Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Vaccine Immunol ; 24(4)2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28179404

RESUMO

Malaria is caused by parasites of the genus Plasmodium, which are transmitted to humans by the bites of Anopheles mosquitoes. After the elimination of Plasmodium falciparum, it is predicted that Plasmodium vivax will remain an important cause of morbidity and mortality outside Africa, stressing the importance of developing a vaccine against P. vivax malaria. In this study, we assessed the immunogenicity and protective efficacy of two P. vivax antigens, apical membrane antigen 1 (AMA1) and the 42-kDa C-terminal fragment of merozoite surface protein 1 (MSP142) in a plasmid recombinant DNA prime/adenoviral (Ad) vector boost regimen in Aotus monkeys. Groups of 4 to 5 monkeys were immunized with plasmid DNA alone, Ad alone, prime/boost regimens with each antigen, prime/boost regimens with both antigens, and empty vector controls and then subjected to blood-stage challenge. The heterologous immunization regimen with the antigen pair was more protective than either antigen alone or both antigens delivered with a single vaccine platform, on the basis of their ability to induce the longest prepatent period and the longest time to the peak level of parasitemia, the lowest peak and mean levels of parasitemia, the smallest area under the parasitemia curve, and the highest self-cure rate. Overall, prechallenge MSP142 antibody titers strongly correlated with a decreased parasite burden. Nevertheless, a significant proportion of immunized animals developed anemia. In conclusion, the P. vivax plasmid DNA/Ad serotype 5 vaccine encoding blood-stage parasite antigens AMA1 and MSP142 in a heterologous prime/boost immunization regimen provided significant protection against blood-stage challenge in Aotus monkeys, indicating the suitability of these antigens and this regimen for further development.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária Vivax/prevenção & controle , Proteínas de Membrana/imunologia , Proteína 1 de Superfície de Merozoito/imunologia , Proteínas de Protozoários/imunologia , Vacinas de DNA/imunologia , Anemia/prevenção & controle , Animais , Anticorpos Antiprotozoários/sangue , Aotidae , Modelos Animais de Doenças , Feminino , Vacinas Antimaláricas/administração & dosagem , Malária Vivax/imunologia , Masculino , Parasitemia/prevenção & controle , Resultado do Tratamento , Vacinas de DNA/administração & dosagem
2.
Malar J ; 15(1): 377, 2016 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-27448805

RESUMO

BACKGROUND: In this phase 1 clinical trial, healthy adult, malaria-naïve subjects were immunized with radiation-attenuated Plasmodium falciparum sporozoites (PfRAS) by mosquito bite and then underwent controlled human malaria infection (CHMI). The PfRAS model for immunization against malaria had previously induced >90 % sterile protection against homologous CHMI. This study was to further explore the safety, tolerability and protective efficacy of the PfRAS model and to provide biological specimens to characterize protective immune responses and identify protective antigens in support of malaria vaccine development. METHODS: Fifty-seven subjects were screened, 41 enrolled and 30 received at least one immunization. The true-immunized subjects received PfRAS via mosquito bite and the mock-immunized subjects received mosquito bites from irradiated uninfected mosquitoes. Sera and peripheral blood mononuclear cells (PBMCs) were collected before and after PfRAS immunizations. RESULTS: Immunization with PfRAS was generally safe and well tolerated, and repeated immunization via mosquito bite did not appear to increase the risk or severity of AEs. Local adverse events (AEs) of true-immunized and mock-immunized groups consisted of erythaema, papules, swelling, and induration and were consistent with reactions from mosquito bites seen in nature. Two subjects, one true- and one mock-immunized, developed large local reactions that completely resolved, were likely a result of mosquito salivary antigens, and were withdrawn from further participation as a safety precaution. Systemic AEs were generally rare and mild, consisting of headache, myalgia, nausea, and low-grade fevers. Two true-immunized subjects experienced fever, malaise, myalgia, nausea, and rigours approximately 16 h after immunization. These symptoms likely resulted from pre-formed antibodies interacting with mosquito salivary antigens. Ten subjects immunized with PfRAS underwent CHMI and five subjects (50 %) were sterilely protected and there was a significant delay to parasitaemia in the other five subjects. All ten subjects developed humoral immune responses to whole sporozoites and to the circumsporozoite protein prior to CHMI, although the differences between protected and non-protected subjects were not statistically significant for this small sample size. CONCLUSIONS: The protective efficacy of this clinical trial (50 %) was notably less than previously reported (>90 %). This may be related to differences in host genetics or the inherent variability in mosquito biting behavior and numbers of sporozoites injected. Differences in trial procedures, such as the use of leukapheresis prior to CHMI and of a longer interval between the final immunization and CHMI in these subjects compared to earlier trials, may also have reduced protective efficacy. This trial has been retrospectively registered at ISRCTN ID 17372582, May 31, 2016.


Assuntos
Anticorpos Antiprotozoários/sangue , Culicidae/fisiologia , Mordeduras e Picadas de Insetos , Vacinas Antimaláricas/efeitos adversos , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Adolescente , Adulto , Animais , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Humanos , Vacinas Antimaláricas/administração & dosagem , Masculino , Pessoa de Meia-Idade , Plasmodium falciparum/efeitos da radiação , Esporozoítos/imunologia , Esporozoítos/efeitos da radiação , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/imunologia , Adulto Jovem
3.
Hum Vaccin Immunother ; 8(11): 1564-84, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23151451

RESUMO

When introduced in the 1990s, immunization with DNA plasmids was considered potentially revolutionary for vaccine development, particularly for vaccines intended to induce protective CD8 T cell responses against multiple antigens. We conducted, in 1997-1998, the first clinical trial in healthy humans of a DNA vaccine, a single plasmid encoding Plasmodium falciparum circumsporozoite protein (PfCSP), as an initial step toward developing a multi-antigen malaria vaccine targeting the liver stages of the parasite. As the next step, we conducted in 2000-2001 a clinical trial of a five-plasmid mixture called MuStDO5 encoding pre-erythrocytic antigens PfCSP, PfSSP2/TRAP, PfEXP1, PfLSA1 and PfLSA3. Thirty-two, malaria-naïve, adult volunteers were enrolled sequentially into four cohorts receiving a mixture of 500 µg of each plasmid plus escalating doses (0, 20, 100 or 500 µg) of a sixth plasmid encoding human granulocyte macrophage-colony stimulating factor (hGM-CSF). Three doses of each formulation were administered intramuscularly by needle-less jet injection at 0, 4 and 8 weeks, and each cohort had controlled human malaria infection administered by five mosquito bites 18 d later. The vaccine was safe and well-tolerated, inducing moderate antigen-specific, MHC-restricted T cell interferon-γ responses but no antibodies. Although no volunteers were protected, T cell responses were boosted post malaria challenge. This trial demonstrated the MuStDO5 DNA and hGM-CSF plasmids to be safe and modestly immunogenic for T cell responses. It also laid the foundation for priming with DNA plasmids and boosting with recombinant viruses, an approach known for nearly 15 y to enhance the immunogenicity and protective efficacy of DNA vaccines.


Assuntos
Antígenos de Protozoários/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/uso terapêutico , Plasmodium falciparum/imunologia , Plasmodium falciparum/patogenicidade , Esporozoítos/imunologia , Vacinas de DNA/imunologia , Vacinas de DNA/uso terapêutico , Adulto , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Vacinas Antimaláricas/administração & dosagem , Masculino , Pessoa de Meia-Idade , Plasmídeos/genética , Vacinas de DNA/efeitos adversos , Adulto Jovem
4.
Malar J ; 6: 135, 2007 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-17925026

RESUMO

The present study has evaluated the immunogenicity of single or multiple Plasmodium falciparum (Pf) antigens administered in a DNA prime/poxvirus boost regimen with or without the poloxamer CRL1005 in rhesus monkeys. Animals were primed with PfCSP plasmid DNA or a mixture of PfCSP, PfSSP2/TRAP, PfLSA1, PfAMA1 and PfMSP1-42 (CSLAM) DNA vaccines in PBS or formulated with CRL1005, and subsequently boosted with ALVAC-Pf7, a canarypox virus expressing the CSLAM antigens. Cell-mediated immune responses were evaluated by IFN-gamma ELIspot and intracellular cytokine staining, using recombinant proteins and overlapping synthetic peptides. Antigen-specific and parasite-specific antibody responses were evaluated by ELISA and IFAT, respectively. Immune responses to all components of the multi-antigen mixture were demonstrated following immunization with either DNA/PBS or DNA/CRL1005, and no antigen interference was observed in animals receiving CSLAM as compared to PfCSP alone. These data support the down-selection of the CSLAM antigen combination. CRL1005 formulation had no apparent effect on vaccine-induced T cell or antibody responses, either before or after viral boost. In high responder monkeys, CD4+IL-2+ responses were more predominant than CD8+ T cell responses. Furthermore, CD8+ IFN-gamma responses were detected only in the presence of detectable CD4+ T cell responses. Overall, this study demonstrates the potential for multivalent Pf vaccines based on rational antigen selection and combination, and suggests that further formulation development to increase the immunogenicity of DNA encoded antigens is warranted.


Assuntos
Antígenos de Protozoários/imunologia , Imunização Secundária/métodos , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Poxviridae/imunologia , Vacinas de DNA/imunologia , Animais , Anticorpos Antiprotozoários/biossíntese , Antígenos de Protozoários/administração & dosagem , Antígenos de Protozoários/genética , Imunização , Macaca mulatta , Vacinas Antimaláricas/imunologia , Plasmídeos , Vacinas de DNA/administração & dosagem
5.
Infect Immun ; 73(5): 2863-72, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15845492

RESUMO

A mixture of DNA plasmids expressing five Plasmodium falciparum pre-erythrocyte-stage antigens was administered with or without a DNA plasmid encoding human granulocyte-macrophage colony-stimulating factor (hGM-CSF) as an immune enhancer. After DNA immunization, antigen-specific gamma interferon (IFN-gamma) responses were detected by ELISPOT in 15/31 volunteers to multiple class I- and/or class II-restricted T-cell epitopes derived from all five antigens. Responses to multiple epitopes (

Assuntos
Antígenos de Protozoários/imunologia , Interferon gama/sangue , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Vacinas de DNA/imunologia , Animais , Antígenos de Protozoários/administração & dosagem , Antígenos de Protozoários/genética , Mordeduras e Picadas , Culicidae , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Imunização , Vacinas Antimaláricas/administração & dosagem , Plasmídeos , Linfócitos T/imunologia , Resultado do Tratamento , Vacinas de DNA/administração & dosagem
6.
Antimicrob Agents Chemother ; 48(7): 2455-63, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15215094

RESUMO

CEL-1000 (DGQEEKAGVVSTGLIGGG) is a novel potential preventative and therapeutic agent. We report that CEL-1000 confers a high degree of protection against Plasmodium sporozoite challenge in a murine model of malaria, as shown by the total absence of blood stage infection following challenge with 100 sporozoites (100% protection) and by a substantial reduction (400-fold) of liver stage parasite RNA following challenge with 50,000 sporozoites. CEL-1000 protection was demonstrated in A/J (H-2(a)) and C3H/HeJ (H-2(k)) mice but not in BALB/c (H-2(d)) or CAF1 (A/J x BALB/c F(1) hybrid) mice. In CEL-1000-treated and protected mice, high levels of gamma interferon (IFN-gamma) in serum and elevated frequencies of hepatic and splenic CD4+ IFN-gamma-positive T cells were detected 24 h after administration of an additional dose of CEL-1000. Treatment of A/J mice that received CEL-1000 with antibodies against IFN-gamma just prior to challenge abolished the protection, and a similar treatment with antibodies against CD4+ T cells partially reduced the level of protection, while treatment with control antibodies or antibodies specific for interleukin-12 (IL-12), CD8+ T cells, or NK cells had no effect. Our data establish that the protection induced by CEL-1000 is dependent on IFN-gamma and is partially dependent on CD4+ T cells but is independent of CD8+ T cells, NK cells, and IL-12 at the effector phase and does not induce a detectable antibody response.


Assuntos
Antígenos de Protozoários/imunologia , Antimaláricos/farmacologia , Genes MHC da Classe II/genética , Malária/prevenção & controle , Sequência de Aminoácidos , Animais , Especificidade de Anticorpos , Citocinas/sangue , Ensaio de Imunoadsorção Enzimática , Feminino , Interferon gama/imunologia , Cinética , Fígado/química , Fígado/parasitologia , Malária/imunologia , Malária/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Plasmodium berghei/imunologia , Plasmodium yoelii/imunologia , RNA/química , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Vaccine ; 22(19): 2368-73, 2004 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-15193396

RESUMO

CEL-1000 (derG, DGQEEKAGVVSTGLIGGG) is a small immunomodulatory peptide which delivers demonstrated protective activity in two infectious disease challenge models (HSV and malaria) and an allogenic tumor vaccine model. CEL-1000 and other activators (defensin-beta, CpG ODN, and imiquimod) of the innate immune system promote IFN-gamma-associated protective responses. CEL-1000 is an improved form of peptide G (a peptide from human MHC II beta chain second domain, aa 135-149) known to enhance immune responses of other immunogenic peptides. Since defensin-beta, CpG ODN, and imiquimod have been shown to possess adjuvant activity, we investigated the adjuvant effect of peptide G and CEL-1000 as conjugates with HIV and malaria peptides. Antibody titers and isotypes were evaluated on serum taken from select days following immunization. Results for CEL-1000 and G peptide conjugates were compared with results for KLH conjugates of the same HIV peptide from the p17 molecule (87-116) referred to as HGP-30. Studies demonstrated that comparable titers were seen on day 28, 42, 63, and 77 with either G or KLH-HGP-30 peptide conjugates. In another study, CEL-1000 conjugates (CEL-1000-HGP-30) demonstrated a 4-10-fold higher titer antibody response than seen with several other peptide conjugates of the same HGP-30 peptide. Improved adjuvant activity of CEL-1000 in peptide conjugates was also demonstrated by a shift in the antibody isotypes toward a Th1 response (IgG2a). The IgG2a/IgG1, ratio for G-HGP-30 HIV or KLH-HGP-30 HIV conjugates were lower than for the CEL-1000-HGP-30 HIV conjugate. A similar favoring of the IgG2a/IgG1 ratio was seen for a malaria peptide conjugate (CEL-1000-SF/GF) compared to the un-conjugated peptide (SF-GF). CEL-1000 also showed adjuvant activity in an allogenic tumor vaccine model. As expected for an adjuvant, CEL-1000 or G does not induce detectable self-directed or cross reactive antibodies. CEL-1000 is currently being investigated for use as an adjuvant with conventional vaccines. It is expected that IgG2a antibodies would be preferably generated by CEL-1000 adjuvancy and could enhance in vivo clearance of antigens or pathogens.


Assuntos
Adjuvantes Imunológicos , Peptídeos/imunologia , Células Th1/imunologia , Animais , Ensaio de Imunoadsorção Enzimática , Camundongos , Peptídeos/efeitos adversos
8.
J Immunol ; 172(9): 5561-9, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15100299

RESUMO

Vaccine-induced protection against diseases like malaria, AIDS, and cancer may require induction of Ag-specific CD8(+) and CD4(+) T cell and Ab responses in the same individual. In humans, a recombinant Plasmodium falciparum circumsporozoite protein (PfCSP) candidate vaccine, RTS,S/adjuvant system number 2A (AS02A), induces T cells and Abs, but no measurable CD8(+) T cells by CTL or short-term (ex vivo) IFN-gamma ELISPOT assays, and partial short-term protection. P. falciparum DNA vaccines elicit CD8(+) T cells by these assays, but no protection. We report that sequential immunization with a PfCSP DNA vaccine and RTS,S/AS02A induced PfCSP-specific Abs and Th1 CD4(+) T cells, and CD8(+) cytotoxic and Tc1 T cells. Depending upon the immunization regime, CD4(+) T cells were involved in both the induction and production phases of PfCSP-specific IFN-gamma responses, whereas, CD8(+) T cells were involved only in the production phase. IFN-gamma mRNA up-regulation was detected in both CD45RA(-) (CD45RO(+)) and CD45RA(+)CD4(+) and CD8(+) T cell populations after stimulation with PfCSP peptides. This finding suggests CD45RA(+) cells function as effector T cells. The induction in humans of the three primary Ag-specific adaptive immune responses establishes a strategy for developing immunization regimens against diseases in desperate need of vaccines.


Assuntos
Anticorpos Antiprotozoários/biossíntese , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Esquemas de Imunização , Vacinas Antimaláricas/administração & dosagem , Plasmodium falciparum/imunologia , Proteínas de Protozoários/administração & dosagem , Vacinas de DNA/administração & dosagem , Sequência de Aminoácidos , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Testes Imunológicos de Citotoxicidade , Epitopos de Linfócito T/administração & dosagem , Epitopos de Linfócito T/imunologia , Anticorpos Anti-Hepatite B/biossíntese , Antígenos de Superfície da Hepatite B/imunologia , Humanos , Imunização Secundária/métodos , Interferon gama/biossíntese , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Dados de Sequência Molecular , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Subpopulações de Linfócitos T , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Vacinas de DNA/genética , Vacinas de DNA/imunologia
9.
Vaccine ; 22(13-14): 1592-603, 2004 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-15068840

RESUMO

Optimal protection against malaria may require induction of high levels of protective antibody and CD8(+) and CD4(+) T cell responses. In humans, malaria DNA vaccines elicit CD8(+) cytotoxic T cells (CTL) and IFNgamma responses as measured by short-term (ex vivo) ELISPOT assays, and recombinant proteins elicit antibodies and excellent T cell responses, but no CD8(+) CTL or CD8(+) IFNgamma-producing cells as measured by ex vivo ELISPOT. Priming with DNA and boosting with recombinant pox virus elicits much better T cell responses than DNA alone, but not antibody responses. In an attempt to elicit antibodies and enhanced T cell responses, we administered RTS,S/AS02A, a partially protective Plasmodium falciparum recombinant circumsporozoite protein (CSP) vaccine in adjuvant, to volunteers previously immunized with a P. falciparum CSP DNA vaccine (VCL-2510) and to naïve volunteers. This vaccine regimen was well tolerated and safe. The volunteers who received RTS,S/AS02A alone had, as expected, antibody and CD4(+) T cell responses, but no CD8(+) T cell responses. Volunteers who received PfCSP DNA followed by RTS,S/AS02A had antibody and CD8(+) and CD4(+) T cell responses (Wang et al., submitted). Sequential immunization with DNA and recombinant protein, also called heterologous prime-boost, led to enhanced immune responses as compared to DNA or recombinant protein alone, suggesting that it might provide enhanced protective immunity.


Assuntos
Anticorpos Antiprotozoários/biossíntese , Vacinas Antimaláricas/imunologia , Plasmodium falciparum/imunologia , Vacinas de DNA/imunologia , Adolescente , Adulto , Animais , Anticorpos Antiprotozoários/análise , Linfócitos B/imunologia , Feminino , Anticorpos Anti-Hepatite B/biossíntese , Antígenos de Superfície da Hepatite B/imunologia , Humanos , Esquemas de Imunização , Vacinas Antimaláricas/efeitos adversos , Masculino , Esporos de Protozoários/imunologia , Linfócitos T/imunologia , Vacinas de DNA/efeitos adversos , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/imunologia
10.
Infect Immun ; 72(3): 1594-602, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14977966

RESUMO

We describe a novel approach for identifying target antigens for preerythrocytic malaria vaccines. Our strategy is to rapidly test hundreds of DNA vaccines encoding exons from the Plasmodium yoelii yoelii genomic sequence. In this antigen identification method, we measure reduction in parasite burden in the liver after sporozoite challenge in mice. Orthologs of protective P. y. yoelii genes can then be identified in the genomic databases of Plasmodium falciparum and Plasmodium vivax and investigated as candidate antigens for a human vaccine. A pilot study to develop the antigen identification method approach used 192 P. y. yoelii exons from genes expressed during the sporozoite stage of the life cycle. A total of 182 (94%) exons were successfully cloned into a DNA immunization vector with the Gateway cloning technology. To assess immunization strategies, mice were vaccinated with 19 of the new DNA plasmids in addition to the well-characterized protective plasmid encoding P. y. yoelii circumsporozoite protein. Single plasmid immunization by gene gun identified a novel vaccine target antigen which decreased liver parasite burden by 95% and which has orthologs in P. vivax and P. knowlesi but not P. falciparum. Intramuscular injection of DNA plasmids produced a different pattern of protective responses from those seen with gene gun immunization. Intramuscular immunization with plasmid pools could reduce liver parasite burden in mice despite the fact that none of the plasmids was protective when given individually. We conclude that high-throughput cloning of exons into DNA vaccines and their screening is feasible and can rapidly identify new malaria vaccine candidate antigens.


Assuntos
Antígenos de Protozoários/genética , Vacinas Antimaláricas/genética , Plasmodium yoelii/genética , Plasmodium yoelii/imunologia , Animais , Anticorpos Antiprotozoários/sangue , Sequência de Bases , Biolística , Clonagem Molecular , Primers do DNA , DNA de Protozoário/genética , Éxons , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Genoma de Protozoário , Humanos , Injeções Intramusculares , Fígado/parasitologia , Malária/imunologia , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos/genética , Plasmodium yoelii/crescimento & desenvolvimento , Reação em Cadeia da Polimerase , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas de DNA/farmacologia
11.
J Biol Chem ; 279(10): 9490-6, 2004 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-14676185

RESUMO

Plasmodium sporozoites are transmitted through the bite of infected mosquitoes and invade hepatocytes as a first and obligatory step of the parasite life cycle in man. Hepatocyte invasion involves proteins secreted from parasite vesicles called micronemes, the most characterized being the thrombospondin-related adhesive protein (TRAP). Here we investigated the expression and function of another microneme protein recently identified in Plasmodium falciparum sporozoites, apical membrane antigen 1 (AMA-1). P. falciparum AMA-1 is expressed in sporozoites and is lost after invasion of hepatocytes, and anti-AMA-1 antibodies inhibit sporozoite invasion, suggesting that the protein is involved during invasion of hepatocytes. As observed with TRAP, AMA-1 is initially mostly sequestered within the sporozoite. Upon microneme exocytosis, AMA-1 and TRAP relocate to the sporozoite surface, where they are proteolytically cleaved, resulting in the shedding of soluble fragments. A subset of serine protease inhibitors blocks the processing and shedding of both AMA-1 and TRAP and inhibits sporozoite infectivity, suggesting that interfering with sporozoite proteolytic processing may constitute a valuable strategy to prevent hepatocyte infection.


Assuntos
Antígenos de Protozoários/metabolismo , Hepatócitos/parasitologia , Proteínas de Membrana/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Animais , Células Cultivadas , Hepatócitos/metabolismo , Humanos , Plasmodium falciparum/patogenicidade , Esporozoítos/metabolismo , Esporozoítos/patogenicidade
13.
Trans R Soc Trop Med Hyg ; 96(5): 560-4, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12474490

RESUMO

The aim of this study was to determine the exposure of child citizens of the United Arab Emirates (UAE) to Plasmodium vivax, and to elucidate if it was related to place of residence or previous international travel to malaria-endemic areas. Blood samples were collected from 1010 primary schoolchildren resident in 7 out of 9 districts of the UAE during October and November 1999. Plasma samples were tested for antibodies against MAP4 (DGQPAGDR)3P2P30, a multiple antigen peptide containing the repeat amino acid sequences of P. vivax circumsporozoite protein (CSP), conjugated to 2 T-helper epitopes, P2 (QYIKANSKFIGITE) and P30 (FNNFTVSFWLRVPKVSASHLE) from tetanus toxin. For confirmation of P. vivax-specific reactivity, positive samples were further tested against (AGDR)6, a synthetic peptide containing 6 copies of a protective epitope within the CSP, and against a recombinant CSP, designated as NS1(81)V20. Results indicated that 3.3% of the children were seropositive. The seropositivity rates differed significantly in relation to place of residence, whereas travel outside the UAE did not significantly affect the exposure rates to P. vivax.


Assuntos
Anticorpos Antiprotozoários/sangue , Malária Vivax/parasitologia , Plasmodium vivax/imunologia , Proteínas de Protozoários/imunologia , Animais , Criança , Estudos Transversais , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Masculino , Prevalência , Emirados Árabes Unidos/epidemiologia
14.
Hum Gene Ther ; 13(13): 1551-60, 2002 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12228010

RESUMO

Introduction of a new vaccine requires choosing a delivery system that provides safe administration and the desired level of immunogenicity. The safety, tolerability, and immunogenicity of three monthly 2.5-mg doses of a PfCSP DNA vaccine were evaluated in healthy volunteers as administered intramuscularly (IM) by needle, IM by jet injection (Biojector or IM/intradermally (ID) by jet injection. Vaccine administration was well-tolerated. Adverse events were primarily mild and limited to the site of injection (98%). Jet injections (either IM or ID) were associated with approximately twice as many adverse events per immunization as needle IM, but nevertheless were strongly and consistently preferred in opinion polls taken during the study. No volunteers had clinically significant biochemical or hematologic changes or detectable anti-dsDNA antibodies. In conclusion, the injection of Plasmodium falciparum circumsporozoite (PfCSP) DNA vaccine appeared to be safe and well-tolerated when administered by any of the three modes of delivery. However, despite improved antibody responses following both jet injection and ID delivery in animal models, no antibodies could be detected in volunteers by immunofluorescence antibody test (IFAT) or enzyme-linked immunosorbent assay (ELISA) after DNA vaccination.


Assuntos
Malária/prevenção & controle , Vacinas de DNA/administração & dosagem , Animais , Humanos , Injeções Intradérmicas , Injeções Intramusculares , Malária/imunologia , Plasmodium falciparum/imunologia , Vacinas de DNA/efeitos adversos , Vacinas de DNA/imunologia
15.
Infect Immun ; 70(7): 3493-9, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12065488

RESUMO

The persistence of immunity to malaria induced in mice by a heterologous DNA priming and poxvirus boosting regimen was characterized. Mice were immunized by priming with DNA vaccine plasmids encoding the Plasmodium yoelii circumsporozoite protein (PyCSP) and murine granulocyte-macrophage colony-stimulating factor and boosting with recombinant vaccinia encoding PyCSP. BALB/c mice immunized with either high-dose (100 microg of p PyCSP plus 30 microg of pGM-CSF) or low-dose (1 microg of p PyCSP plus 1 microg of pGM-CSF DNA) priming were protected against challenge with 50 P. yoelii sporozoites. Protection 2 weeks after immunization was 70 to 100%, persisted at this level for at least 20 weeks, and declined to 30 to 40% by 28 weeks. Eight of eight mice protected at 20 weeks were still protected when rechallenged at 40 weeks. The antigen (Ag)-specific effector CD8(+)-T-cell population present 2 weeks after boosting had ex vivo Ag-specific cytolytic activity, expressed both gamma interferon (IFN-gamma) and tumor necrosis factor alpha, and constituted 12 to 20% of splenic CD8(+) T cells. In contrast, the memory CD8(+)-Ag-specific-cell population at 28 weeks lacked cytolytic activity and constituted only 6% of splenic CD8(+) T cells, but at the single-cell level it produced significantly higher levels of IFN-gamma than the effectors. High levels of Ag- or parasite-specific antibodies present 2 weeks after boosting had declined three- to sevenfold by 28 weeks. Low-dose priming was similarly immunogenic and as protective as high-dose priming against a 50-, but not a 250-, sporozoite challenge. These results demonstrate that a heterologous priming and boosting vaccination can provide lasting protection against malaria in this model system.


Assuntos
Antígenos de Protozoários/imunologia , Linfócitos T CD8-Positivos/imunologia , DNA de Protozoário/imunologia , Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Plasmodium yoelii/imunologia , Proteínas de Protozoários/imunologia , Vacinas de DNA/imunologia , Sequência de Aminoácidos , Animais , Antígenos de Protozoários/genética , Feminino , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Imunização Secundária , Memória Imunológica/imunologia , Malária/imunologia , Vacinas Antimaláricas/genética , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Plasmodium yoelii/genética , Proteínas de Protozoários/genética , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/imunologia , Fatores de Tempo , Vacinas de DNA/genética , Vaccinia virus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...