR+ CD14+ CD16++’ cells. The p values at the bottom of each panel are from testing for a significant difference in controls vs. cases within each panel, and were modeled using a mixed-effects model (using lmer) with match ID as a random effect.

References

    1. Adey A, Morrison HG, Xun X, Kitzman JO, Turner EH, Stackhouse B, MacKenzie AP, Caruccio NC, Zhang X, Shendure J. Rapid, low-input, low-bias construction of shotgun fragment libraries by high-density in vitro transposition. Genome Biology. 2010;11:R119. doi: 10.1186/gb-2010-11-12-r119.
    1. Agnandji ST, Lell B, Soulanoudjingar SS, Fernandes JF, Abossolo BP, Conzelmann C, Methogo B, Doucka Y, Flamen A, Mordmüller B, Issifou S, Kremsner PG, Sacarlal J, Aide P, Lanaspa M, Aponte JJ, Nhamuave A, Quelhas D, Bassat Q, Mandjate S, Macete E, Alonso P, Abdulla S, Salim N, Juma O, Shomari M, Shubis K, Machera F, Hamad AS, Minja R, Mtoro A, Sykes A, Ahmed S, Urassa AM, Ali AM, Mwangoka G, Tanner M, Tinto H, D’Alessandro U, Sorgho H, Valea I, Tahita MC, Kaboré W, Ouédraogo S, Sandrine Y, Guiguemdé RT, Ouédraogo JB, Hamel MJ, Kariuki S, Odero C, Oneko M, Otieno K, Awino N, Omoto J, Williamson J, Muturi-Kioi V, Laserson KF, Slutsker L, Otieno W, Otieno L, Nekoye O, Gondi S, Otieno A, Ogutu B, Wasuna R, Owira V, Jones D, Onyango AA, Njuguna P, Chilengi R, Akoo P, Kerubo C, Gitaka J, Maingi C, Lang T, Olotu A, Tsofa B, Bejon P, Peshu N, Marsh K, Owusu-Agyei S, Asante KP, Osei-Kwakye K, Boahen O, Ayamba S, Kayan K, Owusu-Ofori R, Dosoo D, Asante I, Adjei G, Adjei G, Chandramohan D, Greenwood B, Lusingu J, Gesase S, Malabeja A, Abdul O, Kilavo H, Mahende C, Liheluka E, Lemnge M, Theander T, Drakeley C, Ansong D, Agbenyega T, Adjei S, Boateng HO, Rettig T, Bawa J, Sylverken J, Sambian D, Agyekum A, Owusu L, Martinson F, Hoffman I, Mvalo T, Kamthunzi P, Nkomo R, Msika A, Jumbe A, Chome N, Nyakuipa D, Chintedza J, Ballou WR, Bruls M, Cohen J, Guerra Y, Jongert E, Lapierre D, Leach A, Lievens M, Ofori-Anyinam O, Vekemans J, Carter T, Leboulleux D, Loucq C, Radford A, Savarese B, Schellenberg D, Sillman M, Vansadia P. First results of phase 3 trial of RTS,S/AS01 malaria vaccine in African children. The New England Journal of Medicine. 2011;365:1863–1875. doi: 10.1056/NEJMoa1102287.
    1. Andersen-Nissen E, Fiore-Gartland A, Ballweber Fleming L, Carpp LN, Naidoo AF, Harper MS, Voillet V, Grunenberg N, Laher F, Innes C, Bekker L-G, Kublin JG, Huang Y, Ferrari G, Tomaras GD, Gray G, Gilbert PB, McElrath MJ. Innate immune signatures to a partially-efficacious HIV vaccine predict correlates of HIV-1 infection risk. PLOS Pathogens. 2021;17:e1009363. doi: 10.1371/journal.ppat.1009363.
    1. Antwi-Baffour S, Kyeremeh R, Buabeng D, Adjei JK, Aryeh C, Kpentey G, Seidu MA. Correlation of malaria parasitaemia with peripheral blood monocyte to lymphocyte ratio as indicator of susceptibility to severe malaria in Ghanaian children. Malaria Journal. 2018;17:419. doi: 10.1186/s12936-018-2569-x.
    1. Baldridge JR, McGowan P, Evans JT, Cluff C, Mossman S, Johnson D, Persing D. Taking a Toll on human disease: Toll-like receptor 4 agonists as vaccine adjuvants and monotherapeutic agents. Expert Opinion on Biological Therapy. 2004;4:1129–1138. doi: 10.1517/14712598.4.7.1129.
    1. Beeson JG, Kurtovic L, Dobaño C, Opi DH, Chan JA, Feng G, Good MF, Reiling L, Boyle MJ. Challenges and strategies for developing efficacious and long-lasting malaria vaccines. Science Translational Medicine. 2019;11:474. doi: 10.1126/scitranslmed.aau1458.
    1. Benjamini Y, Hochberg Y. Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. Journal of the Royal Statistical Society. 1995;57:289–300. doi: 10.1111/j.2517-6161.1995.tb02031.x.
    1. Bueno LL, Fujiwara RT, Soares IS, Braga EM. Direct effect of Plasmodium vivax recombinant vaccine candidates AMA-1 and MSP-119 on the innate immune response. Vaccine. 2008;26:1204–1213. doi: 10.1016/j.vaccine.2007.12.031.
    1. Chaudhury S, Ockenhouse CF, Regules JA, Dutta S, Wallqvist A, Jongert E, Waters NC, Lemiale F, Bergmann-Leitner E. The biological function of antibodies induced by the RTS,S/AS01 malaria vaccine candidate is determined by their fine specificity. Malaria Journal. 2016;15:301. doi: 10.1186/s12936-016-1348-9.
    1. Dennison SM, Reichartz M, Abraha M, Spreng RL, Wille-Reece U, Dutta S, Jongert E, Alam SM, Tomaras GD. Magnitude, Specificity, and Avidity of Sporozoite-Specific Antibodies Associate With Protection Status and Distinguish Among RTS,S/AS01 Dose Regimens. Open Forum Infectious Diseases. 2021;8:ofaa644. doi: 10.1093/ofid/ofaa644.
    1. Didierlaurent AM, Laupèze B, Di Pasquale A, Hergli N, Collignon C, Garçon N. Adjuvant system AS01: helping to overcome the challenges of modern vaccines. Expert Review of Vaccines. 2017;16:55–63. doi: 10.1080/14760584.2016.1213632.
    1. Dobaño C, Sanz H, Sorgho H, Dosoo D, Mpina M, Ubillos I, Aguilar R, Ford T, Díez-Padrisa N, Williams NA, Ayestaran A, Traore O, Nhabomba AJ, Jairoce C, Waitumbi J, Agnandji ST, Kariuki S, Abdulla S, Aponte JJ, Mordmüller B, Asante KP, Owusu-Agyei S, Tinto H, Campo JJ, Moncunill G, Gyan B, Valim C, Daubenberger C. Concentration and avidity of antibodies to different circumsporozoite epitopes correlate with RTS,S/AS01E malaria vaccine efficacy. Nature Communications. 2019a;10:2174. doi: 10.1038/s41467-019-10195-z.
    1. Dobaño C, Ubillos I, Jairoce C, Gyan B, Vidal M, Jiménez A, Santano R, Dosoo D, Nhabomba AJ, Ayestaran A, Aguilar R, Williams NA, Díez-Padrisa N, Lanar D, Chauhan V, Chitnis C, Dutta S, Gaur D, Angov E, Asante KP, Owusu-Agyei S, Valim C, Gamain B, Coppel RL, Cavanagh D, Beeson JG, Campo JJ, Moncunill G. RTS,S/AS01E immunization increases antibody responses to vaccine-unrelated Plasmodium falciparum antigens associated with protection against clinical malaria in African children: a case-control study. BMC Medicine. 2019b;17:157. doi: 10.1186/s12916-019-1378-6.
    1. Du Y, Thompson EG, Muller J, Valvo J, Braun J, Shankar S, van den Berg RA, Jongert E, Dover D, Sadoff J, Hendriks J, Gardner MJ, Ballou WR, Regules JA, van der Most R, Aderem A, Ockenhouse CF, Hill AV, Wille-Reece U, Zak DE. The Ratiometric Transcript Signature MX2/GPR183 Is Consistently Associated With RTS,S-Mediated Protection Against Controlled Human Malaria Infection. Frontiers in Immunology. 2020;11:669. doi: 10.3389/fimmu.2020.00669.
    1. Eisele TP. Mass drug administration can be a valuable addition to the malaria elimination toolbox. Malaria Journal. 2019;18:281. doi: 10.1186/s12936-019-2906-8.
    1. Fletcher HA, Snowden MA, Landry B, Rida W, Satti I, Harris SA, Matsumiya M, Tanner R, O’Shea MK, Dheenadhayalan V, Bogardus L, Stockdale L, Marsay L, Chomka A, Harrington-Kandt R, Manjaly-Thomas Z-R, Naranbhai V, Stylianou E, Darboe F, Penn-Nicholson A, Nemes E, Hatherill M, Hussey G, Mahomed H, Tameris M, McClain JB, Evans TG, Hanekom WA, Scriba TJ, McShane H. T-cell activation is an immune correlate of risk in BCG vaccinated infants. Nature Communications. 2016;7:11290. doi: 10.1038/ncomms11290.
    1. Fourati S, Cristescu R, Loboda A, Talla A, Filali A, Railkar R, Schaeffer AK, Favre D, Gagnon D, Peretz Y, Wang I-M, Beals CR, Casimiro DR, Carayannopoulos LN, Sékaly R-P. Pre-vaccination inflammation and B-cell signalling predict age-related hyporesponse to hepatitis B vaccination. Nature Communications. 2016;7:10369. doi: 10.1038/ncomms10369.
    1. Gordon DM, McGovern TW, Krzych U, Cohen JC, Schneider I, LaChance R, Heppner DG, Yuan G, Hollingdale M, Slaoui M. Safety, immunogenicity, and efficacy of a recombinantly produced Plasmodium falciparum circumsporozoite protein-hepatitis B surface antigen subunit vaccine. The Journal of Infectious Diseases. 1995;171:1576–1585. doi: 10.1093/infdis/171.6.1576.
    1. Hawkes N. European Medicines Agency approves first malaria vaccine. BMJ. 2015;351:h4067. doi: 10.1136/bmj.h4067.
    1. Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, Alam SM, Evans DT, Montefiori DC, Karnasuta C, Sutthent R, Liao H-X, DeVico AL, Lewis GK, Williams C, Pinter A, Fong Y, Janes H, DeCamp A, Huang Y, Rao M, Billings E, Karasavvas N, Robb ML, Ngauy V, de Souza MS, Paris R, Ferrari G, Bailer RT, Soderberg KA, Andrews C, Berman PW, Frahm N, De Rosa SC, Alpert MD, Yates NL, Shen X, Koup RA, Pitisuttithum P, Kaewkungwal J, Nitayaphan S, Rerks-Ngarm S, Michael NL, Kim JH. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. The New England Journal of Medicine. 2012;366:1275–1286. doi: 10.1056/NEJMoa1113425.
    1. Healer J, Cowman AF, Kaslow DC, Birkett AJ. Vaccines to Accelerate Malaria Elimination and Eventual Eradication. Cold Spring Harbor Perspectives in Medicine. 2017;7:a025627. doi: 10.1101/cshperspect.a025627.
    1. Hill DL, Carr EJ, Rutishauser T, Moncunill G, Campo JJ, Innocentin S, Mpina M, Nhabomba A, Tumbo A, Jairoce C, Moll HA, van Zelm MC, Dobaño C, Daubenberger C, Linterman MA. Immune system development varies according to age, location, and anemia in African children. Science Translational Medicine. 2020;12:529. doi: 10.1126/scitranslmed.aaw9522.
    1. HIPC-CHI Signatures Project Team. HIPC-I Consortium Multicohort analysis reveals baseline transcriptional predictors of influenza vaccination responses. Science Immunology. 2017;2:eaal4656. doi: 10.1126/sciimmunol.aal4656.
    1. Hoffman SL, Vekemans J, Richie TL, Duffy PE. The march toward malaria vaccines. Vaccine. 2015;33 Suppl 4:D13–D23. doi: 10.1016/j.vaccine.2015.07.091.
    1. Hornung V, Rothenfusser S, Britsch S, Krug A, Jahrsdörfer B, Giese T, Endres S, Hartmann G. Quantitative expression of toll-like receptor 1-10 mRNA in cellular subsets of human peripheral blood mononuclear cells and sensitivity to CpG oligodeoxynucleotides. Journal of Immunology. 2002;168:4531–4537. doi: 10.4049/jimmunol.168.9.4531.
    1. Kazmin D, Nakaya HI, Lee EK, Johnson MJ, van der Most R, van den Berg RA, Ballou WR, Jongert E, Wille-Reece U, Ockenhouse C, Aderem A, Zak DE, Sadoff J, Hendriks J, Wrammert J, Ahmed R, Pulendran B. Systems analysis of protective immune responses to RTS,S malaria vaccination in humans. PNAS. 2017;114:2425–2430. doi: 10.1073/pnas.1621489114.
    1. Kester KE, Cummings JF, Ofori-Anyinam O, Ockenhouse CF, Krzych U, Moris P, Schwenk R, Nielsen RA, Debebe Z, Pinelis E, Juompan L, Williams J, Dowler M, Stewart VA, Wirtz RA, Dubois M-C, Lievens M, Cohen J, Ballou WR, Heppner DG, Jr, RTS,S Vaccine Evaluation Group Randomized, double-blind, phase 2a trial of falciparum malaria vaccines RTS,S/AS01B and RTS,S/AS02A in malaria-naive adults: safety, efficacy, and immunologic associates of protection. The Journal of Infectious Diseases. 2009;200:337–346. doi: 10.1086/600120.
    1. Kotliarov Y, Sparks R, Martins AJ, Mulè MP, Lu Y, Goswami M, Kardava L, Banchereau R, Pascual V, Biancotto A, Chen J, Schwartzberg PL, Bansal N, Liu CC, Cheung F, Moir S, Tsang JS. Broad immune activation underlies shared set point signatures for vaccine responsiveness in healthy individuals and disease activity in patients with lupus. Nature Medicine. 2020;26:618–629. doi: 10.1038/s41591-020-0769-8.
    1. Kzhyshkowska J, Gratchev A, Goerdt S. Stabilin-1, a homeostatic scavenger receptor with multiple functions. Journal of Cellular and Molecular Medicine. 2006;10:635–649. doi: 10.1111/j.1582-4934.2006.tb00425.x.
    1. Law CW, Chen Y, Shi W, Smyth GK. voom: Precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biology. 2014;15:R29. doi: 10.1186/gb-2014-15-2-r29.
    1. Li S, Rouphael N, Duraisingham S, Romero-Steiner S, Presnell S, Davis C, Schmidt DS, Johnson SE, Milton A, Rajam G, Kasturi S, Carlone GM, Quinn C, Chaussabel D, Palucka AK, Mulligan MJ, Ahmed R, Stephens DS, Nakaya HI, Pulendran B. Molecular signatures of antibody responses derived from a systems biology study of five human vaccines. Nature Immunology. 2014;15:195–204. doi: 10.1038/ni.2789.
    1. Lin L, Finak G, Ushey K, Seshadri C, Hawn TR, Frahm N, Scriba TJ, Mahomed H, Hanekom W, Bart P-A, Pantaleo G, Tomaras GD, Rerks-Ngarm S, Kaewkungwal J, Nitayaphan S, Pitisuttithum P, Michael NL, Kim JH, Robb ML, O’Connell RJ, Karasavvas N, Gilbert P, C De Rosa S, McElrath MJ, Gottardo R. COMPASS identifies T-cell subsets correlated with clinical outcomes. Nature Biotechnology. 2015;33:610–616. doi: 10.1038/nbt.3187.
    1. Liu C, Martins AJ, Lau WW, Rachmaninoff N, Chen J, Imberti L, Mostaghimi D, Fink DL, Burbelo PD, Dobbs K, Delmonte OM, Bansal N, Failla L, Sottini A, Quiros-Roldan E, Han KL, Sellers BA, Cheung F, Sparks R, Chun T-W, Moir S, Lionakis MS, NIAID COVID Consortium. COVID Clinicians. Rossi C, Su HC, Kuhns DB, Cohen JI, Notarangelo LD, Tsang JS. Time-resolved systems immunology reveals a late juncture linked to fatal COVID-19. Cell. 2021;184:1836–1857. doi: 10.1016/j.cell.2021.02.018.
    1. Lu P, Guerin DJ, Lin S, Chaudhury S, Ackerman ME, Bolton DL, Wallqvist A. Immunoprofiling Correlates of Protection Against SHIV Infection in Adjuvanted HIV-1 Pox-Protein Vaccinated Rhesus Macaques. Frontiers in Immunology. 2021;12:625030. doi: 10.3389/fimmu.2021.625030.
    1. Mitchell LA, Henderson AJ, Dow SW. Suppression of vaccine immunity by inflammatory monocytes. Journal of Immunology. 2012;189:5612–5621. doi: 10.4049/jimmunol.1202151.
    1. Moncunill G, Han H, Dobaño C, McElrath MJ, De Rosa SC. OMIP-024: pan-leukocyte immunophenotypic characterization of PBMC subsets in human samples. Cytometry. Part A. 2014;85:995–998. doi: 10.1002/cyto.a.22580.
    1. Moncunill G, Dobaño C, McElrath MJ, De Rosa SC. OMIP-025: evaluation of human T- and NK-cell responses including memory and follicular helper phenotype by intracellular cytokine staining. Cytometry. Part A. 2015;87:289–292. doi: 10.1002/cyto.a.22590.
    1. Moncunill G, De Rosa SC, Ayestaran A, Nhabomba AJ, Mpina M, Cohen KW, Jairoce C, Rutishauser T, Campo JJ, Harezlak J, Sanz H, Díez-Padrisa N, Williams NA, Morris D, Aponte JJ, Valim C, Daubenberger C, Dobaño C, McElrath MJ. RTS,S/AS01E Malaria Vaccine Induces Memory and Polyfunctional T Cell Responses in a Pediatric African Phase III Trial. Frontiers in Immunology. 2017a;8:1008. doi: 10.3389/fimmu.2017.01008.
    1. Moncunill G, Mpina M, Nhabomba AJ, Aguilar R, Ayestaran A, Sanz H, Campo JJ, Jairoce C, Barrios D, Dong Y, Díez-Padrisa N, Fernandes JF, Abdulla S, Sacarlal J, Williams NA, Harezlak J, Mordmüller B, Agnandji ST, Aponte JJ, Daubenberger C, Valim C, Dobaño C. Distinct Helper T Cell Type 1 and 2 Responses Associated With Malaria Protection and Risk in RTS,S/AS01E Vaccinees. Clinical Infectious Diseases. 2017b;65:746–755. doi: 10.1093/cid/cix429.
    1. Moncunill G, Scholzen A, Mpina M, Nhabomba A, Hounkpatin AB, Osaba L, Valls R, Campo JJ, Sanz H, Jairoce C, Williams NA, Pasini EM, Arteta D, Maynou J, Palacios L, Duran-Frigola M, Aponte JJ, Kocken CHM, Agnandji ST, Mas JM, Mordmüller B, Daubenberger C, Sauerwein R, Dobaño C. Antigen-stimulated PBMC transcriptional protective signatures for malaria immunization. Science Translational Medicine. 2020;12:543. doi: 10.1126/scitranslmed.aay8924.
    1. Moris P, Jongert E, van der Most RG. Characterization of T-cell immune responses in clinical trials of the candidate RTS,S malaria vaccine. Human Vaccines & Immunotherapeutics. 2018;14:17–27. doi: 10.1080/21645515.2017.1381809.
    1. Norris BA, Uebelhoer LS, Nakaya HI, Price AA, Grakoui A, Pulendran B. Chronic but not acute virus infection induces sustained expansion of myeloid suppressor cell numbers that inhibit viral-specific T cell immunity. Immunity. 2013;38:309–321. doi: 10.1016/j.immuni.2012.10.022.
    1. Ockenhouse CF, Regules J, Tosh D, Cowden J, Kathcart A, Cummings J, Paolino K, Moon J, Komisar J, Kamau E, Oliver T, Chhoeu A, Murphy J, Lyke K, Laurens M, Birkett A, Lee C, Weltzin R, Wille-Reece U, Sedegah M, Hendriks J, Versteege I, Pau MG, Sadoff J, Vanloubbeeck Y, Lievens M, Heerwegh D, Moris P, Guerra Mendoza Y, Jongert E, Cohen J, Voss G, Ballou WR, Vekemans J. Ad35.CS.01-RTS,S/AS01 Heterologous Prime Boost Vaccine Efficacy against Sporozoite Challenge in Healthy Malaria-Naïve Adults. PLOS ONE. 2015;10:e0131571. doi: 10.1371/journal.pone.0131571.
    1. Palani S, Elima K, Ekholm E, Jalkanen S, Salmi M. Monocyte Stabilin-1 Suppresses the Activation of Th1 Lymphocytes. Journal of Immunology. 2016;196:115–123. doi: 10.4049/jimmunol.1500257.
    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. doi: 10.7554/eLife.51889.
    1. Pichichero ME. Challenges in vaccination of neonates, infants and young children. Vaccine. 2014;32:3886–3894. doi: 10.1016/j.vaccine.2014.05.008.
    1. Regules JA, Cicatelli SB, Bennett JW, Paolino KM, Twomey PS, Moon JE, Kathcart AK, Hauns KD, Komisar JL, Qabar AN, Davidson SA, Dutta S, Griffith ME, Magee CD, Wojnarski M, Livezey JR, Kress AT, Waterman PE, Jongert E, Wille-Reece U, Volkmuth W, Emerling D, Robinson WH, Lievens M, Morelle D, Lee CK, Yassin-Rajkumar B, Weltzin R, Cohen J, Paris RM, Waters NC, Birkett AJ, Kaslow DC, Ballou WR, Ockenhouse CF, Vekemans J. Fractional Third and Fourth Dose of RTS,S/AS01 Malaria Candidate Vaccine: A Phase 2a Controlled Human Malaria Parasite Infection and Immunogenicity Study. The Journal of Infectious Diseases. 2016;214:762–771. doi: 10.1093/infdis/jiw237.
    1. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Research. 2015;43:e47. doi: 10.1093/nar/gkv007.
    1. Robinson MD, Oshlack A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biology. 2010;11:R25. doi: 10.1186/gb-2010-11-3-r25.
    1. RTS,S Clinical Trials Partnership A phase 3 trial of RTS, S/AS01 malaria vaccine in African infants. New England Journal of Medicine. 2012;367:2284–2295. doi: 10.1056/NEJMoa1208394.
    1. 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 Medicine. 2014;11:e1001685. doi: 10.1371/journal.pmed.1001685.
    1. RTS,S Clinical Trials Partnership 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. doi: 10.1016/S0140-6736(15)60721-8.
    1. Sammicheli S, Kuka M, Di Lucia P, de Oya NJ, De Giovanni M, Fioravanti J, Cristofani C, Maganuco CG, Fallet B, Ganzer L, Sironi L, Mainetti M, Ostuni R, Larimore K, Greenberg PD, de la Torre JC, Guidotti LG, Iannacone M. Inflammatory monocytes hinder antiviral B cell responses. Science Immunology. 2016;1:eaah6789. doi: 10.1126/sciimmunol.aah6789.
    1. Simon AK, Hollander GA, McMichael A. Evolution of the immune system in humans from infancy to old age. Proceedings. Biological Sciences. 2015;282:20143085. doi: 10.1098/rspb.2014.3085.
    1. Smyth GK. Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Statistical Applications in Genetics and Molecular Biology. 2004;3:e1027. doi: 10.2202/1544-6115.1027.
    1. Soumillon M, Cacchiarelli D, Semrau S, Oudenaarden A, Mikkelsen TS. Characterization of Directed Differentiation by High-Throughput Single-Cell RNA-Seq. bioRxiv. 2014 doi: 10.1101/003236.
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, Mesirov JP. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. PNAS. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102.
    1. Suscovich TJ, Fallon JK, Das J, Demas AR, Crain J, Linde CH, Michell A, Natarajan H, Arevalo C, Broge T, Linnekin T, Kulkarni V, Lu R, Slein MD, Luedemann C, Marquette M, March S, Weiner J, Gregory S, Coccia M, Flores-Garcia Y, Zavala F, Ackerman ME, Bergmann-Leitner E, Hendriks J, Sadoff J, Dutta S, Bhatia SN, Lauffenburger DA, Jongert E, Wille-Reece U, Alter G. Mapping functional humoral correlates of protection against malaria challenge following RTS,S/AS01 vaccination. Science Translational Medicine. 2020;12:553. doi: 10.1126/scitranslmed.abb4757.
    1. Tsang JS, Schwartzberg PL, Kotliarov Y, Biancotto A, Xie Z, Germain RN, Wang E, Olnes MJ, Narayanan M, Golding H, Moir S, Dickler HB, Perl S, Cheung F, Baylor HIPC Center. CHI Consortium Global analyses of human immune variation reveal baseline predictors of postvaccination responses. Cell. 2014;157:499–513. doi: 10.1016/j.cell.2014.03.031.
    1. Tsang JS, Dobaño C, VanDamme P, Moncunill G, Marchant A, Othman RB, Sadarangani M, Koff WC, Kollmann TR. Improving Vaccine-Induced Immunity: Can Baseline Predict Outcome? Trends in Immunology. 2020;41:457–465. doi: 10.1016/j.it.2020.04.001.
    1. Ubillos I, Ayestaran A, Nhabomba AJ, Dosoo D, Vidal M, Jiménez A, Jairoce C, Sanz H, Aguilar R, Williams NA, Díez-Padrisa N, Mpina M, Sorgho H, Agnandji ST, Kariuki S, Mordmüller B, Daubenberger C, Asante KP, Owusu-Agyei S, Sacarlal J, Aide P, Aponte JJ, Dutta S, Gyan B, Campo JJ, Valim C, Moncunill G, Dobaño C. Baseline exposure, antibody subclass, and hepatitis B response differentially affect malaria protective immunity following RTS,S/AS01E vaccination in African children. BMC Medicine. 2018;16:197. doi: 10.1186/s12916-018-1186-4.
    1. Vahey MT, Wang Z, Kester KE, Cummings J, Heppner DG, Jr, Nau ME, Ofori-Anyinam O, Cohen J, Coche T, Ballou WR, Ockenhouse CF. Expression of genes associated with immunoproteasome processing of major histocompatibility complex peptides is indicative of protection with adjuvanted RTS,S malaria vaccine. The Journal of Infectious Diseases. 2010;201:580–589. doi: 10.1086/650310.
    1. van den Berg RA, Coccia M, Ballou WR, Kester KE, Ockenhouse CF, Vekemans J, Jongert E, Didierlaurent AM, van der Most RG. Predicting RTS,S Vaccine-Mediated Protection from Transcriptomes in a Malaria-Challenge Clinical Trial. Frontiers in Immunology. 2017;8:557. doi: 10.3389/fimmu.2017.00557.
    1. Vaure C, Liu YQ. A comparative review of toll-like receptor 4 expression and functionality in different animal species. Frontiers in Immunology. 2014;5:316. doi: 10.3389/fimmu.2014.00316.
    1. Warimwe GM, Fletcher HA, Olotu A, Agnandji ST, Hill AVS, Marsh K, Bejon P. Peripheral blood monocyte-to-lymphocyte ratio at study enrollment predicts efficacy of the RTS,S malaria vaccine: analysis of pooled phase II clinical trial data. BMC Medicine. 2013a;11:184. doi: 10.1186/1741-7015-11-184.
    1. Warimwe GM, Murungi LM, Kamuyu G, Nyangweso GM, Wambua J, Naranbhai V, Fletcher HA, Hill AVS, Bejon P, Osier FHA, Marsh K. The ratio of monocytes to lymphocytes in peripheral blood correlates with increased susceptibility to clinical malaria in Kenyan children. PLOS ONE. 2013b;8:e57320. doi: 10.1371/journal.pone.0057320.
    1. World Health Organization Q&A on the malaria vaccine implementation programme (MVIP. 2019. [November 29, 2019].
    1. World Health Organization World Malaria Report. 2021a. [January 9, 2021].
    1. World Health Organization WHO recommends groundbreaking malaria vaccine for children at risk. News Release. 2021b. [October 6, 2021].
    1. Wu D, Smyth GK. Camera: a competitive gene set test accounting for inter-gene correlation. Nucleic Acids Research. 2012;40:17. doi: 10.1093/nar/gks461.
    1. Yang GG, Kim D, Pham A, Paul CJ. A Meta-Regression Analysis of the Effectiveness of Mosquito Nets for Malaria Control: The Value of Long-Lasting Insecticide Nets. International Journal of Environmental Research and Public Health. 2018;15:E546. doi: 10.3390/ijerph15030546.
    1. Young WC, Carpp LN, Chaudhury S, Regules JA, Bergmann-Leitner ES, Ockenhouse C, Wille-Reece U, deCamp AC, Hughes E, Mahoney C, Pallikkuth S, Pahwa S, Dennison SM, Mudrak SV, Alam SM, Seaton KE, Spreng RL, Fallon J, Michell A, Ulloa-Montoya F, Coccia M, Jongert E, Alter G, Tomaras GD, Gottardo R. Comprehensive Data Integration Approach to Assess Immune Responses and Correlates of RTS,S/AS01-Mediated Protection From Malaria Infection in Controlled Human Malaria Infection Trials. Frontiers in Big Data. 2021;4:672460. doi: 10.3389/fdata.2021.672460.

Source: PubMed

3
Předplatit