Discovery of Novel Plasmodium falciparum Pre-Erythrocytic Antigens for Vaccine Development

Joao C Aguiar, Jessica Bolton, Joyce Wanga, John B Sacci, Hideyuki Iriko, Julie K Mazeika, Eun-Taek Han, Keith Limbach, Noelle B Patterson, Martha Sedegah, Ann-Marie Cruz, Takafumi Tsuboi, Stephen L Hoffman, Daniel Carucci, Michael R Hollingdale, Eileen D Villasante, Thomas L Richie, Joao C Aguiar, Jessica Bolton, Joyce Wanga, John B Sacci, Hideyuki Iriko, Julie K Mazeika, Eun-Taek Han, Keith Limbach, Noelle B Patterson, Martha Sedegah, Ann-Marie Cruz, Takafumi Tsuboi, Stephen L Hoffman, Daniel Carucci, Michael R Hollingdale, Eileen D Villasante, Thomas L Richie

Abstract

Background: Nearly 100% protection against malaria infection can be achieved in humans by immunization with P. falciparum radiation-attenuated sporozoites (RAS). Although it is thought that protection is mediated by T cell and antibody responses, only a few of the many pre-erythrocytic (sporozoite and liver stage) antigens that are targeted by these responses have been identified.

Methodology: Twenty seven P. falciparum pre-erythrocytic antigens were selected using bioinformatics analysis and expression databases and were expressed in a wheat germ cell-free protein expression system. Recombinant proteins were recognized by plasma from RAS-immunized subjects, and 21 induced detectable antibody responses in mice and rabbit and sera from these immunized animals were used to characterize these antigens. All 21 proteins localized to the sporozoite: five localized to the surface, seven localized to the micronemes, cytoplasm, endoplasmic reticulum or nucleus, two localized to the surface and cytoplasm, and seven remain undetermined. PBMC from RAS-immunized volunteers elicited positive ex vivo or cultured ELISpot responses against peptides from 20 of the 21 antigens.

Conclusions: These T cell and antibody responses support our approach of using reagents from RAS-immunized subjects to screen potential vaccine antigens, and have led to the identification of a panel of novel P. falciparum antigens. These results provide evidence to further evaluate these antigens as vaccine candidates.

Trial registration: ClinicalTrials.gov NCT00870987 ClinicalTrials.gov NCT00392015.

Conflict of interest statement

Competing Interests: JCA is an employee of Camris International. JW is an employee of Technical Resources International, Inc. JKM is an employee of EMD Millipore Corporation. There are no patents, products in development, or marketed products to declare. This does not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Fig 1. High-throughput expression and detection of…
Fig 1. High-throughput expression and detection of P. falciparum proteins.
Compartmental wheat germ expression of recombinant proteins represented by five clones as GST (A) and 6xHis fusions (B). [C14]-Leucine labeled proteins detected by radio blot in three fractions; total (T), supernatant (S), and pellet (P). Protein predicted molecular weight sizes are: (1) 32.8 kDa, (2) 50.2 kDa, (3) 37.7 kDa, (4) 59.7 kDa and (5) 32.5 kDa. The molecular sizes for GST-fused proteins (A) include an additional 29 kDa (GST molecular size). The autoradiographs for all the 151 clones are shown in S1 Fig.
Fig 2. Expression and detection of affinity…
Fig 2. Expression and detection of affinity purified P. falciparum proteins.
(A) Coomassie blue stained SDS-PAGE gels of 17 randomly-selected GST-fusion proteins that were affinity purified and cleaved. Asterisk indicates purified protein. (B) Western blot probed with pooled RAS-immune sera. Arrow indicates positive reactivity.
Fig 3. Stage-specific expression of CSP and…
Fig 3. Stage-specific expression of CSP and CelTOS by immunofluorescence IFA and immuno-electron microscopy.
CSP and CelTOS were localized to sporozoites (A, B, D, and E) and 7-day old liver stages (C and F) by IFA (A, C, D, F) and by immune-electron microscopy (B, E). CSP: localized to the sporozoite surface, (arrows, A), outer sporozoites membranes and shed material (arrows B), and the periphery of 5 day liver stage (arrows, C). CelTOS: localized to patches within sporozoites (arrows, D) that are associated with micronemes (arrows, E), was not on the surface, and was in the cytoplasm of five day liver stages (arrow, F).
Fig 4. Stage-specific expression of three novel…
Fig 4. Stage-specific expression of three novel P. falciparum antigens by immunofluorescence and immune-electron microscopy.
Three novel P. falciparum antigens (Pf02, Pf56, Pf78) were localized by immunofluorescence to sporozoites (A, E, I), 7-day liver stages (C, G, K), and blood stages (D, H, L); and by immuno-electron microscopy to sporozoites (B, F, J). Pf02: localized to the sporozoite surface (A), specifically the outer membrane (arrow, B), the cytoplasm of liver stages (C) and cytoplasm of blood stage merozoites (arrows, D). Pf56: localized to patches in sporozoite periphery (arrows, E, F) and sporozoite interior (arrows, F), the cytoplasm of liver stages (arrow, G) and cytoplasm of blood stage merozoites (arrows, H). Pf78: localized as discrete clumps on the sporozoite periphery (arrows, I), that were mostly in the middle and inner surface membranes (arrows, J), as discrete clumps in the liver stage periphery (arrows, K), and in the blood stage schizonts residual body and cytoplasm, but not merozoites (arrows, L).
Fig 5. Stage-specific expression of three novel…
Fig 5. Stage-specific expression of three novel P. falciparum antigens by immunofluorescence and immune-electron microscopy.
Three additional novel P. falciparum antigens (Pf106, Pf121, Pf144) were localized by immunofluorescence to sporozoites (M, Q, U), 7-day liver stages (O, S, W), and blood stages (P, T, X); and by immuno-electron microscopy to sporozoites (N, R, V). Pf106: localized in clumps at the sporozoite anterior pole (arrow, M), in vesicles inside the sporozoite (arrows, N), in liver stage merozoites (arrows, 0), and in blood stage merozoites (arrows, P). Pf121: localized unevenly in the sporozoite periphery/interior (arrows, Q) that appear to localize to micronemes (arrows, R), throughout liver stage parasite (arrow, S) and in clumps in blood stage schizonts but not the residual body (arrows, T). Pf144: localized to clumps inside the sporozoite (arrow, U) that are associated with micronemes (arrows, V), but is only weakly detected in 7-day liver stage parasites (arrow, W) and is dispersed in the blood stage schizonts (arrow, X).
Fig 6. Antigen-specific IFN-γ ELISpot activity of…
Fig 6. Antigen-specific IFN-γ ELISpot activity of protected and non-protected RAS-immunized volunteers targeting CelTOS and CSP.
CelTOS (circles) and CSP (triangles) overlapping peptide pools spanning full length CSP or CelTOS were used to stimulate T cell responses of three protected (v30, v58, v64) and five non-protected (v20, v43, v52, v53, v65) RAS-immunized subjects, and responses were measured by IFN-γ ELISpot, and expressed as spot-forming cells/million PBMC (sf/m). A. In ex vivo assays, samples were scored as positive (filled markers) using high stringency criteria (significant difference between test antigen and medium controls using Student’s t-test, at least a doubling and a difference of at least 10 sfc/m (See Methods) or as negative (open markers). Occasional values of activities that were negative exceeded positive values if the pre-immunization activities were high. Overall, the CelTOS-specific T cell responses among protected subjects (geometric mean 98 sfc/m) were significantly higher than the CelTOS-specific T cell responses of non-protected subjects (geometric mean 12 sfc/m, p = <0.001, Mann-Whitney U test), whereas CSP-specific T cell responses were similar among protected (geometric mean 18 sfc/m) and non-protected (geometric mean 11 sfc/m, not significant) subjects. B. In cultured ELISpot assays, CelTOS-specific T cell responses of protected subjects (geometric mean 505 sfc/m) were also significantly higher than non-protected subjects (geometric mean 157 sfc/m, p = 0.002, Mann-Whitney U test). CSP-specific T cell responses of protected subjects (geometric mean 215 sfc/m) were similar to those of non-protected subjects (geometric mean 235 sfc/m, not significant).

References

    1. Weiss WR, Good MF, Hollingdale MR, Miller LH, Berzofsky JA. Genetic control of immunity to Plasmodium yoelii sporozoites. J Immunol. 1989;143(12):4263–6.
    1. Nussenzweig V, Nussenzweig RS. Circumsporozoite proteins of malaria parasites. Cell. 1985;42(2):401–3.
    1. Gwadz RW, Cochrane AH, Nussenzweig V, Nussenzweig RS. Preliminary studies on vaccination of rhesus monkeys with irradiated sporozoites of Plasmodium knowlesi and characterization of surface antigens of these parasites. Bull World Health Organ. 1979;57 Suppl 1:165–73.
    1. Clyde DF. Immunization of man against falciparum and vivax malaria by use of attenuated sporozoites. Am J Trop Med Hyg. 1975;24(3):397–401.
    1. Rieckmann KH, Beaudoin RL, Cassells JS, Sell KW. Use of attenuated sporozoites in the immunization of human volunteers against falciparum malaria. Bull World Health Organ. 1979;57 Suppl 1:261–5.
    1. Seder RA, Chang LJ, Enama ME, Zephir KL, Sarwar UN, Gordon IJ, et al. Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science. 2013;341(6152):1359–65. 10.1126/science.1241800
    1. Roestenberg M, McCall M, Hopman J, Wiersma J, Luty AJ, van Gemert GJ, et al. Protection against a malaria challenge by sporozoite inoculation. N Engl J Med. 2009;361(5):468–77. 10.1056/NEJMoa0805832
    1. Roestenberg M, Teirlinck AC, McCall MB, Teelen K, Makamdop KN, Wiersma J, et al. Long-term protection against malaria after experimental sporozoite inoculation: an open-label follow-up study. Lancet. 2011;377(9779):1770–6. 10.1016/S0140-6736(11)60360-7
    1. Khan SM, Janse CJ, Kappe SH, Mikolajczak SA. Genetic engineering of attenuated malaria parasites for vaccination. Current opinion in biotechnology. 2012;23(6):908–16. 10.1016/j.copbio.2012.04.003
    1. Spring M, Murphy J, Nielsen R, Dowler M, Bennett JW, Zarling S, et al. First-in-human evaluation of genetically attenuated Plasmodium falciparum sporozoites administered by bite of Anopheles mosquitoes to adult volunteers. Vaccine. 2013;31(43):4975–83. 10.1016/j.vaccine.2013.08.007
    1. Ferreira A, Schofield L, Enea V, Schellekens H, van der Meide P, Collins WE, et al. Inhibition of development of exoerythrocytic forms of malaria parasites by gamma-interferon. Science. 1986;232(4752):881–4.
    1. Kumar KA, Sano G, Boscardin S, Nussenzweig RS, Nussenzweig MC, Zavala F, et al. The circumsporozoite protein is an immunodominant protective antigen in irradiated sporozoites. Nature. 2006;444(7121):937–40.
    1. Weiss WR, Mellouk S, Houghten RA, Sedegah M, Kumar S, Good MF, et al. Cytotoxic T cells recognize a peptide from the circumsporozoite protein on malaria-infected hepatocytes. The Journal of experimental medicine. 1990;171(3):763–73.
    1. Weiss WR, Sedegah M, Beaudoin RL, Miller LH, Good MF. CD8+ T cells (cytotoxic/suppressors) are required for protection in mice immunized with malaria sporozoites. Proc Natl Acad Sci U S A. 1988;85(2):573–6.
    1. Nussenzweig RS, Vanderberg J, Most H, Orton C. Protective immunity produced by the injection of x-irradiated sporozoites of plasmodium berghei. Nature. 1967;216(5111):160–2.
    1. Romero P, Maryanski JL, Corradin G, Nussenzweig RS, Nussenzweig V, Zavala F. Cloned cytotoxic T cells recognize an epitope in the circumsporozoite protein and protect against malaria. Nature. 1989;341(6240):323–6.
    1. Weiss WR, Jiang CG. Protective CD8+ T lymphocytes in primates immunized with malaria sporozoites. PloS one. 2012;7(2):e31247 10.1371/journal.pone.0031247
    1. Hoffman SL, Goh LM, Luke TC, Schneider I, Le TP, Doolan DL, et al. Protection of humans against malaria by immunization with radiation-attenuated Plasmodium falciparum sporozoites. J Infect Dis. 2002;185(8):1155–64.
    1. Lindner SE, Swearingen KE, Harupa A, Vaughan AM, Sinnis P, Moritz RL, et al. Total and putative surface proteomics of malaria parasite salivary gland sporozoites. Molecular & cellular proteomics: MCP. 2013;12(5):1127–43.
    1. Olotu A, Fegan G, Wambua J, Nyangweso G, Awuondo KO, Leach A, et al. Four-year efficacy of RTS,S/AS01E and its interaction with malaria exposure. N Engl J Med. 2013;368(12):1111–20. 10.1056/NEJMoa1207564
    1. Moorthy VS, Ballou WR. Immunological mechanisms underlying protection mediated by RTS,S: a review of the available data. Malar J. 2009;8(1):312.
    1. Kariu T, Ishino T, Yano K, Chinzei Y, Yuda M. CelTOS, a novel malarial protein that mediates transmission to mosquito and vertebrate hosts. Molecular microbiology. 2006;59(5):1369–79.
    1. Bergmann-Leitner ES, Mease RM, De La Vega P, Savranskaya T, Polhemus M, Ockenhouse C, et al. Immunization with pre-erythrocytic antigen CelTOS from Plasmodium falciparum elicits cross-species protection against heterologous challenge with Plasmodium berghei. PloS one. 2010;5(8):e12294 10.1371/journal.pone.0012294
    1. Khusmith S, Charoenvit Y, Kumar S, Sedegah M, Beaudoin RL, Hoffman SL. Protection against malaria by vaccination with sporozoite surface protein 2 plus CS protein. Science. 1991;252(5006):715–8.
    1. Robson KJ, Hall JR, Jennings MW, Harris TJ, Marsh K, Newbold CI, et al. A highly conserved amino-acid sequence in thrombospondin, properdin and in proteins from sporozoites and blood stages of a human malaria parasite. Nature. 1988;335(6185):79–82.
    1. Sanchez GI, Rogers WO, Mellouk S, Hoffman SL. Plasmodium falciparum: exported protein-1, a blood stage antigen, is expressed in liver stage parasites. Exp Parasitol. 1994;79(1):59–62.
    1. Doolan DL, Sedegah M, Hedstrom RC, Hobart P, Charoenvit Y, Hoffman SL. Circumventing genetic restriction of protection against malaria with multigene DNA immunization: CD8+ cell-, interferon gamma-, and nitric oxide-dependent immunity. The Journal of experimental medicine. 1996;183(4):1739–46.
    1. Moelans II, Meis JF, Kocken C, Konings RN, Schoenmakers JG. A novel protein antigen of the malaria parasite Plasmodium falciparum, located on the surface of gametes and sporozoites. Mol Biochem Parasitol. 1991;45(2):193–204.
    1. Fidock DA, Bottius E, Brahimi K, Moelans II, Aikawa M, Konings RN, et al. Cloning and characterization of a novel Plasmodium falciparum sporozoite surface antigen, STARP. Mol Biochem Parasitol. 1994;64(2):219–32.
    1. Guerin-Marchand C, Druilhe P, Galey B, Londono A, Patarapotikul J, Beaudoin RL, et al. A liver-stage-specific antigen of Plasmodium falciparum characterized by gene cloning. Nature. 1987;329(6135):164–7.
    1. Zhu J, Hollingdale MR. Structure of Plasmodium falciparum liver stage antigen-1. Mol Biochem Parasitol. 1991;48(2):223–6.
    1. Mauduit M, Gruner AC, Tewari R, Depinay N, Kayibanda M, Chavatte JM, et al. A role for immune responses against non-CS components in the cross-species protection induced by immunization with irradiated malaria sporozoites. PloS one. 2009;4(11):e7717 10.1371/journal.pone.0007717
    1. Mishra S, Rai U, Shiratsuchi T, Li X, Vanloubbeeck Y, Cohen J, et al. Identification of non-CSP antigens bearing CD8 epitopes in mice immunized with irradiated sporozoites. Vaccine. 2011;29(43):7335–42. 10.1016/j.vaccine.2011.07.081
    1. Doolan DL, Hoffman SL. The complexity of protective immunity against liver-stage malaria. J Immunol. 2000;165(3):1453–62.
    1. Epstein JE, Tewari K, Lyke KE, Sim BK, Billingsley PF, Laurens MB, et al. Live Attenuated Malaria Vaccine Designed to Protect through Hepatic CD8+ T Cell Immunity. Science. 2011;334(6055):475–80. 10.1126/science.1211548
    1. Florens L, Washburn MP, Raine JD, Anthony RM, Grainger M, Haynes JD, et al. A proteomic view of the Plasmodium falciparum life cycle. Nature. 2002;419(6906):520–6.
    1. Trieu A, Kayala MA, Burk C, Molina DM, Freilich DA, Richie TL, et al. Sterile protective immunity to malaria is associated with a panel of novel P. falciparum antigens. Molecular & cellular proteomics: MCP. 2011;10(9):M111 007948.
    1. Doolan DL, Southwood S, Freilich DA, Sidney J, Graber NL, Shatney L, et al. Identification of Plasmodium falciparum antigens by antigenic analysis of genomic and proteomic data. Proc Natl Acad Sci U S A. 2003;100(17):9952–7.
    1. Gardner MJ, Hall N, Fung E, White O, Berriman M, Hyman RW, et al. Genome sequence of the human malaria parasite Plasmodium falciparum. Nature. 2002;419(6906):498–511.
    1. Doolan DL, Aguiar JC, Weiss WR, Sette A, Felgner PL, Regis DP, et al. Utilization of genomic sequence information to develop malaria vaccines. J Exp Biol. 2003;206(Pt 21):3789–802.
    1. Doolan DL, Mu Y, Unal B, Sundaresh S, Hirst S, Valdez C, et al. Profiling humoral immune responses to P. falciparum infection with protein microarrays. Proteomics. 2008;8(22):4680–94. 10.1002/pmic.200800194
    1. Sacci JB Jr., Ribeiro JM, Huang F, Alam U, Russell JA, Blair PL, et al. Transcriptional analysis of in vivo Plasmodium yoelii liver stage gene expression. Mol Biochem Parasitol. 2005;142(2):177–83.
    1. Tarun AS, Peng X, Dumpit RF, Ogata Y, Silva-Rivera H, Camargo N, et al. A combined transcriptome and proteome survey of malaria parasite liver stages. Proc Natl Acad Sci U S A. 2008;105(1):305–10. 10.1073/pnas.0710780104
    1. Haddad D, Bilcikova E, Witney AA, Carlton JM, White CE, Blair PL, et al. Novel antigen identification method for discovery of protective malaria antigens by rapid testing of DNA vaccines encoding exons from the parasite genome. Infect Immun. 2004;72(3):1594–602.
    1. Albuquerque SS, Carret C, Grosso AR, Tarun AS, Peng X, Kappe SH, et al. Host cell transcriptional profiling during malaria liver stage infection reveals a coordinated and sequential set of biological events. BMC Genomics. 2009;10:270 10.1186/1471-2164-10-270
    1. Aguiar JC, LaBaer J, Blair PL, Shamailova VY, Koundinya M, Russell JA, et al. High-throughput generation of P. falciparum functional molecules by recombinational cloning. Genome research. 2004;14(10B):2076–82.
    1. Le Roch KG, Zhou Y, Blair PL, Grainger M, Moch JK, Haynes JD, et al. Discovery of gene function by expression profiling of the malaria parasite life cycle. Science. 2003;301(5639):1503–8.
    1. Sedegah M, Charoenvit Y, Aguiar J, Sacci J, Hedstrom R, Kumar S, et al. Effect on antibody and T-cell responses of mixing five GMP-produced DNA plasmids and administration with plasmid expressing GM-CSF. Genes Immun. 2004;5(7):553–61.
    1. Mehlin C, Boni E, Buckner FS, Engel L, Feist T, Gelb MH, et al. Heterologous expression of proteins from Plasmodium falciparum: results from 1000 genes. Mol Biochem Parasitol. 2006;148(2):144–60.
    1. Vedadi M, Lew J, Artz J, Amani M, Zhao Y, Dong A, et al. Genome-scale protein expression and structural biology of Plasmodium falciparum and related Apicomplexan organisms. Mol Biochem Parasitol. 2007;151(1):100–10.
    1. Kolb VA, Makeyev EV, Spirin AS. Co-translational folding of an eukaryotic multidomain protein in a prokaryotic translation system. J Biol Chem. 2000;275(22):16597–601.
    1. Tsuboi T, Takeo S, Iriko H, Jin L, Tsuchimochi M, Matsuda S, et al. Wheat germ cell-free system-based production of malaria proteins for discovery of novel vaccine candidates. Infect Immun. 2008;76(4):1702–8. 10.1128/IAI.01539-07
    1. Arumugam TU, Ito D, Takashima E, Tachibana M, Ishino T, Torii M, et al. Application of wheat germ cell-free protein expression system for novel malaria vaccine candidate discovery. Expert Rev Vaccines. 2014;13(1):75–85. 10.1586/14760584.2014.861747
    1. Sawasaki T, Ogasawara T, Morishita R, Endo Y. A cell-free protein synthesis system for high-throughput proteomics. Proc Natl Acad Sci U S A. 2002;99(23):14652–7.
    1. Wirtz RA, Zavala F, Charoenvit Y, Campbell GH, Burkot TR, Schneider I, et al. Comparative testing of monoclonal antibodies against Plasmodium falciparum sporozoites for ELISA development. Bull World Health Organ. 1987;65(1):39–45.
    1. Sacci JB Jr., Alam U, Douglas D, Lewis J, Tyrrell DL, Azad AF, et al. Plasmodium falciparum infection and exoerythrocytic development in mice with chimeric human livers. Int J Parasitol. 2006;36(3):353–60.
    1. Sedegah M, Tamminga C, McGrath S, House B, Ganeshan H, Lejano J, et al. Adenovirus 5-vectored P. falciparum Vaccine Expressing CSP and AMA1. Part A: Safety and Immunogenicity in Seronegative Adults. PloS one. 2011;6(10):e24586 10.1371/journal.pone.0024586
    1. Dodoo D, Hollingdale MR, Anum D, Koram KA, Gyan B, Akanmori BD, et al. Measuring naturally acquired immune responses to candidate malaria vaccine antigens in Ghanaian adults. Malar J. 2011;10:168 10.1186/1475-2875-10-168
    1. Reece WH, Pinder M, Gothard PK, Milligan P, Bojang K, Doherty T, et al. A CD4(+) T-cell immune response to a conserved epitope in the circumsporozoite protein correlates with protection from natural Plasmodium falciparum infection and disease. Nat Med. 2004;10(4):406–10.
    1. Dunachie SJ, Walther M, Epstein JE, Keating S, Berthoud T, Andrews L, et al. A DNA prime-modified vaccinia virus ankara boost vaccine encoding thrombospondin-related adhesion protein but not circumsporozoite protein partially protects healthy malaria-naive adults against Plasmodium falciparum sporozoite challenge. Infect Immun. 2006;74(10):5933–42.
    1. Aikawa M, Cochrane AH, Nussenzweig RS, Rabbege J. Freeze-fracture study of malaria sporozoites: antibody-induced changes of the pellicular membrane. J Protozool. 1979;26(2):273–9.
    1. Posthuma G, Meis JF, Verhave JP, Gigengack S, Hollingdale MR, Ponnudurai T, et al. Immunogold determination of Plasmodium falciparum circumsporozoite protein in Anopheles stephensi salivary gland cells. Eur J Cell Biol. 1989;49(1):66–72.
    1. Bergmann-Leitner ES, Legler PM, Savranskaya T, Ockenhouse CF, Angov E. Cellular and humoral immune effector mechanisms required for sterile protection against sporozoite challenge induced with the novel malaria vaccine candidate CelTOS. Vaccine. 2011;29(35):5940–9. 10.1016/j.vaccine.2011.06.053
    1. Hollingdale MR, Leland P, Leef JL, Leef MF, Beaudoin RL. Serological reactivity of in vitro cultured exoerythrocytic stages of Plasmodium berghei in indirect immunofluorescent or immunoperoxidase antibody tests. Am J Trop Med Hyg. 1983;32(1):24–30.
    1. Dunachie SJ, Walther M, Vuola JM, Webster DP, Keating SM, Berthoud T, et al. A clinical trial of prime-boost immunisation with the candidate malaria vaccines RTS,S/AS02A and MVA-CS. Vaccine. 2006;24(15):2850–9.
    1. Chuang I, Sedegah M, Cicatelli S, Spring M, Polhemus M, Tamminga C, 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(2):1371.
    1. Sedegah M, Kim D, Ganeshan H, Huang J, Belmonte M, Abot E, et al. Identification of minimal human MHC-restricted CD8+ T-cell epitopes within the Plasmodium falciparum circumsporozoite protein (CSP). Malar J. 2013;12:185 10.1186/1475-2875-12-185
    1. Washburn MP, Wolters D, Yates JR 3rd. Large-scale analysis of the yeast proteome by multidimensional protein identification technology. Nature biotechnology. 2001;19(3):242–7.
    1. Krzych U, Dalai S, Zarling S, Pichugin A. Memory CD8 T cells specific for plasmodia liver-stage antigens maintain protracted protection against malaria. Frontiers in immunology. 2012;3:370 10.3389/fimmu.2012.00370
    1. Calarota SA, Baldanti F. Enumeration and characterization of human memory T cells by enzyme-linked immunospot assays. Clinical & developmental immunology. 2013;2013:637649.
    1. Limbach K, Aguiar J, Gowda K, Patterson N, Abot E, Sedegah M, et al. Identification of two new protective pre-erythrocytic malaria vaccine antigen candidates. Malar J. 2011;10:65 10.1186/1475-2875-10-65
    1. Rennenberg A, Lehmann C, Heitmann A, Witt T, Hansen G, Nagarajan K, et al. Exoerythrocytic Plasmodium parasites secrete a cysteine protease inhibitor involved in sporozoite invasion and capable of blocking cell death of host hepatocytes. PLoS Pathog. 2010;6(3):e1000825 10.1371/journal.ppat.1000825
    1. Boysen KE, Matuschewski K. Inhibitor of cysteine proteases is critical for motility and infectivity of Plasmodium sporozoites. mBio. 2013;4(6):e00874–13. 10.1128/mBio.00874-13

Source: PubMed

3
Předplatit