Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Commun ; 4: 2836, 2013.
Article in English | MEDLINE | ID: mdl-24284865

ABSTRACT

Induction of antigen-specific CD8(+) T cells offers the prospect of immunization against many infectious diseases, but no subunit vaccine has induced CD8(+) T cells that correlate with efficacy in humans. Here we demonstrate that a replication-deficient chimpanzee adenovirus vector followed by a modified vaccinia virus Ankara booster induces exceptionally high frequency T-cell responses (median >2400 SFC/10(6) peripheral blood mononuclear cells) to the liver-stage Plasmodium falciparum malaria antigen ME-TRAP. It induces sterile protective efficacy against heterologous strain sporozoites in three vaccinees (3/14, 21%), and delays time to patency through substantial reduction of liver-stage parasite burden in five more (5/14, 36%), P=0.008 compared with controls. The frequency of monofunctional interferon-γ-producing CD8(+) T cells, but not antibodies, correlates with sterile protection and delay in time to patency (P(corrected)=0.005). Vaccine-induced CD8(+) T cells provide protection against human malaria, suggesting that a major limitation of previous vaccination approaches has been the insufficient magnitude of induced T cells.


Subject(s)
Adenoviruses, Simian/immunology , CD8-Positive T-Lymphocytes/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Vaccinia virus/immunology , Adenoviruses, Simian/genetics , Adolescent , Adult , Animals , Antibodies, Protozoan/immunology , Female , Genetic Vectors/genetics , Genetic Vectors/immunology , Humans , Immunity, Cellular , Immunization , Immunization, Secondary , Interferon-gamma/immunology , Leukocytes, Mononuclear , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Male , Middle Aged , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Vaccinia virus/genetics , Young Adult
2.
PLoS One ; 8(6): e65960, 2013.
Article in English | MEDLINE | ID: mdl-23823332

ABSTRACT

BACKGROUND: Controlled human malaria infection (CHMI) studies have become a routine tool to evaluate efficacy of candidate anti-malarial drugs and vaccines. To date, CHMI trials have mostly been conducted using the bite of infected mosquitoes, restricting the number of trial sites that can perform CHMI studies. Aseptic, cryopreserved P. falciparum sporozoites (PfSPZ Challenge) provide a potentially more accurate, reproducible and practical alternative, allowing a known number of sporozoites to be administered simply by injection. METHODOLOGY: We sought to assess the infectivity of PfSPZ Challenge administered in different dosing regimens to malaria-naive healthy adults (n = 18). Six participants received 2,500 sporozoites intradermally (ID), six received 2,500 sporozoites intramuscularly (IM) and six received 25,000 sporozoites IM. FINDINGS: Five out of six participants receiving 2,500 sporozoites ID, 3/6 participants receiving 2,500 sporozoites IM and 6/6 participants receiving 25,000 sporozoites IM were successfully infected. The median time to diagnosis was 13.2, 17.8 and 12.7 days for 2,500 sporozoites ID, 2,500 sporozoites IM and 25,000 sporozoites IM respectively (Kaplan Meier method; p = 0.024 log rank test). CONCLUSIONS: 2,500 sporozoites ID and 25,000 sporozoites IM have similar infectivities. Given the dose response in infectivity seen with IM administration, further work should evaluate increasing doses of PfSPZ Challenge IM to identify a dosing regimen that reliably infects 100% of participants. TRIAL REGISTRATION: ClinicalTrials.gov NCT01465048.


Subject(s)
Malaria, Falciparum/parasitology , Needles , Syringes , Cryopreservation , Enzyme-Linked Immunosorbent Assay , Humans , Malaria, Falciparum/prevention & control , Pilot Projects
3.
Hum Vaccin Immunother ; 9(9): 1831-40, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23778312

ABSTRACT

In the absence of any highly effective vaccine candidate against Plasmodium falciparum malaria, it remains imperative for the field to pursue all avenues that may lead to the successful development of such a formulation. The development of a subunit vaccine targeting the asexual blood-stage of Plasmodium falciparum malaria infection has proven particularly challenging with only limited success to date in clinical trials. However, only a fraction of potential blood-stage vaccine antigens have been evaluated as targets, and a number of new promising candidate antigen formulations and delivery platforms are approaching clinical development. It is therefore essential that reliable and sensitive methods of detecting, or ruling out, even modest efficacy of blood-stage vaccines in small clinical trials be established. In this article we evaluate the challenges facing blood-stage vaccine developers, assess the appropriateness and limitations of various in vivo approaches for efficacy assessment and suggest future directions for the field.


Subject(s)
Biomedical Research/methods , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Humans , Malaria Vaccines/administration & dosage
4.
J Infect Dis ; 208(2): 340-5, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23570846

ABSTRACT

Controlled human malaria infection is used to measure efficacy of candidate malaria vaccines before field studies are undertaken. Mathematical modeling using data from quantitative polymerase chain reaction (qPCR) parasitemia monitoring can discriminate between vaccine effects on the parasite's liver and blood stages. Uncertainty regarding the most appropriate modeling method hinders interpretation of such trials. We used qPCR data from 267 Plasmodium falciparum infections to compare linear, sine-wave, and normal-cumulative-density-function models. We find that the parameters estimated by these models are closely correlated, and their predictive accuracy for omitted data points was similar. We propose that future studies include the linear model.


Subject(s)
Liver/parasitology , Malaria Vaccines/pharmacology , Malaria, Falciparum/parasitology , Models, Biological , Parasitemia/parasitology , Plasmodium falciparum/drug effects , Animals , Humans , Liver/drug effects , Liver/immunology , Malaria Vaccines/blood , Malaria Vaccines/immunology , Malaria, Falciparum/genetics , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Parasitemia/genetics , Parasitemia/immunology , Parasitemia/prevention & control , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology
5.
PLoS One ; 8(3): e57726, 2013.
Article in English | MEDLINE | ID: mdl-23526949

ABSTRACT

BACKGROUND: Heterologous prime boost immunization with chimpanzee adenovirus 63 (ChAd63) and Modified vaccinia Virus Ankara (MVA) vectored vaccines is a strategy recently shown to be capable of inducing strong cell mediated responses against several antigens from the malaria parasite. ChAd63-MVA expressing the Plasmodium falciparum pre-erythrocytic antigen ME-TRAP (multiple epitope string with thrombospondin-related adhesion protein) is a leading malaria vaccine candidate, capable of inducing sterile protection in malaria naïve adults following controlled human malaria infection (CHMI). METHODOLOGY: We conducted two Phase Ib dose escalation clinical trials assessing the safety and immunogenicity of ChAd63-MVA ME-TRAP in 46 healthy malaria exposed adults in two African countries with similar malaria transmission patterns. RESULTS: ChAd63-MVA ME-TRAP was shown to be safe and immunogenic, inducing high-level T cell responses (median >1300 SFU/million PBMC). CONCLUSIONS: ChAd63-MVA ME-TRAP is a safe and highly immunogenic vaccine regimen in adults with prior exposure to malaria. Further clinical trials to assess safety and immunogenicity in children and infants and protective efficacy in the field are now warranted. TRIAL REGISTRATION: Pactr.org PACTR2010020001771828 Pactr.org PACTR201008000221638 ClinicalTrials.gov NCT01373879 NCT01373879 ClinicalTrials.gov NCT01379430 NCT01379430.


Subject(s)
Malaria Vaccines/administration & dosage , Plasmodium falciparum/immunology , Adenoviruses, Simian/genetics , Adult , Antigens, Protozoan/genetics , Gambia , Genetic Vectors , Humans , Immunization, Secondary , Interferon-gamma/blood , Kenya , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Male , Middle Aged , Plasmodium falciparum/genetics , T-Lymphocytes/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccinia virus/genetics , Young Adult
6.
J Immunol ; 190(3): 1135-47, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23293353

ABSTRACT

Overcoming antigenic variation is one of the major challenges in the development of an effective vaccine against Plasmodium falciparum, a causative agent of human malaria. Inclusion of multiple Ag variants in subunit vaccine candidates is one strategy that has aimed to overcome this problem for the leading blood-stage malaria vaccine targets, that is, merozoite surface protein 1 (MSP1) and apical membrane Ag 1 (AMA1). However, previous studies, utilizing malaria Ags, have concluded that inclusion of multiple allelic variants, encoding altered peptide ligands, in such a vaccine may be detrimental to both the priming and in vivo restimulation of Ag-experienced T cells. In this study, we analyze the T cell responses to two alleles of MSP1 and AMA1 induced by vaccination of malaria-naive adult volunteers with bivalent viral-vectored vaccine candidates. We show a significant bias to the 3D7/MAD20 allele compared with the Wellcome allele for the 33 kDa region of MSP1, but not for the 19 kDa fragment or the AMA1 Ag. Although this bias could be caused by "immune interference" at priming, the data do not support a significant role for "immune antagonism" during memory T cell restimulation, despite observation of the latter at a minimal epitope level in vitro. A lack of class I HLA epitopes in the Wellcome allele that are recognized by vaccinated volunteers may in fact contribute to the observed bias. We also show that controlled infection with 3D7 strain P. falciparum parasites neither boosts existing 3D7-specific T cell responses nor appears to "immune divert" cellular responses toward the Wellcome allele.


Subject(s)
Antigens, Protozoan/immunology , Immunologic Memory/immunology , Malaria Vaccines/immunology , Membrane Proteins/immunology , Merozoite Surface Protein 1/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , T-Lymphocytes/immunology , Adenoviridae/genetics , Adult , Alleles , Antibodies, Protozoan/immunology , Antigenic Variation/genetics , Antigens, Protozoan/genetics , Defective Viruses/genetics , Epitopes/immunology , Erythrocytes/parasitology , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , HLA Antigens/immunology , Humans , Interferon-gamma/biosynthesis , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Membrane Proteins/genetics , Merozoite Surface Protein 1/genetics , Peptide Fragments/genetics , Peptide Fragments/immunology , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Protozoan Proteins/genetics , Vaccination , Vaccines, Subunit/immunology , Vaccinia virus/genetics
7.
Mol Ther ; 20(12): 2355-68, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23089736

ABSTRACT

The induction of cellular immunity, in conjunction with antibodies, may be essential for vaccines to protect against blood-stage infection with the human malaria parasite Plasmodium falciparum. We have shown that prime-boost delivery of P. falciparum blood-stage antigens by chimpanzee adenovirus 63 (ChAd63) followed by the attenuated orthopoxvirus MVA is safe and immunogenic in healthy adults. Here, we report on vaccine efficacy against controlled human malaria infection delivered by mosquito bites. The blood-stage malaria vaccines were administered alone, or together (MSP1+AMA1), or with a pre-erythrocytic malaria vaccine candidate (MSP1+ME-TRAP). In this first human use of coadministered ChAd63-MVA regimes, we demonstrate immune interference whereby responses against merozoite surface protein 1 (MSP1) are dominant over apical membrane antigen 1 (AMA1) and ME-TRAP. We also show that induction of strong cellular immunity against MSP1 and AMA1 is safe, but does not impact on parasite growth rates in the blood. In a subset of vaccinated volunteers, a delay in time to diagnosis was observed and sterilizing protection was observed in one volunteer coimmunized with MSP1+AMA1-results consistent with vaccine-induced pre-erythrocytic, rather than blood-stage, immunity. These data call into question the utility of T cell-inducing blood-stage malaria vaccines and suggest that the focus should remain on high-titer antibody induction against susceptible antigen targets.


Subject(s)
Antigens, Protozoan/immunology , Culicidae/parasitology , Culicidae/pathogenicity , Malaria Vaccines/therapeutic use , Merozoite Surface Protein 1/immunology , Adenoviruses, Simian/genetics , Animals , Flow Cytometry , Humans , Malaria Vaccines/administration & dosage , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Orthopoxvirus/immunology , Pan troglodytes/virology
8.
Clin Transl Sci ; 5(4): 348-50, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22883613

ABSTRACT

Previously unrecognized medical conditions identified in volunteers for early phase clinical studies have significant clinical and ethical implications for the participant. It is therefore crucial that the potential for unexpected diagnosis is addressed during the informed consent process. But the frequency of incidental diagnosis in healthy volunteers who attend for clinical trial screening remains unclear. To assess this we retrospectively analyzed 1,131 independent screening visits for 990 volunteers at a single academic center over a 10-year period to describe the frequency and nature of new clinical findings. Overall 23 of 990 volunteers (2.3%) were excluded at screening for a newly diagnosed medical abnormality. Some clinically important conditions, such as nephrotic syndrome and familial hypercholesterolemia were identified. The frequency of abnormalities was associated with increasing age in males (p= 0.02 χ(2) for trend) but not females (p= 0.82). These data will assist those planning and conducting phase I/II vaccine trials in healthy volunteers, and importantly should strengthen the informed consent of future trial participants.


Subject(s)
Clinical Trials as Topic , Health , Incidental Findings , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult
9.
Travel Med Infect Dis ; 10(4): 192-6, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22531678

ABSTRACT

During the H1N1 influenza pandemic (pH1N1/09) diagnostic algorithms were developed to guide antiviral provision. However febrile illnesses are notoriously difficult to distinguish clinically. Recent evidence highlights the importance of incorporating travel history into diagnostic algorithms to prevent the catastrophic misdiagnosis of life-threatening infections such as malaria. We applied retrospectively the UK pH1N1/09 case definition to a unique cohort of healthy adult volunteers exposed to Plasmodium falciparum malaria or influenza to assess the predictive value of this case definition, and to explore the distinguishing clinical features of early phase infection with these pathogens under experimental conditions. For influenza exposure the positive predictive value of the pH1N1/09 case definition was only 0.38 (95% CI: 0.06-0.60), with a negative predictive value of 0.27 (95% CI: 0.02-0.51). Interestingly, 8/11 symptomatic malaria-infected adults would have been inappropriately classified with influenza by the pH1N1/09 case definition, while 5/8 symptomatic influenza-exposed volunteers would have been classified without influenza (P = 0.18 Fisher's exact). Cough (P = 0.005) and nasal symptoms (P = 0.001) were the only clinical features that distinguished influenza-exposed from malaria-exposed volunteers. An open mind regarding the clinical cause of undifferentiated febrile illness, particularly in the absence of upper respiratory tract symptoms, remains important even during influenza pandemic settings. These data support incorporating travel history into pandemic algorithms.


Subject(s)
Influenza A Virus, H3N2 Subtype , Influenza, Human/diagnosis , Malaria/diagnosis , Adult , Diagnosis, Differential , Diagnostic Errors , Female , Humans , Male , Predictive Value of Tests , Young Adult
10.
Clin Infect Dis ; 55(1): 19-25, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22441650

ABSTRACT

BACKGROUND: The novel influenza vaccine MVA-NP+M1 is designed to boost cross-reactive T-cell responses to internal antigens of the influenza A virus that are conserved across all subtypes, providing protection against both influenza disease and virus shedding against all influenza A viruses. Following a phase 1 clinical study that demonstrated vaccine safety and immunogenicity, a phase 2a vaccination and influenza challenge study has been conducted in healthy adult volunteers. METHODS: Volunteers with no measurable serum antibodies to influenza A/Wisconsin/67/2005 received either a single vaccination with MVA-NP+M1 or no vaccination. T-cell responses to the vaccine antigens were measured at enrollment and again prior to virus challenge. All volunteers underwent intranasal administration of influenza A/Wisconsin/67/2005 while in a quarantine unit and were monitored for symptoms of influenza disease and virus shedding. RESULTS: Volunteers had a significantly increased T-cell response to the vaccine antigens following a single dose of the vaccine, with an increase in cytolytic effector molecules. Intranasal influenza challenge was undertaken without safety issues. Two of 11 vaccinees and 5 of 11 control subjects developed laboratory-confirmed influenza (symptoms plus virus shedding). Symptoms of influenza were less pronounced in the vaccinees and there was a significant reduction in the number of days of virus shedding in those vaccinees who developed influenza (mean, 1.09 days in controls, 0.45 days in vaccinees, P = .036). CONCLUSIONS: This study provides the first demonstration of clinical efficacy of a T-cell-based influenza vaccine and indicates that further clinical development should be undertaken. CLINICAL TRIALS REGISTRATION: NCT00993083.


Subject(s)
Antibodies, Viral/immunology , Influenza A virus/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , T-Lymphocytes/immunology , Administration, Intranasal , Adolescent , Adult , Antibodies, Viral/blood , Antigens, Viral/immunology , HLA-A2 Antigen , Humans , Influenza Vaccines/administration & dosage , Influenza Vaccines/adverse effects , Influenza, Human/virology , Interferon-gamma , Middle Aged , Nucleocapsid Proteins , Pilot Projects , RNA-Binding Proteins/immunology , Viral Core Proteins/immunology , Viral Matrix Proteins/immunology , Virus Shedding
11.
PLoS One ; 7(2): e31208, 2012.
Article in English | MEDLINE | ID: mdl-22363582

ABSTRACT

BACKGROUND: Traditionally, vaccine development against the blood-stage of Plasmodium falciparum infection has focused on recombinant protein-adjuvant formulations in order to induce high-titer growth-inhibitory antibody responses. However, to date no such vaccine encoding a blood-stage antigen(s) alone has induced significant protective efficacy against erythrocytic-stage infection in a pre-specified primary endpoint of a Phase IIa/b clinical trial designed to assess vaccine efficacy. Cell-mediated responses, acting in conjunction with functional antibodies, may be necessary for immunity against blood-stage P. falciparum. The development of a vaccine that could induce both cell-mediated and humoral immune responses would enable important proof-of-concept efficacy studies to be undertaken to address this question. METHODOLOGY: We conducted a Phase Ia, non-randomized clinical trial in 16 healthy, malaria-naïve adults of the chimpanzee adenovirus 63 (ChAd63) and modified vaccinia virus Ankara (MVA) replication-deficient viral vectored vaccines encoding two alleles (3D7 and FVO) of the P. falciparum blood-stage malaria antigen; apical membrane antigen 1 (AMA1). ChAd63-MVA AMA1 administered in a heterologous prime-boost regime was shown to be safe and immunogenic, inducing high-level T cell responses to both alleles 3D7 (median 2036 SFU/million PBMC) and FVO (median 1539 SFU/million PBMC), with a mixed CD4(+)/CD8(+) phenotype, as well as substantial AMA1-specific serum IgG responses (medians of 49 µg/mL and 41 µg/mL for 3D7 and FVO AMA1 respectively) that demonstrated growth inhibitory activity in vitro. CONCLUSIONS: ChAd63-MVA is a safe and highly immunogenic delivery platform for both alleles of the AMA1 antigen in humans which warrants further efficacy testing. ChAd63-MVA is a promising heterologous prime-boost vaccine strategy that could be applied to numerous other diseases where strong cellular and humoral immune responses are required for protection. TRIAL REGISTRATION: ClinicalTrials.gov NCT01095055.


Subject(s)
Adenoviruses, Simian/genetics , Antigens, Protozoan/immunology , Genetic Vectors/genetics , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Vaccinia virus/genetics , Adolescent , Adult , Animals , Antibodies, Neutralizing/immunology , Antibodies, Protozoan/immunology , Enzyme-Linked Immunospot Assay , Female , Humans , Immunization , Interferon-gamma/immunology , Life Cycle Stages , Malaria, Falciparum/immunology , Male , Middle Aged , Plasmodium falciparum/growth & development , T-Lymphocytes/immunology , Young Adult
12.
J Infect Dis ; 205(5): 772-81, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22275401

ABSTRACT

BACKGROUND: Vaccine development in human Plasmodium falciparum malaria has been hampered by the exceptionally high levels of CD8(+) T cells required for efficacy. Use of potently immunogenic human adenoviruses as vaccine vectors could overcome this problem, but these are limited by preexisting immunity to human adenoviruses. METHODS: From 2007 to 2010, we undertook a phase I dose and route finding study of a new malaria vaccine, a replication-incompetent chimpanzee adenovirus 63 (ChAd63) encoding the preerythrocytic insert multiple epitope thrombospondin-related adhesion protein (ME-TRAP; n = 54 vaccinees) administered alone (n = 28) or with a modified vaccinia virus Ankara (MVA) ME-TRAP booster immunization 8 weeks later (n = 26). We observed an excellent safety profile. High levels of TRAP antigen-specific CD8(+) and CD4(+) T cells, as detected by interferon γ enzyme-linked immunospot assay and flow cytometry, were induced by intramuscular ChAd63 ME-TRAP immunization at doses of 5 × 10(10) viral particles and above. Subsequent administration of MVA ME-TRAP boosted responses to exceptionally high levels, and responses were maintained for up to 30 months postvaccination. CONCLUSIONS: The ChAd63 chimpanzee adenovirus vector appears safe and highly immunogenic, providing a viable alternative to human adenoviruses as vaccine vectors for human use. CLINICAL TRIALS REGISTRATION: NCT00890019.


Subject(s)
Adenoviruses, Simian/immunology , Malaria Vaccines/administration & dosage , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Protozoan Proteins/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Adenoviruses, Simian/genetics , Animals , Antibodies, Neutralizing/blood , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Epitopes , Flow Cytometry , Humans , Interferon-gamma/metabolism , Interleukin-2/metabolism , Malaria Vaccines/adverse effects , Tumor Necrosis Factor-alpha/metabolism , Vaccines, DNA/adverse effects
13.
Mol Ther ; 19(12): 2269-76, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21862998

ABSTRACT

Efficacy trials of antibody-inducing protein-in-adjuvant vaccines targeting the blood-stage Plasmodium falciparum malaria parasite have so far shown disappointing results. The induction of cell-mediated responses in conjunction with antibody responses is thought to be one alternative strategy that could achieve protective efficacy in humans. Here, we prepared chimpanzee adenovirus 63 (ChAd63) and modified vaccinia virus Ankara (MVA) replication-deficient vectors encoding the well-studied P. falciparum blood-stage malaria antigen merozoite surface protein 1 (MSP1). A phase Ia clinical trial was conducted in healthy adults of a ChAd63-MVA MSP1 heterologous prime-boost immunization regime. The vaccine was safe and generally well tolerated. Fewer systemic adverse events (AEs) were observed following ChAd63 MSP1 than MVA MSP1 administration. Exceptionally strong T-cell responses were induced, and these displayed a mixed of CD4(+) and CD8(+) phenotype. Substantial MSP1-specific serum immunoglobulin G (IgG) antibody responses were also induced, which were capable of recognizing native parasite antigen, but these did not reach titers sufficient to neutralize P. falciparum parasites in vitro. This viral vectored vaccine regime is thus a leading approach for the induction of strong cellular and humoral immunogenicity against difficult disease targets in humans. Further studies are required to assess whether this strategy can achieve protective efficacy against blood-stage malaria infection.


Subject(s)
Adenoviridae/genetics , CD4-Positive T-Lymphocytes/immunology , Genetic Vectors/therapeutic use , Malaria, Falciparum/immunology , Malaria, Falciparum/therapy , Merozoite Surface Protein 1/immunology , Vaccinia virus/genetics , Adjuvants, Immunologic , Adult , Animals , Antibodies, Protozoan/immunology , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunity, Cellular , Immunoglobulin G/immunology , Immunologic Memory , Macaca mulatta , Malaria, Falciparum/blood , Male , Merozoite Surface Protein 1/blood , Merozoite Surface Protein 1/genetics , Mice , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Vaccination , Young Adult
14.
PLoS One ; 6(7): e22271, 2011.
Article in English | MEDLINE | ID: mdl-21799809

ABSTRACT

BACKGROUND: Inhibition of parasite growth is a major objective of blood-stage malaria vaccines. The in vitro assay of parasite growth inhibitory activity (GIA) is widely used as a surrogate marker for malaria vaccine efficacy in the down-selection of candidate blood-stage vaccines. Here we report the first study to examine the relationship between in vivo Plasmodium falciparum growth rates and in vitro GIA in humans experimentally infected with blood-stage malaria. METHODS: In this phase I/IIa open-label clinical trial five healthy malaria-naive volunteers were immunised with AMA1/C1-Alhydrogel+CPG 7909, and together with three unvaccinated controls were challenged by intravenous inoculation of P. falciparum infected erythrocytes. RESULTS: A significant correlation was observed between parasite multiplication rate in 48 hours (PMR) and both vaccine-induced growth-inhibitory activity (Pearson r = -0.93 [95% CI: -1.0, -0.27] P = 0.02) and AMA1 antibody titres in the vaccine group (Pearson r = -0.93 [95% CI: -0.99, -0.25] P = 0.02). However immunisation failed to reduce overall mean PMR in the vaccine group in comparison to the controls (vaccinee 16 fold [95% CI: 12, 22], control 17 fold [CI: 0, 65] P = 0.70). Therefore no impact on pre-patent period was observed (vaccine group median 8.5 days [range 7.5-9], control group median 9 days [range 7-9]). CONCLUSIONS: Despite the first observation in human experimental malaria infection of a significant association between vaccine-induced in vitro growth inhibitory activity and in vivo parasite multiplication rate, this did not translate into any observable clinically relevant vaccine effect in this small group of volunteers. TRIAL REGISTRATION: ClinicalTrials.gov [NCT00984763].


Subject(s)
Adjuvants, Immunologic , Malaria Vaccines/immunology , Malaria/prevention & control , Malaria/parasitology , Plasmodium falciparum/growth & development , Plasmodium falciparum/immunology , Vaccination/methods , Adjuvants, Immunologic/adverse effects , Adolescent , Adult , Aluminum Hydroxide/immunology , Antibodies/immunology , Antigens, Protozoan/immunology , Female , Humans , Malaria Vaccines/adverse effects , Male , Membrane Proteins/immunology , Middle Aged , Oligodeoxyribonucleotides/immunology , Protozoan Proteins/immunology , Vaccination/adverse effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...