DNA prime/Adenovirus boost malaria vaccine encoding P. falciparum CSP and AMA1 induces sterile protection associated with cell-mediated immunity

Ilin Chuang, Martha Sedegah, Susan Cicatelli, Michele Spring, Mark Polhemus, Cindy Tamminga, Noelle Patterson, Melanie Guerrero, Jason W Bennett, Shannon McGrath, Harini Ganeshan, Maria Belmonte, Fouzia Farooq, Esteban Abot, Jo Glenna Banania, Jun Huang, Rhonda Newcomer, Lisa Rein, Dianne Litilit, Nancy O Richie, Chloe Wood, Jittawadee Murphy, Robert Sauerwein, Cornelus C Hermsen, Andrea J McCoy, Edwin Kamau, James Cummings, Jack Komisar, Awalludin Sutamihardja, Meng Shi, Judith E Epstein, Santina Maiolatesi, Donna Tosh, Keith Limbach, Evelina Angov, Elke Bergmann-Leitner, Joseph T Bruder, Denise L Doolan, C Richter King, Daniel Carucci, Sheetij Dutta, Lorraine Soisson, Carter Diggs, Michael R Hollingdale, Christian F Ockenhouse, Thomas L Richie, Ilin Chuang, Martha Sedegah, Susan Cicatelli, Michele Spring, Mark Polhemus, Cindy Tamminga, Noelle Patterson, Melanie Guerrero, Jason W Bennett, Shannon McGrath, Harini Ganeshan, Maria Belmonte, Fouzia Farooq, Esteban Abot, Jo Glenna Banania, Jun Huang, Rhonda Newcomer, Lisa Rein, Dianne Litilit, Nancy O Richie, Chloe Wood, Jittawadee Murphy, Robert Sauerwein, Cornelus C Hermsen, Andrea J McCoy, Edwin Kamau, James Cummings, Jack Komisar, Awalludin Sutamihardja, Meng Shi, Judith E Epstein, Santina Maiolatesi, Donna Tosh, Keith Limbach, Evelina Angov, Elke Bergmann-Leitner, Joseph T Bruder, Denise L Doolan, C Richter King, Daniel Carucci, Sheetij Dutta, Lorraine Soisson, Carter Diggs, Michael R Hollingdale, Christian F Ockenhouse, Thomas L Richie

Abstract

Background: Gene-based vaccination using prime/boost regimens protects animals and humans against malaria, inducing cell-mediated responses that in animal models target liver stage malaria parasites. We tested a DNA prime/adenovirus boost malaria vaccine in a Phase 1 clinical trial with controlled human malaria infection.

Methodology/principal findings: The vaccine regimen was three monthly doses of two DNA plasmids (DNA) followed four months later by a single boost with two non-replicating human serotype 5 adenovirus vectors (Ad). The constructs encoded genes expressing P. falciparum circumsporozoite protein (CSP) and apical membrane antigen-1 (AMA1). The regimen was safe and well-tolerated, with mostly mild adverse events that occurred at the site of injection. Only one AE (diarrhea), possibly related to immunization, was severe (Grade 3), preventing daily activities. Four weeks after the Ad boost, 15 study subjects were challenged with P. falciparum sporozoites by mosquito bite, and four (27%) were sterilely protected. Antibody responses by ELISA rose after Ad boost but were low (CSP geometric mean titer 210, range 44-817; AMA1 geometric mean micrograms/milliliter 11.9, range 1.5-102) and were not associated with protection. Ex vivo IFN-γ ELISpot responses after Ad boost were modest (CSP geometric mean spot forming cells/million peripheral blood mononuclear cells 86, range 13-408; AMA1 348, range 88-1270) and were highest in three protected subjects. ELISpot responses to AMA1 were significantly associated with protection (p = 0.019). Flow cytometry identified predominant IFN-γ mono-secreting CD8+ T cell responses in three protected subjects. No subjects with high pre-existing anti-Ad5 neutralizing antibodies were protected but the association was not statistically significant.

Significance: The DNA/Ad regimen provided the highest sterile immunity achieved against malaria following immunization with a gene-based subunit vaccine (27%). Protection was associated with cell-mediated immunity to AMA1, with CSP probably contributing. Substituting a low seroprevalence vector for Ad5 and supplementing CSP/AMA1 with additional antigens may improve protection.

Trial registration: ClinicalTrials.govNCT00870987.

Conflict of interest statement

Competing Interests: CD and LS from USAID (funders) played a role in study design. JTB and CRK worked for Gen Vec, Inc. (financial interest in the vaccine) and helped design the vaccine constructs. This does not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Figure 1. Trial design.
Figure 1. Trial design.
Subjects were immunized week 0, 4, 8 and 24 and challenged week 28 (blue arrows). Samples for measuring cell-mediated immunity (ELISpot assay and flow cytometry) were collected at six time points (black arrows), and for measuring antibody levels (ELISA, IFA and growth inhibition assay) at similar time points plus after the DNA immunizations (gray arrows). See text for details.
Figure 2. Schematic of DNA and Adenovirus…
Figure 2. Schematic of DNA and Adenovirus CSP and AMA1 vaccines.
Each panel presents the native protein (top of each panel) and the protein expressed by the DNA or Ad construct (middle and bottom of each panel) for the CSP (Panel A) and AMA1 (Panel B) vaccine antigens. N = amino terminus; C = carboxy terminus; TPA = human tissue plasminogen activator signal sequence; TM = transmembrane domain. See text for explanation. Identical colors indicate identical sequences. Not represented is a single amino acid substitution (G → R) in the AMA DNA construct at position 143.
Figure 3. Flow diagram of immunized and…
Figure 3. Flow diagram of immunized and control volunteers.
Thirty-seven volunteers met all eligibility criteria and were allocated to the immunization group (n = 20) and infectivity controls (n = 6), and 11 were either alternates (n = 6) or not used. WBC = white blood count; DVT = deep venous thrombosis. See text for explanation.
Figure 4. Development of parasitemia in the…
Figure 4. Development of parasitemia in the immunized and infectivity control subjects.
Panel A: Parasitemia-free survival curves (Kaplan-Meier) for immunized volunteers and infectivity controls based on microscopic examination of peripheral blood smears. Panel B: Quantitative(q)-PCR measurements of parasitemia in immunized and challenge controls (error bars show standard deviation) (see reference 28).
Figure 5. Pre-existing NAb to Ad5 may…
Figure 5. Pre-existing NAb to Ad5 may interfere with protection.
Pre-existing NAb titers to Ad5 were measured prior to Ad immunization and are compared with days to patency by microscopy after CHMI. Three of six volunteers who were seronegative (NAb titer 500 (above horizontal red line) became patent at a rate similar to subjects with NAb titer

Figure 6. Antibody responses by ELISA to…

Figure 6. Antibody responses by ELISA to CSP and AMA1.

The box plots (see Statistical…

Figure 6. Antibody responses by ELISA to CSP and AMA1.
The box plots (see Statistical Analysis section for description) represent anti-CSP titers and anti-AMA1 antibody concentration in µg/mL by ELISA for all 15 challenged volunteers. The time points on the x-axis are described in Figure 1. Four protected volunteers are shown as larger, color-coded dots. For the protected volunteers, the antibody titer to CSP of v11 post-DNA and the antibody concentration to AMA1 of v11 post-Ad are box plot outliers. Group geomean CSP and AMA1 ELISA activities for the fifteen recipients were significantly higher than baseline (*) post-DNA, post-Ad, post-Ch and post-Ch final relative to pre-immunization levels (p = 

Figure 7. Ex vivo T cell IFN-γ…

Figure 7. Ex vivo T cell IFN-γ activities by ELISpot Assay for CSP and AMA1.

Figure 7. Ex vivo T cell IFN-γ activities by ELISpot Assay for CSP and AMA1.
The box plots (see Statistical Analysis section for description) represent CSP and AMA1 IFN-γ ELISpot responses (summed peptide pool-specific responses) in spot forming cells per million PBMCs for all 15 challenged volunteers. The time points on the x-axis are described in Figure 1. For the protected volunteers, the IFN-γ ELISpot responses to CSP of v11 and v18 post-Ad are box plot suspected outliers. Group geomean CSP and AMA1 IFN-γ ELISpot activities for the fifteen recipients were not significantly higher than baseline post-DNA, pre-Ad, post-Ad, post-Ch or post-Ch final relative to pre-immunization levels (CSP p = 0.057, AMA1 p = 0.16, mixed linear model).

Figure 8. IFN-γ activities by flow cytometry…

Figure 8. IFN-γ activities by flow cytometry for CSP and AMA1.

The box plots (see…

Figure 8. IFN-γ activities by flow cytometry for CSP and AMA1.
The box plots (see Statistical Analysis section for description) represent IFN-γ -producing CD4+ or CD8+ T cell frequencies as percentage of gated CD4+ or CD8+ T cells, measured by flow cytometry assays after stimulation with a single CSP or AMA1 megapool containing all individual peptide pools for each antigen, for all 15 challenged volunteers. The time points on the x-axis are described in Figure 1. The four protected volunteers are shown as larger, color-coded dots. For the protected volunteers, the CD4+ T cell AMA1 response of v06 at pre-Ad is a box plot outlier, and the CD8+ T cell CSP responses of v11 and v18 post-Ad, and the CD8+ T cell AMA1 responses of v18 post-Ad and post-Ch are box plot suspected outliers. The dotted lines represent positive cutoff (0.03% as described in Methods). IFN-γ -producing CD4+ T cell frequencies were significantly higher than baseline (*) post-DNA (p = 0.047), post-Ad (p = 0.0097) and post-Ch (p = 0.004) for AMA1 (mixed linear model). IFN-γ -producing CD8+ T cell frequencies were significantly higher than baseline (*) post-Ad for CSP (p = 0.007) and post-Ad for AMA1 (0.002) (mixed linear model).
All figures (8)
Similar articles
Cited by
References
    1. WHO (2011) World Malaria Report.
    1. PMI (2011) Fifth Annual Report to Congress.
    1. Clyde DF (1975) Immunization of man against falciparum and vivax malaria by use of attenuated sporozoites. Am J Trop Med Hyg 24: 397–401. - PubMed
    1. Roestenberg M, McCall M, Hopman J, Wiersma J, Luty AJ, et al. (2009) Protection against a malaria challenge by sporozoite inoculation. N Engl J Med 361: 468–477. - PubMed
    1. Roestenberg M, Teirlinck AC, McCall MB, Teelen K, Makamdop KN, et al. (2011) Long-term protection against malaria after experimental sporozoite inoculation: an open-label follow-up study. Lancet 377: 1770–1776. - PubMed
Show all 53 references
Publication types
MeSH terms
Associated data
Related information
Grant support
This work was supported by USAID “Development of Adenovirus-Vectored Malaria Vaccines” Grant #: GHA-P-00-03-00006-01, Project Number 936-3118; and the Congressionally Directed Medical Research Program “Development of Recombinant Adenoviral-based Vaccines against Malaria” Grant #: W81XWH-05-2-0041. Website: https://cdmrp.org. Military Infectious Research Program “Phase 1/2a clinical trials assessing the safety, tolerability, immunogenicity &protective efficacy of Ad5-CA, a two-antigen, adenovirus-vectored Plasmodium falciparum malaria vaccine, in healthy, malaria-nave adults”, work unit number 62787A 870 F 1432. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 6. Antibody responses by ELISA to…
Figure 6. Antibody responses by ELISA to CSP and AMA1.
The box plots (see Statistical Analysis section for description) represent anti-CSP titers and anti-AMA1 antibody concentration in µg/mL by ELISA for all 15 challenged volunteers. The time points on the x-axis are described in Figure 1. Four protected volunteers are shown as larger, color-coded dots. For the protected volunteers, the antibody titer to CSP of v11 post-DNA and the antibody concentration to AMA1 of v11 post-Ad are box plot outliers. Group geomean CSP and AMA1 ELISA activities for the fifteen recipients were significantly higher than baseline (*) post-DNA, post-Ad, post-Ch and post-Ch final relative to pre-immunization levels (p = 

Figure 7. Ex vivo T cell IFN-γ…

Figure 7. Ex vivo T cell IFN-γ activities by ELISpot Assay for CSP and AMA1.

Figure 7. Ex vivo T cell IFN-γ activities by ELISpot Assay for CSP and AMA1.
The box plots (see Statistical Analysis section for description) represent CSP and AMA1 IFN-γ ELISpot responses (summed peptide pool-specific responses) in spot forming cells per million PBMCs for all 15 challenged volunteers. The time points on the x-axis are described in Figure 1. For the protected volunteers, the IFN-γ ELISpot responses to CSP of v11 and v18 post-Ad are box plot suspected outliers. Group geomean CSP and AMA1 IFN-γ ELISpot activities for the fifteen recipients were not significantly higher than baseline post-DNA, pre-Ad, post-Ad, post-Ch or post-Ch final relative to pre-immunization levels (CSP p = 0.057, AMA1 p = 0.16, mixed linear model).

Figure 8. IFN-γ activities by flow cytometry…

Figure 8. IFN-γ activities by flow cytometry for CSP and AMA1.

The box plots (see…

Figure 8. IFN-γ activities by flow cytometry for CSP and AMA1.
The box plots (see Statistical Analysis section for description) represent IFN-γ -producing CD4+ or CD8+ T cell frequencies as percentage of gated CD4+ or CD8+ T cells, measured by flow cytometry assays after stimulation with a single CSP or AMA1 megapool containing all individual peptide pools for each antigen, for all 15 challenged volunteers. The time points on the x-axis are described in Figure 1. The four protected volunteers are shown as larger, color-coded dots. For the protected volunteers, the CD4+ T cell AMA1 response of v06 at pre-Ad is a box plot outlier, and the CD8+ T cell CSP responses of v11 and v18 post-Ad, and the CD8+ T cell AMA1 responses of v18 post-Ad and post-Ch are box plot suspected outliers. The dotted lines represent positive cutoff (0.03% as described in Methods). IFN-γ -producing CD4+ T cell frequencies were significantly higher than baseline (*) post-DNA (p = 0.047), post-Ad (p = 0.0097) and post-Ch (p = 0.004) for AMA1 (mixed linear model). IFN-γ -producing CD8+ T cell frequencies were significantly higher than baseline (*) post-Ad for CSP (p = 0.007) and post-Ad for AMA1 (0.002) (mixed linear model).
All figures (8)
Figure 7. Ex vivo T cell IFN-γ…
Figure 7. Ex vivo T cell IFN-γ activities by ELISpot Assay for CSP and AMA1.
The box plots (see Statistical Analysis section for description) represent CSP and AMA1 IFN-γ ELISpot responses (summed peptide pool-specific responses) in spot forming cells per million PBMCs for all 15 challenged volunteers. The time points on the x-axis are described in Figure 1. For the protected volunteers, the IFN-γ ELISpot responses to CSP of v11 and v18 post-Ad are box plot suspected outliers. Group geomean CSP and AMA1 IFN-γ ELISpot activities for the fifteen recipients were not significantly higher than baseline post-DNA, pre-Ad, post-Ad, post-Ch or post-Ch final relative to pre-immunization levels (CSP p = 0.057, AMA1 p = 0.16, mixed linear model).
Figure 8. IFN-γ activities by flow cytometry…
Figure 8. IFN-γ activities by flow cytometry for CSP and AMA1.
The box plots (see Statistical Analysis section for description) represent IFN-γ -producing CD4+ or CD8+ T cell frequencies as percentage of gated CD4+ or CD8+ T cells, measured by flow cytometry assays after stimulation with a single CSP or AMA1 megapool containing all individual peptide pools for each antigen, for all 15 challenged volunteers. The time points on the x-axis are described in Figure 1. The four protected volunteers are shown as larger, color-coded dots. For the protected volunteers, the CD4+ T cell AMA1 response of v06 at pre-Ad is a box plot outlier, and the CD8+ T cell CSP responses of v11 and v18 post-Ad, and the CD8+ T cell AMA1 responses of v18 post-Ad and post-Ch are box plot suspected outliers. The dotted lines represent positive cutoff (0.03% as described in Methods). IFN-γ -producing CD4+ T cell frequencies were significantly higher than baseline (*) post-DNA (p = 0.047), post-Ad (p = 0.0097) and post-Ch (p = 0.004) for AMA1 (mixed linear model). IFN-γ -producing CD8+ T cell frequencies were significantly higher than baseline (*) post-Ad for CSP (p = 0.007) and post-Ad for AMA1 (0.002) (mixed linear model).

References

    1. WHO (2011) World Malaria Report.
    1. PMI (2011) Fifth Annual Report to Congress.
    1. Clyde DF (1975) Immunization of man against falciparum and vivax malaria by use of attenuated sporozoites. Am J Trop Med Hyg 24: 397–401.
    1. Roestenberg M, McCall M, Hopman J, Wiersma J, Luty AJ, et al. (2009) Protection against a malaria challenge by sporozoite inoculation. N Engl J Med 361: 468–477.
    1. Roestenberg M, Teirlinck AC, McCall MB, Teelen K, Makamdop KN, et al. (2011) Long-term protection against malaria after experimental sporozoite inoculation: an open-label follow-up study. Lancet 377: 1770–1776.
    1. Weiss WR, Jiang CG (2012) Protective CD8+ T lymphocytes in primates immunized with malaria sporozoites. PLoS One 7: e31247.
    1. Overstreet MG, Cockburn IA, Chen YC, Zavala F (2008) Protective CD8 T cells against Plasmodium liver stages: immunobiology of an 'unnatural' immune response. Immunol Rev 225: 272–283.
    1. Doolan DL, Martinez-Alier N (2006) Immune response to pre-erythrocytic stages of malaria parasites. Curr Mol Med 6: 169–185.
    1. Butler NS, Schmidt NW, Vaughan AM, Aly AS, Kappe SH, et al. (2011) Superior antimalarial immunity after vaccination with late liver stage-arresting genetically attenuated parasites. Cell Host Microbe 9: 451–462.
    1. Epstein JE, Tewari K, Lyke KE, Sim BK, Billingsley PF, et al. (2011) Live attenuated malaria vaccine designed to protect through hepatic CD8(+) T cell immunity. Science 334: 475–480.
    1. Kester KE, Cummings JF, Ofori-Anyinam O, Ockenhouse CF, Krzych U, et al. (2009) 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. J Infect Dis 200: 337–346.
    1. Agnandji ST, Lell B, Soulanoudjingar SS, Fernandes JF, Abossolo BP, et al. (2011) First results of phase 3 trial of RTS,S/AS01 malaria vaccine in African children. N Engl J Med 365: 1863–1875.
    1. Moorthy VS, Ballou WR (2009) Immunological mechanisms underlying protection mediated by RTS,S: a review of the available data. Malar J 8: 312.
    1. Halstead SB, Thomas SJ (2011) New Japanese encephalitis vaccines: alternatives to production in mouse brain. Expert Rev Vaccines 10: 355–364.
    1. Sedegah M, Tamminga C, McGrath S, House B, Ganeshan H, et al. (2011) Adenovirus 5-vectored P. falciparum Vaccine Expressing CSP and AMA1. Part A: Safety and Immunogenicity in Seronegative Adults. PLoS One 6: e24586.
    1. Tamminga C, Sedegah M, Regis D, Chuang I, Epstein JE, et al. (2011) Adenovirus-5-vectored P. falciparum Vaccine Expressing CSP and AMA1. Part B: Safety, Immunogenicity and Protective Efficacy of the CSP component. PLoS One 6: e25868.
    1. Webster DP, Dunachie S, Vuola JM, Berthoud T, Keating S, et al. (2005) Enhanced T cell-mediated protection against malaria in human challenges by using the recombinant poxviruses FP9 and modified vaccinia virus Ankara. Proc Natl Acad Sci U S A 102: 4836–4841.
    1. Wang R, Epstein J, Baraceros FM, Gorak EJ, Charoenvit Y, et al. (2001) Induction of CD4(+) T cell-dependent CD8(+) type 1 responses in humans by a malaria DNA vaccine. Proc Natl Acad Sci U S A 98: 10817–10822.
    1. Wang R, Doolan DL, Charoenvit Y, Hedstrom RC, Gardner MJ, et al. (1998) Simultaneous induction of multiple antigen-specific cytotoxic T lymphocytes in nonhuman primates by immunization with a mixture of four Plasmodium falciparum DNA plasmids. Infect Immun 66: 4193–4202.
    1. Reyes-Sandoval A, Berthoud T, Alder N, Siani L, Gilbert SC, et al. (2010) Prime-boost immunization with adenoviral and modified vaccinia virus Ankara vectors enhances the durability and polyfunctionality of protective malaria CD8+ T-cell responses. Infect Immun 78: 145–153.
    1. Jiang G, Shi M, Conteh S, Richie N, Banania G, et al. (2009) Sterile protection against Plasmodium knowlesi in rhesus monkeys from a malaria vaccine: comparison of heterologous prime boost strategies. PLoS One 4: e6559.
    1. Silvie O, Franetich JF, Charrin S, Mueller MS, Siau A, et al. (2004) A role for apical membrane antigen 1 during invasion of hepatocytes by Plasmodium falciparum sporozoites. J Biol Chem 279: 9490–9496.
    1. Stowers AW, Kennedy MC, Keegan BP, Saul A, Long CA, et al. (2002) Vaccination of monkeys with recombinant Plasmodium falciparum apical membrane antigen 1 confers protection against blood-stage malaria. Infect Immun 70: 6961–6967.
    1. Polley SD, Mwangi T, Kocken CH, Thomas AW, Dutta S, et al. (2004) Human antibodies to recombinant protein constructs of Plasmodium falciparum Apical Membrane Antigen 1 (AMA1) and their associations with protection from malaria. Vaccine 23: 718–728.
    1. Coppi A, Natarajan R, Pradel G, Bennett BL, James ER, et al. (2011) The malaria circumsporozoite protein has two functional domains, each with distinct roles as sporozoites journey from mosquito to mammalian host. J Exp Med 208: 341–356.
    1. Bongfen SE, Torgler R, Romero JF, Renia L, Corradin G (2007) Plasmodium berghei-infected primary hepatocytes process and present the circumsporozoite protein to specific CD8+ T cells in vitro. J Immunol 178: 7054–7063.
    1. Dunachie SJ, Walther M, Epstein JE, Keating S, Berthoud T, et al. (2006) 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 74: 5933–5942.
    1. Hermsen CC, de Vlas SJ, van Gemert GJ, Telgt DS, Verhage DF, et al. (2004) Testing vaccines in human experimental malaria: statistical analysis of parasitemia measured by a quantitative real-time polymerase chain reaction. Am J Trop Med Hyg 71: 196–201.
    1. Gaziano TA, Young CR, Fitzmaurice G, Atwood S, Gaziano JM (2008) Laboratory-based versus non-laboratory-based method for assessment of cardiovascular disease risk: the NHANES I Follow-up Study cohort. Lancet 371: 923–931.
    1. Sprangers MC, Lakhai W, Koudstaal W, Verhoeven M, Koel BF, et al. (2003) Quantifying adenovirus-neutralizing antibodies by luciferase transgene detection: addressing preexisting immunity to vaccine and gene therapy vectors. J Clin Microbiol 41: 5046–5052.
    1. Le TP, Coonan KM, Hedstrom RC, Charoenvit Y, Sedegah M, et al. (2000) Safety, tolerability and humoral immune responses after intramuscular administration of a malaria DNA vaccine to healthy adult volunteers. Vaccine 18: 1893–1901.
    1. Epstein JE, Gorak EJ, Charoenvit Y, Wang R, Freydberg N, et al. (2002) Safety, tolerability, and lack of antibody responses after administration of a PfCSP DNA malaria vaccine via needle or needle-free jet injection, and comparison of intramuscular and combination intramuscular/intradermal routes. Hum Gene Ther 13: 1551–1560.
    1. Wang R, Doolan DL, Le TP, Hedstrom RC, Coonan KM, et al. (1998) Induction of antigen-specific cytotoxic T lymphocytes in humans by a malaria DNA vaccine. Science 282: 476–480.
    1. Charoenvit Y, Mellouk S, Cole C, Bechara R, Leef MF, et al. (1991) Monoclonal, but not polyclonal, antibodies protect against Plasmodium yoelii sporozoites. J Immunol 146: 1020–1025.
    1. McGill R (1978) Variations of Box Plots. Am Stat 32: 12–16.
    1. Chulay JD, Schneider I, Cosgriff TM, Hoffman SL, Ballou WR, et al. (1986) Malaria transmitted to humans by mosquitoes infected from cultured Plasmodium falciparum. Am J Trop Med Hyg 35: 66–68.
    1. Sheehy SH, Duncan CJ, Elias SC, Biswas S, Collins KA, et al. (2012) 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 7: e31208.
    1. Tamminga C, Sedegah M, Regis D, Chuang I, Epstein JE, et al. (2011) Adenovirus-5-vectored P. falciparum vaccine expressing CSP and AMA1. Part B: safety, immunogenicity and protective efficacy of the CSP component. PLoS One 6: e25868.
    1. Brun A, Barcena J, Blanco E, Borrego B, Dory D, et al. (2011) Current strategies for subunit and genetic viral veterinary vaccine development. Virus Res 157: 1–12.
    1. Liu MA (2010) Gene-based vaccines: Recent developments. Curr Opin Mol Ther 12: 86–93.
    1. Redding L, Weiner DB (2009) DNA vaccines in veterinary use. Expert Rev Vaccines 8: 1251–1276.
    1. Peiperl L, Morgan C, Moodie Z, Li H, Russell N, et al. (2010) Safety and immunogenicity of a replication-defective adenovirus type 5 HIV vaccine in Ad5-seronegative persons: a randomized clinical trial (HVTN 054). PLoS One 5: e13579.
    1. Radosevic K, Rodriguez A, Lemckert AA, van der Meer M, Gillissen G, et al. The Th1 immune response to Plasmodium falciparum circumsporozoite protein is boosted by adenovirus vectors 35 and 26 with a homologous insert. Clin Vaccine Immunol 17: 1687–1694.
    1. Plotkin SA (2008) Vaccines: correlates of vaccine-induced immunity. Clin Infect Dis 47: 401–409.
    1. Kester KE, Cummings JF, Ockenhouse CF, Nielsen R, Hall BT, et al. (2008) Phase 2a trial of 0, 1, and 3 month and 0, 7, and 28 day immunization schedules of malaria vaccine RTS,S/AS02 in malaria-naive adults at the Walter Reed Army Institute of Research. Vaccine 26: 2191–2202.
    1. Frimpong-Boateng K, van Rooijen N, Geiben-Lynn R (2010) Regulatory T cells suppress natural killer cells during plasmid DNA vaccination in mice, blunting the CD8+ T cell immune response by the cytokine TGFbeta. PLoS One 5: e12281.
    1. Nwanegbo E, Vardas E, Gao W, Whittle H, Sun H, et al. (2004) Prevalence of neutralizing antibodies to adenoviral serotypes 5 and 35 in the adult populations of The Gambia, South Africa, and the United States. Clin Diagn Lab Immunol 11: 351–357.
    1. Thorner AR, Vogels R, Kaspers J, Weverling GJ, Holterman L, et al. (2006) Age dependence of adenovirus-specific neutralizing antibody titers in individuals from sub-Saharan Africa. J Clin Microbiol 44: 3781–3783.
    1. Buchbinder SP, Mehrotra DV, Duerr A, Fitzgerald DW, Mogg R, et al. (2008) Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-of-concept trial. Lancet 372: 1881–1893.
    1. Gilbert SC, Moorthy VS, Andrews L, Pathan AA, McConkey SJ, et al. (2006) Synergistic DNA-MVA prime-boost vaccination regimes for malaria and tuberculosis. Vaccine 24: 4554–4561.
    1. Asmuth DM, Brown EL, DiNubile MJ, Sun X, del Rio C, et al. (2009) Comparative cell-mediated immunogenicity of DNA/DNA, DNA/adenovirus type 5 (Ad5), or Ad5/Ad5 HIV-1 clade B gag vaccine prime-boost regimens. J Infect Dis 201: 132–141.
    1. Jaoko W, Karita E, Kayitenkore K, Omosa-Manyonyi G, Allen S, et al. (2010) Safety and immunogenicity study of Multiclade HIV-1 adenoviral vector vaccine alone or as boost following a multiclade HIV-1 DNA vaccine in Africa. PLoS One 5: e12873.
    1. Koup RA, Roederer M, Lamoreaux L, Fischer J, Novik L, et al. (2010) Priming immunization with DNA augments immunogenicity of recombinant adenoviral vectors for both HIV-1 specific antibody and T-cell responses. PLoS One 5: e9015.

Source: PubMed

3
Sottoscrivi