Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Trends Parasitol ; 23(7): 293-6, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17512252

RESUMO

To date, the only pre-blood stage vaccine to confer protection against malaria in field trials elicits both antigen-specific antibody and T-cell responses. Recent clinical trials of new heterologous prime-boost malaria vaccine regimens using DNA, fowlpox or MVA, have chiefly elicited T-cell responses that have promisingly reduced hepatic merozoites in challenge trials, but failed to protect in field trials. These encouraging results suggest further augmentation of T-cell responses to pre-blood stage antigens might one day contribute to a highly protective vaccine. We envision that a highly protective pre-erythrocytic vaccine will likely be based upon a heterologous prime-boost regimen that induces both appropriate T-cell responses as well as robust and protracted antibody production.


Assuntos
Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Vacinas de DNA/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Ensaios Clínicos Fase II como Assunto , Vírus da Varíola das Aves Domésticas/imunologia , Humanos , Memória Imunológica/imunologia , Vacinas Antimaláricas/uso terapêutico , Malária Falciparum/prevenção & controle , Vacinas de DNA/uso terapêutico
2.
Clin Vaccine Immunol ; 20(6): 803-10, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23536694

RESUMO

Circumsporozoite protein (CSP) of Plasmodium falciparum is a protective human malaria vaccine candidate. There is an urgent need for models that can rapidly down-select novel CSP-based vaccine candidates. In the present study, the mouse-mosquito transmission cycle of a transgenic Plasmodium berghei malaria parasite stably expressing a functional full-length P. falciparum CSP was optimized to consistently produce infective sporozoites for protection studies. A minimal sporozoite challenge dose was established, and protection was defined as the absence of blood-stage parasites 14 days after intravenous challenge. The specificity of protection was confirmed by vaccinating mice with multiple CSP constructs of differing lengths and compositions. Constructs that induced high NANP repeat-specific antibody titers in enzyme-linked immunosorbent assays were protective, and the degree of protection was dependent on the antigen dose. There was a positive correlation between antibody avidity and protection. The antibodies in the protected mice recognized the native CSP on the parasites and showed sporozoite invasion inhibitory activity. Passive transfer of anti-CSP antibodies into naive mice also induced protection. Thus, we have demonstrated the utility of a mouse efficacy model to down-select human CSP-based vaccine formulations.


Assuntos
Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Parasitemia/prevenção & controle , Proteínas de Protozoários/imunologia , Vacinação/métodos , Animais , Anticorpos Antiprotozoários/sangue , Culicidae , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/isolamento & purificação , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium berghei/genética , Plasmodium berghei/imunologia , Plasmodium falciparum/genética , Plasmodium falciparum/imunologia , Proteínas de Protozoários/genética , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/isolamento & purificação
3.
Trends Parasitol ; 27(7): 306-14, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21435951

RESUMO

The development of a vaccine against malaria is a major research priority given the burden of disease, death and economic loss inflicted upon the tropical world by this parasite. Despite decades of effort, however, a vaccine remains elusive. The best candidate is a subunit vaccine termed RTS,S but this provides only partial protection against clinical disease. This review examines what is known about protective immunity against pre-erythrocytic stage malaria by considering the humoral and T cell-mediated immune responses that are induced by attenuated sporozoites and by the RTS,S vaccine. On the basis of these observations a set of research priorities are defined that are crucial for the development of a vaccine capable of inducing long-lasting and high-grade protection against malaria.


Assuntos
Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Linfócitos T CD4-Positivos , Interações Hospedeiro-Parasita/imunologia , Humanos , Imunidade Humoral , Malária/epidemiologia , Malária/imunologia , Esporozoítos/imunologia , Esporozoítos/fisiologia
4.
PLoS One ; 6(7): e20775, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21779319

RESUMO

A phase 2a RTS,S/AS malaria vaccine trial, conducted previously at the Walter Reed Army Institute of Research, conferred sterile immunity against a primary challenge with infectious sporozoites in 40% of the 80 subjects enrolled in the study. The frequency of Plasmodium falciparum circumsporozoite protein (CSP)-specific CD4(+) T cells was significantly higher in protected subjects as compared to non-protected subjects. Intrigued by these unique vaccine-related correlates of protection, in the present study we asked whether RTS,S also induced effector/effector memory (T(E/EM)) and/or central memory (T(CM)) CD4(+) T cells and whether one or both of these sub-populations is the primary source of cytokine production. We showed for the first time that PBMC from malaria-non-exposed RTS,S-immunized subjects contain both T(E/EM) and T(CM) cells that generate strong IL-2 responses following re-stimulation in vitro with CSP peptides. Moreover, both the frequencies and the total numbers of IL-2-producing CD4(+) T(E/EM) cells and of CD4(+) T(CM) cells from protected subjects were significantly higher than those from non-protected subjects. We also demonstrated for the first time that there is a strong association between the frequency of CSP peptide-reactive CD4(+) T cells producing IL-2 and the titers of CSP-specific antibodies in the same individual, suggesting that IL-2 may be acting as a growth factor for follicular Th cells and/or B cells. The frequencies of CSP peptide-reactive, TNF-α-producing CD4(+) T(E/EM) cells and of CD4(+) T(E/EM) cells secreting both IL-2 and TNF-α were also shown to be higher in protected vs. non-protected individuals. We have, therefore, demonstrated that in addition to TNF-α, IL-2 is also a significant contributing factor to RTS,S/AS vaccine induced immunity and that both T(E/EM) and T(CM) cells are major producers of IL-2.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Interleucina-2/metabolismo , Vacinas Antimaláricas/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Vacinas Antimaláricas/uso terapêutico , Masculino
5.
Vaccine ; 28(31): 5135-44, 2010 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-19737527

RESUMO

Plasmodium falciparum Liver Stage Antigen 1 (LSA-1) is a pre-erythrocytic stage antigen. Our LSA-1 vaccine candidate is a recombinant protein with full-length C- and N-terminal flanking domains and two of the 17 amino acid repeats from the central repeat region termed "LSA-NRC." We describe the first Phase I/II study of this recombinant LSA-NRC protein formulated with either the AS01 or AS02 adjuvant system. We conducted an open-label Phase I/II study. Thirty-six healthy malaria-naïve adults received one of four formulations by intra-deltoid injection on a 0 and 1 month schedule; low dose (LD) LSA-NRC/AS01:10microg LSA-NRC/0.5ml AS01 (n=5), high dose (HD) LSA-NRC/AS01: 50microg LSA-NRC/0.5ml AS01 (n=13); LD LSA-NRC/AS02: 10microg LSA-NRC/0.5ml AS02 (n=5) and HD LSA-NRC/AS02: 50microg LSA-NRC/0.5ml AS02 (n=13). Two weeks post-second immunization, the high dose vaccinees and 6 non-immunized infectivity controls underwent experimental malaria sporozoite challenge. The vaccines showed a reassuring safety profile but were moderately reactogenic. There were no serious adverse events. All subjects seroconverted after the first immunization. Following the second immunization, LSA-1-specific CD4+ T cells producing two cytokines (IL-2 and IFN-gamma) were found by intra-cellular staining in all subjects in the LD LSA-NRC/AS01B group and in 3 of 5 subjects in the LD LSA-NRC/AS02 group. In contrast, the HD LSA-NRC/AS01 and HD LSA-NRC/AS02 group subjects had fewer LSA-1-specific CD4+ T cells, and minimal to no IFN-gamma responses. There was no increase in LSA-1-specific CD8+ T cells found in any group. Per protocol, 22 high dose vaccinees, but no low dose vaccinees, underwent P. falciparum homologous malaria challenge (3D7 clone). All vaccinees became parasitemic and there was no delay in their pre-patent period versus controls (p=0.95). LSA-NRC/AS01 and LSA-NRC/AS02 elicited antigen-specific antibody and CD4+ T cell responses, but elicited no protective immunity. Although the optimal antigen dose of LSA-NRC may not have been selected for the challenge portion of the protocol, further vaccine development based upon LSA-1 should not be excluded and should include alternative vaccine platforms able to elicit additional effector mechanisms such as CD8+ T cells.


Assuntos
Antígenos de Protozoários/imunologia , Linfócitos T CD4-Positivos/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Adjuvantes Imunológicos/farmacologia , Adulto , Anticorpos Antiprotozoários/sangue , Formação de Anticorpos , Feminino , Humanos , Imunidade Celular , Imunidade Humoral , Esquemas de Imunização , Imunização Secundária , Interferon gama/imunologia , Interleucina-2/imunologia , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/efeitos adversos , Malária Falciparum/imunologia , Masculino , Parasitemia/imunologia , Plasmodium falciparum/imunologia , Proteínas Recombinantes/imunologia , Esporozoítos/imunologia , Adulto Jovem
6.
Vaccine ; 26(18): 2191-202, 2008 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-18387719

RESUMO

BACKGROUND: Immunization with RTS,S/AS02 consistently protects some vaccinees against malaria infection in experimental challenges and in field trials. A brief immunization schedule against falciparum malaria would be compatible with the Expanded Programme on Immunization, or in combination with other prevention measures, interrupt epidemic malaria or protect individuals upon sudden travel to an endemic area. METHODS: We conducted an open label, Phase 2a trial of two different full dose schedules of RTS,S/AS02 in 40 healthy malaria-naïve adults. Cohort 1 (n=20) was immunized on a 0, 1, and 3 month schedule and Cohort 2 (n=20) on a 0, 7, and 28 day schedule. Three weeks later, 38 vaccinees and 12 unimmunized infectivity controls underwent malaria challenge. RESULTS: Both regimens had a good safety and tolerability profile. Peak GMCs of antibody to the circumsporozoite protein (CSP) were similar in Cohort 1 (78 microg/mL; 95% CI: 45-134) and Cohort 2 (65 microg/mL; 95% CI: 40-104). Vaccine efficacy for Cohort 1 was 45% (95% CI: 18-62%) and for Cohort 2, 39% (95% CI: 11-56%). Protected volunteers had a higher GMC of anti-CSP antibody (114 microg/mL) than did volunteers with a 2-day delay (70 microg/mL) or no delay (30 microg/mL) in the time to onset of parasitemia (Kruskal-Wallis, p=0.019). A trend was seen for higher CSP-specific IFN-gamma responses in PBMC from protected volunteers only in Cohort 1, but not in Cohort 2, for ex vivo and for cultured ELISPOT assays. CONCLUSION: In malaria-naïve adults, the efficacy of three-dose RTS,S/AS02 regimens on either a 0, 1, and 3 month schedule or an abbreviated 0, 7, and 28 day schedule was not discernibly different from two previously reported trials of two-dose regimens given at 0, 1 month that conferred 47% (95% CI: -19 to 76%) protection and in another trial 42% (95% CI: 5-63%). A strong association of CSP-specific antibody with protection against malaria challenge is observed and confirms similar observations made in other studies. Subsequent trials of adjuvanted RTS,S in African children and infants on a 0, 1, and 2 month schedule have demonstrated a favorable safety and efficacy profile.


Assuntos
Esquemas de Imunização , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Adolescente , Adulto , Anticorpos Antiprotozoários/sangue , Células Cultivadas , Feminino , Humanos , Interferon gama/biossíntese , Leucócitos Mononucleares/imunologia , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/efeitos adversos , Masculino , Pessoa de Meia-Idade , Parasitemia/prevenção & controle , Proteínas de Protozoários/imunologia
7.
J Infect Dis ; 196(4): 599-607, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17624847

RESUMO

At present, radiation-attenuated plasmodia sporozoites ( gamma -spz) is the only vaccine that induces sterile and lasting protection in malaria-naive humans and laboratory rodents. However, gamma -spz are not without risks. For example, the heterogeneity of the gamma -spz could explain occasional breakthrough infections. To avoid this possibility, we constructed a double-knockout P. berghei parasite by removing 2 genes, UIS3 and UIS4, that are up-regulated in infective spz. We evaluated the double-knockout Pbuis3(-)/4(-) parasites for protective efficacy and the contribution of CD8(+) T cells to protection. Pbuis3(-)/4(-) spz induced sterile and protracted protection in C57BL/6 mice. Protection was linked to CD8(+) T cells, given that mice deficient in beta (2)m were not protected. Pbuis3(-)/4(-) spz-immune CD8(+) T cells consisted of effector/memory phenotypes and produced interferon- gamma . On the basis of these observations, we propose that the development of genetically attenuated P. falciparum parasites is warranted for tests in clinical trials as a pre-erythrocytic stage vaccine candidate.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/genética , Malária/imunologia , Malária/prevenção & controle , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/genética , Esporozoítos/imunologia , Vacinação , Animais , Linfócitos T CD8-Positivos/metabolismo , Avaliação Pré-Clínica de Medicamentos , Feminino , Deleção de Genes , Antígenos de Histocompatibilidade Classe I/genética , Esquemas de Imunização , Memória Imunológica , Interferon gama/biossíntese , Fígado/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Microglobulina beta-2/genética
8.
J Immunol ; 171(4): 2024-34, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12902507

RESUMO

We previously demonstrated that protection induced by radiation-attenuated (gamma) Plasmodium berghei sporozoites is linked to MHC class I-restricted CD8(+) T cells specific for exoerythrocytic-stage Ags, and that activated intrahepatic memory CD8(+) T cells are associated with protracted protection. In this study, we further investigated intrahepatic memory CD8(+) T cells to elucidate mechanisms required for their maintenance. Using phenotypic markers indicative of activation (CD44, CD45RB), migration (CD62L), and IFN-gamma production, we identified two subsets of intrahepatic memory CD8(+) T cells: the CD44(high)CD45RB(low)CD62L(low)CD122(low) phenotype, representing the dominant effector memory set, and the CD44(high)CD45RB(high)CD62L(low/high)CD122(high) phenotype, representing the central memory set. Only the effector memory CD8(+) T cells responded swiftly to sporozoite challenge by producing sustained IFN-gamma; the central memory T cells responded with delay, and the IFN-gamma reactivity was short-lived. In addition, the subsets of liver memory CD8(+) T cells segregated according to the expression of CD122 (IL-15R) in that only the central memory CD8(+) T cells were CD122(high), whereas the effector memory CD8(+) T cells were CD122(low). Moreover, the effector memory CD8(+) T cells declined as protection waned in mice treated with primaquine, a drug that interferes with the formation of liver-stage Ags. We propose that protracted protection induced by P. berghei radiation-attenuated sporozoites depends in part on a network of interactive liver memory CD8(+) T cell subsets, each representing a different phase of activation or differentiation, and the balance of which is profoundly affected by the repository of liver-stage Ag and IL-15.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Memória Imunológica , Fígado/imunologia , Malária/imunologia , Malária/prevenção & controle , Plasmodium berghei/imunologia , Animais , Antígenos de Protozoários/metabolismo , Antígenos de Protozoários/fisiologia , Linfócitos T CD8-Positivos/parasitologia , Linfócitos T CD8-Positivos/transplante , Sobrevivência Celular/imunologia , Feminino , Raios gama , Receptores de Hialuronatos/biossíntese , Imunização , Imunofenotipagem , Interferon gama/metabolismo , Selectina L/biossíntese , Antígenos Comuns de Leucócito/biossíntese , Fígado/parasitologia , Fígado/patologia , Malária/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasmodium berghei/crescimento & desenvolvimento , Receptores de Interleucina-2/biossíntese , Esporozoítos/imunologia , Esporozoítos/efeitos da radiação , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/parasitologia , Subpopulações de Linfócitos T/transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA