Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Infect Dis ; 19(1): 920, 2019 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-31664924

RESUMO

BACKGROUND: The only licensed malaria vaccine, RTS,S/AS01, has been developed for morbidity-control in young children. The potential impact on transmission of deploying such anti-infective vaccines to wider age ranges, possibly with co-administration of antimalarial treatment, is unknown. Combinations of existing malaria interventions is becoming increasingly important as evidence mounts that progress on reducing malaria incidence is stalling and threatened by resistance. METHODS: Malaria transmission and intervention dynamics were simulated using OpenMalaria, an individual-based simulation model of malaria transmission, by considering a seasonal transmission setting and by varying epidemiological and setting parameters such as transmission intensity, case management, intervention types and intervention coverages. Chemopreventive drugs and anti-infective vaccine efficacy profiles were based on previous studies in which model parameters were fitted to clinical trial data. These intervention properties were used to evaluate the potential of seasonal mass applications of preventative anti-infective malaria vaccines, alone or in combination with chemoprevention, to reduce malaria transmission, prevent resurgence, and/or reach transmission interruption. RESULTS: Deploying a vaccine to all ages on its own is a less effective intervention strategy compared to chemoprevention alone. However, vaccines combined with drugs are likely to achieve dramatic prevalence reductions and in few settings, transmission interruption. The combined mass intervention will result in lower prevalence following the intervention compared to chemoprevention alone and will increase chances of interruption of transmission resulting from a synergistic effect between both interventions. The combination of vaccine and drug increases the time before transmission resurges after mass interventions cease compared to mass treatment alone. Deploying vaccines and drugs together requires fewer rounds of mass intervention and fewer years of intervention to achieve the same public health impact as chemoprevention alone. CONCLUSIONS: Through simulations we identified a previously unidentified value of deploying vaccines with drugs, namely the greatest benefit will be in preventing and delaying transmission resurgence for longer periods than with other human targeted interventions. This is suggesting a potential role for deploying vaccines alongside drugs in transmission foci as part of surveillance-response strategies.


Assuntos
Antimaláricos/administração & dosagem , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/epidemiologia , Malária Falciparum/prevenção & controle , Administração Massiva de Medicamentos , Vacinação em Massa , Modelos Teóricos , Estações do Ano , Adulto , Quimioprevenção/métodos , Criança , Pré-Escolar , Transmissão de Doença Infecciosa/prevenção & controle , Quimioterapia Combinada , Humanos , Lactente , Malária Falciparum/tratamento farmacológico , Plasmodium falciparum/imunologia , Prevalência
2.
Malar J ; 14: 72, 2015 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-25885325

RESUMO

BACKGROUND: The malaria vaccine RTS,S induces antibodies against the Plasmodium falciparum circumsporozoite protein (CSP) and the concentration of Immunoglobulin G (IgG) against the repeat region of CSP following vaccination is associated with protection from P. falciparum malaria. So far, only the quantity of anti-CSP IgG has been measured and used to predict vaccination success, although quality (measured as avidity) of the antigen-antibody interaction shall be important since only a few sporozoites circulate for a short time after an infectious mosquito bite, likely requiring fast and strong binding. METHODS: Quantity and avidity of anti-CSP IgG in African infants who received RTS,S/AS01E in a 0-1-2-month or a 0-1-7-month schedule in a phase 2 clinical trial were measured by enzyme-linked immunosorbent assay. Antibody avidity was defined as the proportion of IgG able to bind in the presence of a chaotropic agent (avidity index). The effect of CSP-specific IgG concentration and avidity on protective efficacy was modelled using Cox proportional hazards. RESULTS: After the third dose, quantity and avidity were similar between the two vaccination schedules. IgG avidity after the last vaccine injection was not associated with protection, whereas the change in avidity following second and third RTS,S/AS01E injection was associated with a 54% risk reduction of getting malaria (hazard ratio: 0.46; 95% confidence interval (CI): 0.22-0.99) in those participants with a change in avidity above the median. The change in anti-CSP IgG concentration following second and third injection was associated with a 77% risk reduction of getting malaria (hazard ratio: 0.23, 95% CI: 0.11-0.51). CONCLUSIONS: Change in IgG response between vaccine doses merits further evaluation as a surrogate marker for RTS,S efficacy. TRIAL REGISTRATION: ClinicalTrials.gov Identifier NCT00436007 .


Assuntos
Esquemas de Imunização , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Proteínas de Protozoários/imunologia , Afinidade de Anticorpos/imunologia , Ensaio de Imunoadsorção Enzimática , Humanos , Estimativa de Kaplan-Meier , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia
3.
PLoS One ; 9(11): e109654, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25405724

RESUMO

BACKGROUND: Controlled human malaria infection (CHMI) studies which recapitulate mosquito-borne infection are a critical tool to identify protective vaccine and drug candidates for advancement to field trials. In partnership with the Walter Reed Army Institute of Research, the CHMI model was established at the Seattle Biomedical Research Institute's Malaria Clinical Trials Center (MCTC). Activities and reagents at both centers were aligned to ensure comparability and continued safety of the model. To demonstrate successful implementation, CHMI was performed in six healthy malaria-naïve volunteers. METHODS: All volunteers received NF54 strain Plasmodium falciparum by the bite of five infected Anopheles stephensi mosquitoes under controlled conditions and were monitored for signs and symptoms of malaria and for parasitemia by peripheral blood smear. Subjects were treated upon diagnosis with chloroquine by directly observed therapy. Immunological (T cell and antibody) and molecular diagnostic (real-time quantitative reverse transcriptase polymerase chain reaction [qRT-PCR]) assessments were also performed. RESULTS: All six volunteers developed patent parasitemia and clinical malaria. No serious adverse events occurred during the study period or for six months post-infection. The mean prepatent period was 11.2 days (range 9-14 days), and geometric mean parasitemia upon diagnosis was 10.8 parasites/µL (range 2-69) by microscopy. qRT-PCR detected parasites an average of 3.7 days (range 2-4 days) earlier than blood smears. All volunteers developed antibodies to the blood-stage antigen merozoite surface protein 1 (MSP-1), which persisted up to six months. Humoral and cellular responses to pre-erythrocytic antigens circumsporozoite protein (CSP) and liver-stage antigen 1 (LSA-1) were limited. CONCLUSION: The CHMI model was safe, well tolerated and characterized by consistent prepatent periods, pre-symptomatic diagnosis in 3/6 subjects and adverse event profiles as reported at established centers. The MCTC can now evaluate candidates in the increasingly diverse vaccine and drug pipeline using the CHMI model. TRIAL REGISTRATION: ClinicalTrials.gov NCT01058226.


Assuntos
Experimentação Humana , Malária Falciparum/diagnóstico , Plasmodium falciparum/patogenicidade , Esporozoítos , Adulto , Animais , Anopheles/parasitologia , Anopheles/fisiologia , Mordeduras e Picadas/parasitologia , Feminino , Humanos , Malária Falciparum/complicações , Malária Falciparum/etiologia , Malária Falciparum/imunologia , Masculino , Plasmodium falciparum/fisiologia
4.
PLoS One ; 4(11): e7849, 2009 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-19924281

RESUMO

OBJECTIVE: RTS,S, a candidate vaccine for malaria, is a recombinant protein expressed in yeast containing part of the circumsporozoite protein (CSP) sequence of 3D7 strain of Plasmodium falciparum linked to the hepatitis B surface antigen in a hybrid protein. The RTS,S antigen is formulated with GSK Biologicals' proprietary Adjuvant Systems AS02(A) or AS01(B). A recent trial of the RTS,S/AS02(A) and RTS,S/AS01(B) vaccines evaluated safety, immunogenicity and impact on the development of parasitemia of the two formulations. Parasite isolates from this study were used to determine the molecular impact of RTS,S/AS02(A) and RTS,S/AS01(B) on the multiplicity of infection (MOI) and the csp allelic characteristics of subsequent parasitemias. DESIGN: The distribution of csp sequences and the MOI of the infecting strains were examined at baseline and in break-through infections from vaccinated individuals and from those receiving a non-malarial vaccine. SETTING: The study was conducted in Kombewa District, western Kenya. PARTICIPANTS: Semi-immune adults from the three study arms provided isolates at baseline and during break-through infections. OUTCOME: Parasite isolates used for determining MOI and divergence of csp T cell-epitopes were 191 at baseline and 87 from break-through infections. RESULTS: Grouping recipients of RTS,S/AS01(A) and RTS,S/AS02(B) together, vaccine recipients identified as parasite-positive by microscopy contained significantly fewer parasite genotypes than recipients of the rabies vaccine comparator (median in pooled RTS,S groups: 3 versus 4 in controls, P = 0.0313). When analyzed separately, parasitaemic individuals in the RTS,S/AS01(B) group, but not the RTS,S/AS02(A) group, were found to have significantly fewer genotypes than the comparator group. Two individual amino acids found in the vaccine construct (Q339 in Th2R and D371 in Th3R) were observed to differ in incidence between vaccine and comparator groups but in different directions; parasites harboring Q339 were less common among pooled RTS,S/AS vaccine recipients than among recipients of rabies vaccine, whereas parasites with D371 were more common among the RTS,S/AS groups. CONCLUSIONS: It is concluded that both RTS,S/AS vaccines reduce multiplicity of infection. Our results do not support the hypothesis that RTS,S/AS vaccines elicit preferential effects against pfcsp alleles with sequence similarity to the 3D7 pfcsp sequence employed in the vaccine construct.


Assuntos
Vacinas Antimaláricas/uso terapêutico , Malária/prevenção & controle , Malária/parasitologia , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Adolescente , Adulto , Alelos , Epitopos de Linfócito T/química , Feminino , Genótipo , Haplótipos , Humanos , Masculino , Polimorfismo Genético , Análise de Sequência de DNA
5.
Vaccine ; 25(41): 7007-11, 2007 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-17728022

RESUMO

As the 21st century unfolds, infectious diseases remain one of the most significant threats to our economy, our food animal production systems, animal welfare, and most importantly, the lives of people worldwide, regardless of their economic standing. The potential use of biological threat agents for terrorism or biowarfare further undermines the security of our society. Arguably, vaccines represent the single most cost-effective, medically delivered strategy for confronting these challenges. The workshop "Advances in Immunology and Vaccine Discovery" was organized to address these challenges, based on the conviction that the interface between immunology and vaccinology offers the best prospects for major breakthroughs in vaccine discovery and development. Six focus areas were identified by workshop organizers: (1) pathogen immune evasion; (2) innate immunity; (3) mucosal immunity; (4) immunogenetics; (5) comparative immunology; and (6) genomics. These areas provided opportunities to elucidate how protective immunity may relate to the disruption of the molecular mechanisms that underlie host-pathogen interactions. A report generated by workshop organizers and participants provides key recommendations and identifies important research gaps, needs, future steps, and potential strategic US-EU collaborations. The report is available on line through ScienceDirect (URL).


Assuntos
Controle de Doenças Transmissíveis/métodos , Doenças Transmissíveis/imunologia , Vacinas/imunologia , Humanos
6.
Vaccine ; 25(21): 4203-12, 2007 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-17442466

RESUMO

We report the first safety and immunogenicity trial of the Plasmodium falciparum vaccine candidate FMP2.1/AS02A, a recombinant E. coli-expressed protein based upon the apical membrane antigen-1 (AMA-1) of the 3D7 clone formulated with the AS02A adjuvant. We conducted an open-label, staggered-start, dose-escalating Phase I trial in 23 malaria-naïve volunteers who received 8, 20 or 40microg of FMP2.1 in a fixed volume of 0.5mL of AS02A on a 0, 1, and 2 month schedule. Nineteen of 23 volunteers received all three scheduled immunizations. The most frequent solicited local and systemic adverse events associated with immunization were injection site pain (68%) and headache (29%). There were no significant laboratory abnormalities or vaccine-related serious adverse events. All volunteers seroconverted after second immunization as determined by ELISA. Immune sera recognized sporozoites and merozoites by immunofluorescence assay (IFA), and exhibited both growth inhibition and processing inhibition activity against homologous (3D7) asexual stage parasites. Post-immunization, peripheral blood mononuculear cells exhibited FMP2.1-specific lymphoproliferation and IFN-gamma and IL-5 ELISPOT assay responses. This is the first PfAMA-1-based vaccine shown to elicit both potent humoral and cellular immunity in humans. Encouraged by the potential of FMP1/AS02A to target host immunity against PfAMA-1 that is known to be expressed by sporozoite, hepatic and erythrocytic stages, we have initiated field trials of FMP2.1/AS02A in an endemic population in the Republic of Mali.


Assuntos
Antígenos de Protozoários/imunologia , Lipídeo A/análogos & derivados , Vacinas Antimaláricas/efeitos adversos , Vacinas Antimaláricas/imunologia , Proteínas de Membrana/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Saponinas/imunologia , Adjuvantes Imunológicos , Adolescente , Adulto , Animais , Anticorpos Antiprotozoários/sangue , Linhagem Celular , Proliferação de Células , Células Cultivadas , Cricetinae , Combinação de Medicamentos , Ensaio de Imunoadsorção Enzimática , Escherichia coli/genética , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Cefaleia , Humanos , Imunização Secundária , Interferon gama/biossíntese , Interleucina-5/biossíntese , Leucócitos Mononucleares/imunologia , Lipídeo A/imunologia , Vacinas Antimaláricas/administração & dosagem , Masculino , Merozoítos/imunologia , Mesocricetus , Pessoa de Meia-Idade , Dor , Plasmodium falciparum/crescimento & desenvolvimento , Esporozoítos/imunologia , Vacinas Sintéticas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...