Controlled Human Malaria Infection (CHMI) differentially affects cell-mediated and antibody responses to CSP and AMA1 induced by adenovirus vaccines with and without DNA-priming

Martha Sedegah, Michael R Hollingdale, Fouzia Farooq, Harini Ganeshan, Maria Belmonte, Jun Huang, Esteban Abot, Keith Limbach, Ilin Chuang, Cindy Tamminga, Judith E Epstein, Eileen Villasante, Martha Sedegah, Michael R Hollingdale, Fouzia Farooq, Harini Ganeshan, Maria Belmonte, Jun Huang, Esteban Abot, Keith Limbach, Ilin Chuang, Cindy Tamminga, Judith E Epstein, Eileen Villasante

Abstract

We have previously shown that a DNA-prime followed by an adenovirus-5 boost vaccine containing CSP and AMA1 (DNA/Ad) successfully protected 4 of 15 subjects to controlled human malaria infection (CHMI). However, the adenovirus-5 vaccine alone (AdCA) failed to induce protection despite eliciting cellular responses that were often higher than those induced by DNA/Ad. Here we determined the effect of CHMI on pre-CHMI cellular and antibody responses against CSP and AMA1 expressed as fold-changes in activities. Generally, in the DNA/Ad trial, CHMI caused pre-CHMI ELISpot IFN-γ and CD8+ T cell IFN-γ responses of the protected subjects to fall but among non-protected subjects, CHMI caused rises of pre-CHMI ELISpot IFN-γ but falls of CD8+ T cell IFN-γ responses. In contrast in the AdCA trial, CHMI caused both pre-CHMI ELISpot IFN-γ and CD8+ T cell IFN-γ responses of the AdCA subjects to fall. We suggest that the falls in activities are due to migration of peripheral CD8+ T cells to the liver in response to developing liver stage parasites, and this fall, in the DNA/Ad trial, is masked in ELISpot responses of the non-protected subjects by rises in other immune cell types. In addition, CHMI caused falls in antibody activities of protected subjects, but rises in non-protected subjects in both trials to CSP, and dramatically in the AdCA trial to AMA1, reaching 380 μg/ml that is probably due to boosting by transient blood stage infection before chloroquine treatment. Taken together, these results further define differences in cellular responses between DNA/Ad and AdCA trials, and suggest that natural transmission may boost responses induced by these malaria vaccines especially when protection is not achieved.

Trial registration: ClinicalTrials.gov NCT00392015 NCT00870987.

Keywords: AMA1; CHMI; CSP; DNA -prime; T cells; adenovirus-boost; antibody; efficacy; malaria; vaccine.

Figures

Figure 1.
Figure 1.
Ex vivo ELISpot IFN-γ activities of infectivity controls. Five infectivity controls from the DNA/Ad trial and 4 infectivity controls from the AdCA trial were used. The associations of fold-changes of pre-CHMI and post-CHMI activities with pre-CHMI activities are shown as log-transformed values, and the dotted line represents no-change. The shaded box shows ±1.5 range (log ±0.18)-. (A) CSP: the fold change was ≤1.5, except one outlier, and all 9 subjects remained negative after CHMI. (B) AMA1: the fold change of all subjects was <1.5, and 6/9 subjects remained negative after CHMI, but 3/9 subjects developed positive activities after CHMI each to a single AMA1 peptide pool (not shown).
Figure 2.
Figure 2.
DNA/Ad trial: ex vivo ELISpot IFN-γ activities to CSP and AMA1. The associations of fold-changes of pre-CHMI and post-CHMI activities with pre-CHMI activities are shown as log-transformed values, and the dotted line represents no-change. The shaded box shows ±1.5 range. (A) CSP: there was a significant relationship between fold-change and pre-CHMI activities; the fold changes of 2 protected (v11, v18) and 3 non-protected subjects were greater than ±1.5 (shaded box). (B) AMA1: there was a significant relationship between fold-change and pre-CHMI activities; the fold changes of theAMA1 immunodominant pools of v10 (Ap8), v11 (Ap10) and v18 (Ap8) were used as they represented most of the total summed activities. The fold changes of 3 protected subjects (v10, v11, v18) and 9 non-protected subjects were greater than ±1.5 (shaded box).
Figure 3.
Figure 3.
DNA/Ad trial: CD8+ T cell IFN-γ activities to CSP and AMA1. The associations of fold-changes of pre-CHMI and post-CHMI activities with pre-CHMI activities are shown as log-transformed values, and the dotted line represents no-change. The shaded box shows ±1.5 range. (A) CSP: there was a significant relationship between fold-change and pre-CHMI activities; the fold changes of 2 protected (v11, v18) and 3 non-protected subjects were greater than ±1.5 (shaded box). (B) AMA1: there was no significant relationship between fold-change and pre-CHMI activities; the fold changes of 3 protected (v10, v11, v18) and 4 non-protected subjects were greater than −1.5 (shaded box).
Figure 4.
Figure 4.
AdCA trial: ex vivo ELISpot IFN-γ activities to CSP and AMA1. The associations of fold-changes of pre-CHMI and post-CHMI activities with pre-CHMI activities are shown as log-transformed values, and the dotted line represents no-change. The shaded box shows ±1.5 range. (A) CSP: there was a significant negative association between fold-change and pre-CHMI activities; activities of all 13/17 positive before CHMI fell after CHMI. (B) AMA1: although activities of all 17/17 subjects that were positive pre-CHMI all fell after CHMI, there was no relationship between fold-change and pre-CHMI activities as only 10/17 were greater than −1.5.
Figure 5.
Figure 5.
AdCA trial: CD8+ T cell IFN-γ activities to CSP and AMA1. The associations of fold-changes of pre-CHMI and post-CHMI activities with pre-CHMI activities are shown as log-transformed values, and the dotted line represents no-change. The shaded box shows ±1.5 range. (A) CSP: there was no significant relationship between fold-change and pre-CHMI activities, although activities fell in 5/15 subjects. (B) AMA1: There was a significant relationship between fold-change and pre-CHMI and activities fell in 9/15 subjects.
Figure 6.
Figure 6.
DNA/Ad and AdCA trials: association of fold changes of antibody responses to CSP and AMA1 with pre-CHMI activities. The associations of fold-changes of pre-CHMI and post-CHMI activities with pre-CHMI activities are shown as log-transformed values, and the dotted line represents no-change. (A) DNA-Ad trial: AMA1: there was no relationship between fold change after CHMI and pre-CHMI activities. (B) AdCA trial: CSP: there was a significant relationship between fold change after CHMI and pre-CHMI activities. (C): AdCA trial: AMA1: there was a stronger significant relationship than CSP between fold change and pre-CHMI activities. In the AdCA trial, CHMI had a greater effect on lower than higher pre-CHMI activities, and for AMA1, CHMI greatly increased pre-CHMI activities.

References

    1. Chuang I, Sedegah M, Cicatelli S, Spring M, Polhemus M, Tamminga C, Patterson N, Guerrero M, Bennett JW, McGrath MG, et al.. DNA prime/adenovirus boost malaria vaccine encoding P. falciparum CSP and AMA1 induces sterile protection associated with cell-mediated immunity. PLoS One 2013; 8:1371; PMID:23457473;
    1. Tamminga C, Sedegah M, Maiolatesi S, Fedders C, Reyes S, Reyes A, Vasquez C, Alcorta Y, Chuang I, Spring M, et al.. Human adenovirus 5-vectored Plasmodium falciparum NMRC-M3V-Ad-PfCA vaccine encoding CSP and AMA1 is safe, well-tolerated and immunogenic but does not protect against controlled human malaria infection. Hum Vaccin Immunother 2013; 9:2165-2177; PMID:23899517;
    1. Sedegah M, Hollingdale MR, Farooq F, Ganeshan H, Belmonte M, Kim Y, Peters B, Sette A, Huang J, McGrath S, et al.. Sterile Immunity to Malaria after DNA Prime/Adenovirus Boost Immunization Is Associated with Effector Memory CD8+T Cells Targeting AMA1 Class I Epitopes. PLoS One 2014; 9:e106241; PMID:25211344;
    1. Zarling S, Berenzon D, Dalai S, Liepinsh D, Steers N, Krzych U. The survival of memory CD8 T cells that is mediated by IL-15 correlates with sustained protection against malaria. J Immunol 2013; 190:5128-5141; PMID:23589611;
    1. Nganou-Makamdop K, van Gemert GJ, Arens T, Hermsen CC, Sauerwein RW. Long term protection after immunization with P. berghei sporozoites correlates with sustained IFNgamma responses of hepatic CD8+ memory T cells. PLoS One 2012; 7:e36508; PMID:22563506;
    1. Epstein JE, Tewari K, Lyke KE, Sim BK, Billingsley PF, Laurens MB, Gunasekera A, Chakravarty S, James ER, Sedegah M, et al.. Live attenuated malaria vaccine designed to protect through hepatic CD8(+) T cell immunity. Science 2011; 334:475-480; PMID:21903775;
    1. Sedegah M, Tamminga C, McGrath S, House B, Ganeshan H, Lejano J, Abot E, Banania G, Sayo R, Farooq F, et al.. Adenovirus 5-vectored P. falciparum Vaccine Expressing CSP and AMA1. Part A: Safety and Immunogenicity in Seronegative Adults. PLoS One 2011; 6:e24586; PMID:22003383;
    1. Tamminga C, Sedegah M, Regis D, Chuang I, Epstein JE, Spring M, Mendoza-Silveiras J, McGrath S, Maiolatesi S, Reyes S, et al.. Adenovirus-5-vectored P. falciparum vaccine expressing CSP and AMA1. Part B: safety, immunogenicity and protective efficacy of the CSP component. PLoS One 2011; 6:e25868; PMID:22003411;
    1. Tamminga C, Sedegah M, Maiolatesi S, Fedders C, Reyes S, Reyes A, Vasquez C, Alcorta Y, Chuang I, Spring M, et al.. Human Adenovirus 5-Vectored Plasmodium falciparum NMRC-M3V-Ad-PfCA Vaccine encoding CSP and AMA1 is safe, well tolerated and immunogenic but does not protect against controlled human malaria infection. Hum Vaccin Immunother 2013; 9:2165-2177; PMID:23899517;
    1. Teirlinck AC, McCall MB, Roestenberg M, Scholzen A, Woestenenk R, de Mast Q, van der Ven AJ, Hermsen CC, Luty AJ, Sauerwein RW. Longevity and composition of cellular immune responses following experimental Plasmodium falciparum malaria infection in humans. PLoS Pathog 2011; 7:e1002389; PMID:22144890;
    1. Elias SC, Choudhary P, de Cassan SC, Biswas S, Collins KA, Halstead FD, Bliss CM, Ewer KJ, Hodgson SH, Duncan CJ, et al.. Analysis of human B-cell responses following ChAd63-MVA MSP1 and AMA1 immunization and controlled malaria infection. Immunology 2014; 141:628-644; PMID:24303947;
    1. Stoyanov CT, Boscardin SB, Deroubaix S, Barba-Spaeth G, Franco D, Nussenzweig RS, Nussenzweig M, Rice CM. Immunogenicity and protective efficacy of a recombinant yellow fever vaccine against the murine malarial parasite Plasmodium yoelii. Vaccine 2010; 28:4644-4652; PMID:20451637;
    1. Biswas S, Choudhary P, Elias SC, Miura K, Milne KH, de Cassan SC, Collins KA, Halstead FD, Bliss CM, Ewer KJ, et al.. Assessment of Humoral Immune Responses to Blood-Stage Malaria Antigens following ChAd63-MVA Immunization, Controlled Human Malaria Infection and Natural Exposure. PLoS One 2014; 9:e107903; PMID:25254500;
    1. Ockenhouse CF, Sun PF, Lanar DE, Wellde BT, Hall BT, Kester K, Stoute JA, Magill A, Krzych U, Farley L, et al.. Phase I/IIa safety, immunogenicity, and efficacy trial of NYVAC-Pf7, a pox-vectored, multiantigen, multistage vaccine candidate for Plasmodium falciparum malaria. J Infect Dis 1998; 177:1664-1673; PMID:9607847;
    1. Hamid MM, Remarque EJ, El Hassan IM, Hussain AA, Narum DL, Thomas AW, Kocken CH, Weiss WR, Faber BW. Malaria infection by sporozoite challenge induces high functional antibody titres against blood stage antigens after a DNA prime, poxvirus boost vaccination strategy in Rhesus macaques. Malar J 2011; 10:29; PMID:21303498;
    1. Tartz S, Deschermeier C, Retzlaff S, Heussler V, Sebo P, Fleischer B, Jacobs T. Plasmodium berghei sporozoite challenge of vaccinated BALB/c mice leads to the induction of humoral immunity and improved function of CD8(+) memory T cells. Eur J Immunol 2012; 43:693-704; PMID:23229763;
    1. Epstein JE, Tewari K, Lyke KE, Sim BK, Billingsley PF, Laurens MB, Gunasekera A, Chakravarty S, James ER, Sedegah M, et al.. Live Attenuated Malaria Vaccine Designed to Protect through Hepatic CD8+ T Cell Immunity. Science 2011; 334(6055):475-480; PMID:21903775;
    1. Seder RA, Chang LJ, Enama ME, Zephir KL, Sarwar UN, Gordon IJ, Holman LA, James ER, Billingsley PF, Gunasekera A, et al.. Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science 2013; 341:1359-1365; PMID:23929949;
    1. Sedegah M, Hollingdale MR, Farooq F, Ganeshan H, Belmonte M, Kim Y, Peters B, Sette A, Huang J, McGrath MG, et al.. Sterile Immunity to Hepatic Stage Malaria after DNA Prime/Adenovirus Boost Immunization is Associated with Effector Memory CD8+T Cells Targeting AMA1 Class I Epitopes. PLoS One 2014; 9(9):e106241; PMID:25211344;
    1. Krzych U, Dalai S, Zarling S, Pichugin A. Memory CD8 T cells specific for plasmodia liver-stage antigens maintain protracted protection against malaria. Front Immunol 2012; 3:370; PMID:23233854;
    1. Tse SW, Radtke AJ, Zavala F. Induction and maintenance of protective CD8+ T cells against malaria liver stages: implications for vaccine development. Mem Inst Oswaldo Cruz 2011; 106(Suppl 1):172-178; PMID:21881772;
    1. Tse SW, Cockburn IA, Zhang H, Scott AL, Zavala F. Unique transcriptional profile of liver-resident memory CD8 T cells induced by immunization with malaria sporozoites. Genes Immun 2013; 14(5):302-9; PMID:23594961;
    1. Chen Q, Amaladoss A, Ye W, Liu M, Dummler S, Kong F, Wong LH, Loo HL, Loh E, Tan SQ, et al.. Human natural killer cells control Plasmodium falciparum infection by eliminating infected red blood cells. Proc Natl Acad Sci U S A 2014; 111:1479-1484; PMID:24474774;
    1. Horowitz A, Hafalla JC, King E, Lusingu J, Dekker D, Leach A, Moris P, Cohen J, Vekemans J, Villafana T, et al.. Antigen-specific IL-2 secretion correlates with NK cell responses after immunization of Tanzanian children with the RTS,S/AS01 malaria vaccine. J Immunol 2012; 188:5054-5062; PMID:22504653;
    1. McCall MB, Roestenberg M, Ploemen I, Teirlinck A, Hopman J, de Mast Q, Dolo A, Doumbo OK, Luty A, van der Ven AJ, et al.. Memory-like IFN-gamma response by NK cells following malaria infection reveals the crucial role of T cells in NK cell activation by P. falciparum. Eur J Immunol 2010; 40:3472-3477; PMID:21072880;
    1. Quinn KM, Da Costa A, Yamamoto A, Berry D, Lindsay RW, Darrah PA, Wang L, Cheng C, Kong WP, Gall JG, et al.. Comparative analysis of the magnitude, quality, phenotype, and protective capacity of simian immunodeficiency virus gag-specific CD8+ T cells following human-, simian-, and chimpanzee-derived recombinant adenoviral vector immunization. J Immunol 2013; 190:2720-2735; PMID:23390298;
    1. Yang SH, Lee CG, Park SH, Im SJ, Kim YM, Son JM, Wang JS, Yoon SK, Song MK, Ambrozaitis A, et al.. Correlation of antiviral T-cell responses with suppression of viral rebound in chronic hepatitis B carriers: a proof-of-concept study. Gene Ther 2006; 13:1110-1117; PMID:16525482;
    1. Wherry EJ, Blattman JN, Murali-Krishna K, van der Most R, Ahmed R. Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. J Virol 2003; 77:4911-4927; PMID:12663797;
    1. Hafalla JC, Claser C, Couper KN, Grau GE, Renia L, de Souza JB, Riley EM. The CTLA-4 and PD-1/PD-L1 inhibitory pathways independently regulate host resistance to Plasmodium-induced acute immune pathology. PLoS Pathog 2012; 8:e1002504; PMID:22319445;
    1. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol 2008; 26:677-704; PMID:18173375;
    1. Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat Immunol 2007; 8:239-245; PMID:17304234;
    1. Wykes MN, Horne-Debets JM, Leow CY, Karunarathne DS. Malaria drives T cells to exhaustion. Front Microbiol 2014; 5:249; PMID:24904561;
    1. Illingworth J, Butler NS, Roetynck S, Mwacharo J, Pierce SK, Bejon P, Crompton PD, Marsh K, Ndungu FM. Chronic exposure to Plasmodium falciparum is associated with phenotypic evidence of B and T cell exhaustion. J Immunol 2013; 190:1038-1047; PMID:23264654;
    1. Thera MA, Doumbo OK, Coulibaly D, Laurens MB, Ouattara A, Kone AK, Guindo AB, Traore K, Traore I, Kouriba B, et al.. A field trial to assess a blood-stage malaria vaccine. N Engl J Med 2011; 365:1004-1013; PMID:21916638;
    1. Bergmann-Leitner ES, Duncan EH, Burge JR, Spring M, Angov E. Miniaturization of a high-throughput pLDH-based Plasmodium falciparum growth inhibition assay for small volume samples from preclinical and clinical vaccine trials. Am J Trop Med Hyg 2008; 78:468-471; PMID:18337345
    1. Hamann D, Baars PA, Rep MH, Hooibrink B, Kerkhof-Garde SR, Klein MR, van Lier RA. Phenotypic and functional separation of memory and effector human CD8+ T cells. J Exp Med 1997; 186:1407-1418; PMID:9348298;
    1. Tamminga C. Adenovirus-5-Vectored P. falciparumVaccine Expressing CSP and AMA1. Part B: Safety, immunogenicity and Protective Efficacy of the CSP Component. PLoS One 2011; 6:e25868; PMID:22003411;
    1. Wessa P. Free Statistics Software. Offic Res Dev Edu 2014; 1(1):23-r7

Source: PubMed

3
Tilaa