Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
BMC Med ; 22(1): 170, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38649867

RESUMEN

BACKGROUND: The stalling global progress in malaria control highlights the need for novel tools for malaria elimination, including transmission-blocking vaccines. Transmission-blocking vaccines aim to induce human antibodies that block parasite development in the mosquito and mosquitoes becoming infectious. The Pfs48/45 protein is a leading Plasmodium falciparum transmission-blocking vaccine candidate. The R0.6C fusion protein, consisting of Pfs48/45 domain 3 (6C) and the N-terminal region of P. falciparum glutamate-rich protein (R0), has previously been produced in Lactococcus lactis and elicited functional antibodies in rodents. Here, we assess the safety and transmission-reducing efficacy of R0.6C adsorbed to aluminium hydroxide with and without Matrix-M™ adjuvant in humans. METHODS: In this first-in-human, open-label clinical trial, malaria-naïve adults, aged 18-55 years, were recruited at the Radboudumc in Nijmegen, the Netherlands. Participants received four intramuscular vaccinations on days 0, 28, 56 and 168 with either 30 µg or 100 µg of R0.6C and were randomised for the allocation of one of the two different adjuvant combinations: aluminium hydroxide alone, or aluminium hydroxide combined with Matrix-M1™ adjuvant. Adverse events were recorded from inclusion until 84 days after the fourth vaccination. Anti-R0.6C and anti-6C IgG titres were measured by enzyme-linked immunosorbent assay. Transmission-reducing activity of participants' serum and purified vaccine-specific immunoglobulin G was assessed by standard membrane feeding assays using laboratory-reared Anopheles stephensi mosquitoes and cultured P. falciparum gametocytes. RESULTS: Thirty-one participants completed four vaccinations and were included in the analysis. Administration of all doses was safe and well-tolerated, with one related grade 3 adverse event (transient fever) and no serious adverse events occurring. Anti-R0.6C and anti-6C IgG titres were similar between the 30 and 100 µg R0.6C arms, but higher in Matrix-M1™ arms. Neat participant sera did not induce significant transmission-reducing activity in mosquito feeding experiments, but concentrated vaccine-specific IgGs purified from sera collected two weeks after the fourth vaccination achieved up to 99% transmission-reducing activity. CONCLUSIONS: R0.6C/aluminium hydroxide with or without Matrix-M1™ is safe, immunogenic and induces functional Pfs48/45-specific transmission-blocking antibodies, albeit at insufficient serum concentrations to result in transmission reduction by neat serum. Future work should focus on identifying alternative vaccine formulations or regimens that enhance functional antibody responses. TRIAL REGISTRATION: The trial is registered with ClinicalTrials.gov under identifier NCT04862416.


Asunto(s)
Vacunas contra la Malaria , Malaria Falciparum , Glicoproteínas de Membrana , Plasmodium falciparum , Proteínas Protozoarias , Adolescente , Adulto , Animales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , Adyuvantes Inmunológicos/administración & dosificación , Hidróxido de Aluminio/administración & dosificación , Anticuerpos Antiprotozoarios , Vacunas contra la Malaria/inmunología , Vacunas contra la Malaria/administración & dosificación , Malaria Falciparum/prevención & control , Malaria Falciparum/transmisión , Malaria Falciparum/inmunología , Países Bajos , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología
2.
Immunol Lett ; 258: 20-23, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37075916

RESUMEN

BACKGROUND: Here we assessed a possible relationship between baseline TGF-ß concentrations and acquisition of sterile immunity after Plasmodium falciparum sporozoite immunization. METHODS: TGF-ß concentrations were determined in samples of 65 malaria-naive volunteers in 4 studies either prior to and after challenge infection, or prior to and after first immunizing infection under chemoprophylaxis with P. falciparum sporozoites. RESULTS: High baseline TGF-ß concentrations were associated with rapid acquisition of sterile protection (p = 0.028). CONCLUSION: Baseline TGF-ß concentrations predict the efficiency of acquisition of sterile immunity following sporozoite immunization and may represent a steady-state regulatory mechanism to keep in check immune systems with a low threshold for activation.


Asunto(s)
Malaria Falciparum , Malaria , Animales , Humanos , Plasmodium falciparum , Esporozoítos , Malaria Falciparum/prevención & control , Malaria Falciparum/tratamiento farmacológico , Inmunización
3.
Ned Tijdschr Geneeskd ; 1642020 09 03.
Artículo en Holandés | MEDLINE | ID: mdl-33030326

RESUMEN

Five years ago, the very first malaria vaccine, RTS,S/AS01, received a positive evaluation by the European Medicines Agency. Although this vaccine does not achieve the WHO's target of 75% protection, it does set the standard for all new vaccine candidates. In this article, we describe the progress made in the development of several second-generation malaria vaccines, an area where Dutch research has made major contributions. These include vaccines with live, attenuated Plasmodium falciparumsporozoites and transmission-blocking vaccines. Thanks in part to Dutch contributions, the development of vaccines against malaria has recently made significant progress on the way to the finish line: a vaccine that provides protection to the most vulnerable population - young children in Africa.


Asunto(s)
Vacunas contra la Malaria/uso terapéutico , Malaria Falciparum/prevención & control , África , Niño , Preescolar , Humanos , Programas de Inmunización , Cooperación Internacional , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Países Bajos , Plasmodium falciparum/inmunología
4.
Ned Tijdschr Geneeskd ; 160: D444, 2016.
Artículo en Holandés | MEDLINE | ID: mdl-27848905

RESUMEN

BACKGROUND: Hyper-reactive malaria splenomegaly (HMS) is a rare and potentially severe complication of malaria. It is likely that the incidence of patients with HMS will rise in the Netherlands due to the recent increase in asylum-seekers from Sub-Saharan Africa. It can be difficult to diagnose this disease, as this case shows. CASE DESCRIPTION: A 31-year-old male from Eritrea was admitted with fever and dyspnea, caused by an influenza A-infection. The patient also presented with cachexia, pronounced hepatosplenomegaly and pancytopenia. Microscopic diagnostic analysis for malaria was negative. HMS was eventually diagnosed through high-sensitivity qPCR for malaria, which showed the presence of a very low level of Plasmodium falciparum parasitemia; furthermore, IgM levels were high and malaria serology was strongly positive. CONCLUSION: HMS should be considered in patients from malaria-endemic areas presenting with splenomegaly and pancytopenia. Because standard diagnostics for malaria are often negative in this population, malaria serology and sensitive qPCR play an important diagnostic role.


Asunto(s)
Malaria/diagnóstico , Malaria/tratamiento farmacológico , Refugiados , Esplenomegalia/diagnóstico , Esplenomegalia/tratamiento farmacológico , Adulto , Eritrea , Hepatomegalia , Humanos , Malaria/parasitología , Masculino , Países Bajos , Esplenomegalia/parasitología , Síndrome
6.
Parasite Immunol ; 37(11): 590-8, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26363409

RESUMEN

Immunization of malaria-naïve volunteers under chemoprophylaxis with Plasmodium falciparum sporozoites (CPS) efficiently and reproducibly induces sterile protection and thus constitutes an excellent model to study protective immune responses against malaria. Here, we performed the first longitudinal assessment of lymphocyte activation and differentiation kinetics during sporozoite immunization in 15 volunteers by ex vivo lymphocyte flow cytometry analysis. Both CD4 and CD8 T cells as well as γδT cells, NK cells and CD3+ CD56+ cells showed increased activation and proliferation following immunization. Transient induction of the transcription factor T-bet and the cytotoxic molecule granzyme B indicated a role of Th1 responses and cytotoxic T cells in CPS-induced immunity. The absolute number of γδT cells as well as the proportion of granzyme B-containing γδT cells showed a significant and sustained increase. Regulatory T-cell (Treg) proliferation was significantly higher after the second immunization in subjects subsequently not protected against challenge infection. These findings indicate an important role for γδT cells, Th1 and cytotoxic responses in whole sporozoite immunization with a possibly suppressive role of Tregs.


Asunto(s)
Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Malaria Falciparum/prevención & control , Plasmodium falciparum/inmunología , Esporozoítos/inmunología , Adulto , Animales , Citometría de Flujo , Granzimas/inmunología , Humanos , Inmunofenotipificación , Activación de Linfocitos , Malaria Falciparum/parasitología , Plasmodium falciparum/crecimiento & desarrollo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Adulto Joven
7.
Vaccine ; 33(52): 7476-82, 2015 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-26409813

RESUMEN

Stimulated by recent advances in malaria control and increased funding, the elimination of malaria is now considered to be an attainable goal for an increasing number of malaria-endemic regions. This has boosted the interest in transmission-reducing interventions including vaccines that target sexual, sporogenic, and/or mosquito-stage antigens to interrupt malaria transmission (SSM-VIMT). SSM-VIMT aim to prevent human malaria infection in vaccinated communities by inhibiting parasite development within the mosquito after a blood meal taken from a gametocyte carrier. Only a handful of target antigens are in clinical development and progress has been slow over the years. Major stumbling blocks include (i) the expression of appropriately folded target proteins and their downstream purification, (ii) insufficient induction of sustained functional blocking antibody titers by candidate vaccines in humans, and (iii) validation of a number of (bio)-assays as correlate for blocking activity in the field. Here we discuss clinical manufacturing and testing of current SSM-VIMT candidates and the latest bio-assay development for clinical evaluation. New testing strategies are discussed that may accelerate the evaluation and application of SSM-VIMT.


Asunto(s)
Vacunas contra la Malaria/inmunología , Malaria/prevención & control , Malaria/transmisión , Plasmodium vivax/inmunología , Animales , Anopheles/parasitología , Anticuerpos Antiprotozoarios/sangre , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Antígenos de Superficie/inmunología , Bioensayo , Ensayos Clínicos como Asunto , Erradicación de la Enfermedad , Humanos , Insectos Vectores/parasitología , Malaria/inmunología , Vacunas contra la Malaria/administración & dosificación , Malaria Falciparum/inmunología , Malaria Falciparum/prevención & control , Malaria Falciparum/transmisión , Malaria Vivax/prevención & control , Malaria Vivax/transmisión , Plasmodium falciparum/inmunología
8.
J Antimicrob Chemother ; 70(5): 1357-66, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25667405

RESUMEN

OBJECTIVES: The development of drugs to reduce malaria transmission is an important part of malaria eradication plans. We set out to develop and validate a combination of new screening assays for prioritization of transmission-blocking molecules. METHODS: We developed high-throughput assays for screening compounds against gametocytes, the parasite stages responsible for onward transmission to mosquitoes. An existing gametocyte parasitic lactate dehydrogenase (pLDH) assay was adapted for use in 384-well plates, and a novel homogeneous immunoassay to monitor the functional transition of female gametocytes into gametes was developed. A collection of 48 marketed and experimental antimalarials was screened and subsequently tested for impact on sporogony in Anopheles mosquitoes, to directly quantify the transmission-blocking properties of antimalarials in relation to their effects on gametocyte pLDH activity or gametogenesis. RESULTS AND CONCLUSIONS: The novel screening assays revealed distinct stage-specific kinetics and dynamics of drug effects. Peroxides showed the most potent transmission-blocking effects, with an intermediate speed of action and IC50 values that were 20-40-fold higher than the IC50s against the asexual stages causing clinical malaria. Finally, the novel synthetic peroxide OZ439 appeared to be a promising drug candidate as it exerted gametocytocidal and transmission-blocking effects at clinically relevant concentrations.


Asunto(s)
Antimaláricos/aislamiento & purificación , Evaluación Preclínica de Medicamentos/métodos , Plasmodium/efectos de los fármacos , Animales , Anopheles/parasitología , Supervivencia Celular/efectos de los fármacos , Femenino , Ensayos Analíticos de Alto Rendimiento/métodos , Concentración 50 Inhibidora , L-Lactato Deshidrogenasa/análisis , Plasmodium/enzimología
9.
Parasite Immunol ; 34(12): 562-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23171040

RESUMEN

Studies in animal models suggest that protection against malaria induced by intradermal (ID) administration of sporozoites is less effective compared to intravenous injection (IV). We investigated in a murine model the protective efficacy and immune responses after ID or IV immunization of sporozoites. Mice were immunized via either IV or ID route with Plasmodium berghei sporozoites in combination with chloroquine treatment (CPS) (allowing full liver stage development) or by γ-radiation-attenuated sporozoites (RAS) (early liver stage arrest). While IV immunization with both RAS and CPS generated 90-100% protection, ID immunization resulted in reduced levels of protection with either immunization strategy in both Balb/cByJ (50%) and C57BL/6j mice (7-13%). Lower protection by ID routing associated with a 30-fold lower parasite liver load [P < 0.001 (χ(2) = 49.08, d.f. = 1)] assessed by real-time in vivo imaging of bioluminescent P. berghei parasites. Unlike IV, ID immunization did not result in expansion of CD8+ T cells with effector memory phenotype and showed lower IFNγ responses irrespective of the immunization regime. In conclusion, protection against sporozoite infection is likely dependent on parasite liver infection and subsequently generated cellular immune responses.


Asunto(s)
Hígado/parasitología , Vacunas contra la Malaria/inmunología , Carga de Parásitos , Plasmodium berghei/inmunología , Esporozoítos/inmunología , Animales , Antimaláricos/administración & dosificación , Linfocitos T CD8-positivos/inmunología , Cloroquina/administración & dosificación , Modelos Animales de Enfermedad , Rayos gamma , Inmunización/métodos , Inyecciones Intradérmicas , Inyecciones Intravenosas , Interferón gamma/metabolismo , Malaria/inmunología , Malaria/prevención & control , Vacunas contra la Malaria/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Plasmodium berghei/patogenicidad , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología
10.
Antimicrob Agents Chemother ; 56(7): 3544-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22508309

RESUMEN

The global malaria agenda has undergone a reorientation from control of clinical cases to entirely eradicating malaria. For that purpose, a key objective is blocking transmission of malaria parasites from humans to mosquito vectors. The new antimalarial drug candidate NITD609 was evaluated for its transmission-reducing potential and compared to a few established antimalarials (lumefantrine, artemether, primaquine), using a suite of in vitro assays. By the use of a microscopic readout, NITD609 was found to inhibit the early and late development of Plasmodium falciparum gametocytes in vitro in a dose-dependent fashion over a range of 5 to 500 nM. In addition, using the standard membrane feeding assay, NITD609 was also found to be a very effective drug in reducing transmission to the Anopheles stephensi mosquito vector. Collectively, our data suggest a strong transmission-reducing effect of NITD609 acting against different P. falciparum transmission stages.


Asunto(s)
Anopheles/parasitología , Antimaláricos/farmacología , Gametogénesis/efectos de los fármacos , Indoles/farmacología , Insectos Vectores/parasitología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/patogenicidad , Compuestos de Espiro/farmacología , Animales
11.
Vaccine ; 27(42): 5719-25, 2009 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-19654061

RESUMEN

Development and optimization of first generation malaria vaccine candidates has been facilitated by the existence of a well-established Plasmodium falciparum clinical challenge model in which infectious sporozoites are administered to human subjects via mosquito bite. While ideal for testing pre-erythrocytic stage vaccines, some researchers believe that the sporozoite challenge model is less appropriate for testing blood stage vaccines. Here we report a consultation, co-sponsored by PATH MVI, USAID, EMVI and WHO, where scientists from all institutions globally that have conducted such clinical challenges in recent years and representatives from regulatory agencies and funding agencies met to discuss clinical malaria challenge models. Participants discussed strengthening and harmonizing the sporozoite challenge model and considered the pros and cons of further developing a blood stage challenge possibly better suited for evaluating the efficacy of blood stage vaccines. This report summarizes major findings and recommendations, including an update on the Plasmodium vivax clinical challenge model, the prospects for performing experimental challenge trials in malaria endemic countries and an update on clinical safety data. While the focus of the meeting was on the optimization of clinical challenge models for evaluation of blood stage candidate malaria vaccines, many of the considerations are relevant for the application of challenge trials to other purposes.


Asunto(s)
Ensayos Clínicos como Asunto , Vacunas contra la Malaria/administración & dosificación , Malaria/prevención & control , Animales , Humanos , Malaria/sangre , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium vivax/crecimiento & desarrollo
12.
Immunol Lett ; 122(2): 115-7, 2009 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-19100773

RESUMEN

Malaria is a major economic and public health problem in mainly sub-Saharan Africa. Globally 300-500 million new infections occur each year with 1-3 million fatal cases in particular young children. The most effective way to reduce disease and death from infectious diseases is to vaccinate susceptible populations. Increasing parasite resistance against available cheap anti-malarial drugs urges the need for development of malaria vaccines.


Asunto(s)
Vacunas contra la Malaria/inmunología , Malaria/inmunología , África , Animales , Investigación Biomédica , Humanos , Inmunidad , Malaria/epidemiología , Malaria/prevención & control , Modelos Animales , Prevalencia
13.
Parasite Immunol ; 30(3): 133-8, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18179626

RESUMEN

Macrophage migration inhibitory factor (MIF) has recently been implicated in the pathogenesis of malarial anaemia. However, field studies have reported contradictory results on circulating MIF concentrations in patients with clinically overt Plasmodium falciparum malaria. We determined plasma MIF levels over time in 10 healthy volunteers during experimental P. falciparum infection. Under fully controlled conditions, MIF levels decreased significantly during early blood-stage infection and reached a nadir at day 8 post-infection. A decrease in the number of circulating lymphocytes, which are an important source of MIF production, paralleled the decrease in MIF levels. Monocyte/macrophage counts remained unchanged. At MIF nadir, the anti-inflammatory cytokine interleukin (IL)-10, which is an inhibitor of T-cell MIF production, was detectable in only 2 of 10 volunteers. Plasma concentrations of the pro-inflammatory cytokines IL-8 and IL-1beta were only marginally elevated. We conclude that circulating MIF levels decrease early in blood-stage malaria as a result of the decline in circulating lymphocytes.


Asunto(s)
Linfocitos/sangre , Factores Inhibidores de la Migración de Macrófagos/sangre , Malaria Falciparum/parasitología , Plasmodium falciparum/inmunología , Adolescente , Adulto , Animales , Femenino , Humanos , Interleucina-10/sangre , Interleucina-1beta/sangre , Interleucina-8/sangre , Recuento de Linfocitos , Macrófagos/inmunología , Malaria Falciparum/inmunología , Masculino , Monocitos/inmunología , Factores de Tiempo
14.
Parasite Immunol ; 29(6): 309-17, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17518949

RESUMEN

Next to children, adults form a substantial part of the infectious reservoir that is responsible for the spread of malaria. In this longitudinal study, we determined sexual stage immune responses to Plasmodium falciparum and infectiousness to mosquitoes in adults from an area with intense malaria transmission. A cohort of 43 Tanzanian adults was followed for 18 months. Parasitological data were collected monthly; serum once every three months. Antibody prevalences were determined for sexual stage antigens Pfs230 and Pfs48/45 and circumsporozoite protein (NANP5)(n = 199). Functional transmission reducing activity (TRA) was assessed by standard membrane feeding assay (SMFA; n = 85). Cumulative parasite prevalence was 67.4% (29/43) for asexual stages and 34.9% (15/43) for gametocytes. Enrolment antibody prevalence was 95.3% (41/43) for NANP5, 18.9% (7/37) for Pfs230 and 7% (3/43) for Pfs48/45 epitope 3. TRA > 50% reduction was seen in 48.2% (41/85) and TRA > 90% reduction in 4.7% (4/85) of the samples. Our findings of low and inconsistent sexual stage immune responses are likely to be the result of a low exposure to gametocytes in this older age group. This may in turn be caused by effective asexual stage immunity. We conclude that the infectivity of older individuals is less likely to be affected by sexual stage immunity.


Asunto(s)
Anopheles/parasitología , Anticuerpos Antiprotozoarios/inmunología , Insectos Vectores/parasitología , Malaria Falciparum/inmunología , Parasitemia/inmunología , Plasmodium falciparum/inmunología , Adolescente , Adulto , Animales , Anticuerpos Antiprotozoarios/sangre , Estudios de Cohortes , Humanos , Modelos Logísticos , Estudios Longitudinales , Malaria Falciparum/epidemiología , Malaria Falciparum/parasitología , Malaria Falciparum/transmisión , Persona de Mediana Edad , Parasitemia/epidemiología , Parasitemia/transmisión , Población Rural , Estaciones del Año , Estudios Seroepidemiológicos , Tanzanía/epidemiología
15.
Antimicrob Agents Chemother ; 51(3): 1064-70, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17178799

RESUMEN

During asexual intraerythrocytic growth, Plasmodium falciparum utilizes hemoglobin obtained from the host red blood cell (RBC) as a nutrient source. Papain-like cysteine proteases, falcipains 2 and 3, have been reported to be involved in hemoglobin digestion and are targets of current antimalarial drug development efforts. However, their expression during gametocytogenesis, which is required for malaria parasite transmission, has not been studied. Many of the available antimalarials do not inhibit development of sexual stage parasites, and therefore, the persistence of gametocytes after drug treatment allows continued transmission of the disease. In the work reported here, incubation of stage V gametocytes with membrane-permeant cysteine protease inhibitor E64d significantly inhibited oocyst production (80 to 100%). The same conditions inhibited processing of gametocyte-surface antigen Pfs230 during gametogenesis but did not alter the morphology of the food vacuole in gametocytes, inhibit emergence, or block male exflagellation. E64d reduced the level of oocyst production more effectively than that reported previously for falcipain 1-knockout parasites, suggesting that falcipains 2 and 3 may also be involved in malaria parasite transmission. However, in this study only falcipain 3 and not falcipain 2 was found to be expressed in stage V gametocytes. Interestingly, during gametocytogenesis falcipain 3 was transported into the red blood cell and by stage V was localized in vesicles along the RBC surface, consistent with a role during gamete emergence. The ability of a membrane-permeant cysteine protease inhibitor to significantly reduce malaria parasite transmission suggests that future drug design should include evaluation of gametogenesis and sporogonic development.


Asunto(s)
Antimaláricos/farmacología , Inhibidores de Cisteína Proteinasa/farmacología , Leucina/análogos & derivados , Oocistos/efectos de los fármacos , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/crecimiento & desarrollo , Animales , Culicidae , Cisteína Endopeptidasas/biosíntesis , Eritrocitos/parasitología , Técnica del Anticuerpo Fluorescente Indirecta , Gametogénesis/efectos de los fármacos , Humanos , Immunoblotting , Técnicas In Vitro , Leucina/farmacología , Plasmodium falciparum/metabolismo
16.
Parasite Immunol ; 28(5): 185-90, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16629703

RESUMEN

Immunity to the sexual stages of Plasmodium falciparum is induced during natural infections and can significantly reduce the transmission of parasites to mosquitoes (transmission reducing activity; TRA) but little is known about how these responses develop with increasing age/exposure to malaria. Routinely TRA is measured in the standard membrane feeding assay (SMFA). Sera were collected from a total of 199 gametocyte carriers (median age 4 years, quartiles 2 and 9 years) near Ifakara, Tanzania; 128 samples were tested in the SMFA and generated TRA data classified as a reduction of > 50% and > 90% of transmission. TRA of > 50% was highest in young children (aged 1-2) with a significant decline with age (chi(2) trend = 5.79, P = 0.016) and in logistic regression was associated with prevalence of antibodies to both Pfs230 and Pfs48/45 (OR 4.03, P = 0.011 and OR 2.43 P = 0.059, respectively). A TRA of > 90% reduction in transmission was not age related but was associated with antibodies to Pfs48/45 (OR 2.36, P = 0.055). Our data confirm that antibodies are an important component of naturally induced TRA. However, whilst a similar but small proportion of individuals at all ages have TRA > 90%, the gradual deterioration of TRA > 50% with age suggests decreased antibody concentration or affinity. This may be due to decreased exposure to gametocytes, probably as a result of increased asexual and/or gametocyte specific immunity.


Asunto(s)
Portador Sano/parasitología , Malaria Falciparum/inmunología , Malaria Falciparum/transmisión , Plasmodium falciparum/inmunología , Adolescente , Factores de Edad , Animales , Anopheles/parasitología , Anticuerpos Antiprotozoarios/sangre , Portador Sano/inmunología , Portador Sano/transmisión , Niño , Preescolar , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Lactante , Modelos Lineales , Malaria Falciparum/sangre , Malaria Falciparum/parasitología , Masculino , Plasmodium falciparum/crecimiento & desarrollo
17.
Curr Mol Med ; 6(2): 223-9, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16515512

RESUMEN

Gametocytes and sporogonic stages are responsible for the spread of disease and drug resistance in the population. Sexual stage immunity affects the infectiousness of gametocytes to mosquitoes. Specific antibodies including anti-Pfs48/45 and anti-Pfs230 antibodies are found in individuals with limited prior exposure to malaria. Sexual stage antibodies are rapidly acquired after infection and are relatively prevalent in gametocytaemic individuals. Functional transmission reducing activity (TRA) is found after primary infections and in young children and appears to depend on recent rather than cumulative exposure to gametocytes. Exposure to gametocytes decreases with age most likely as a consequence of the acquisition of asexual-stage immunity that controls asexual parasite density and consequently gametocytaemia. This results in lower exposure to the antigenic load of gametocytes in semi-immune individuals. Since sexual stage immunity is probably short-lived in the absence of gametocytes, we hypothesize that sexual stage immunity will wane, resulting in low antibody and TRA prevalences in clinically semi-immune carriers.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Formación de Anticuerpos , Estadios del Ciclo de Vida , Malaria Falciparum/inmunología , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/inmunología , Animales , Enfermedades Endémicas , Humanos , Malaria Falciparum/epidemiología , Malaria Falciparum/transmisión , Plasmodium falciparum/patogenicidad , Apoyo a la Investigación como Asunto
18.
Neth J Med ; 63(2): 52-8, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15768480

RESUMEN

BACKGROUND: Human experimental malaria infections have been safely carried out previously. The objective of this study was to evaluate infection rates and clinical safety of different protocols for human experimental malaria induced by Plasmodium falciparum-infected mosquitoes. METHODS: Thirty nonimmune volunteers were infected by bites of 1-2 or 4-7 Anopheles stephensi mosquitoes infected with the NF54 strain of P. falciparum. RESULTS: A 100 or 50% infection rate was obtained after bites of 4-7 and 1-2 infected mosquitoes, respectively. Median prepatent period was 8.8 days. The most common symptoms after a median incubation time of eight days were headache, malaise/fatigue and fever. There was no significant difference in clinical and parasitological presentation between groups infected by 4-7 or 1-2 mosquitoes. Delay of treatment by maximally 48 hours after the first positive thick smear was generally well tolerated but fever was higher and more frequently observed. The most prominent laboratory abnormality was uncomplicated thrombocytopenia. Two volunteers with parasitaemia developed psychiatric side effects after chloroquine treatment. CONCLUSION: With stringent inclusion criteria, close monitoring and immediate administration of treatment upon detection of parasitaemia, experimental human malaria challenges can be considered safe and generally well tolerated.


Asunto(s)
Antimaláricos/uso terapéutico , Experimentación Humana , Malaria Falciparum/fisiopatología , Plasmodium falciparum/patogenicidad , Adolescente , Adulto , Animales , Análisis Químico de la Sangre , Culicidae , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Humanos , Malaria Falciparum/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Cooperación del Paciente , Probabilidad , Proyectos de Investigación , Medición de Riesgo , Sensibilidad y Especificidad , Índice de Severidad de la Enfermedad , Estadísticas no Paramétricas
19.
Parasitology ; 130(Pt 1): 13-22, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15700753

RESUMEN

Host responses to the transmittable stages of the malaria parasite may reduce transmission effectively. Transmission-reducing activity (TRA) of human serum can be determined as a percentage, using the Standard Membrane Feeding Assay (SMFA). This laboratory assay was evaluated using the results of 121 experiments with malaria-endemic sera among which many repeated measurements were obtained. The assay consists of the feeding of Anopheles stephensi mosquitoes with cultured Plasmodium falciparum gametocytes, mixed with human red blood cells, and control and experimental sera. The TRA of individual sera was determined by the comparison of oocyst densities between these sera. Bootstrap data on oocyst densities in individual mosquitoes in control feeds were used to construct confidence limits for TRA percentages of serum feeds. Low (<20%) and high TRA (>90%) values for individual sera were usually reproduced in a second experiment, whereas this was more difficult for values between 20% and 90%. The observed variability of TRA values is explained in part by the variability in oocyst density per mosquito. Oocyst densities in control feeds varied more between experiments than within experiments and showed a slight decline over the 3 years of experiments. Reproducibility of TRA of field sera was low (20%) between experiments, but much higher (61 %) within experiments. A minimum of 35 oocysts per mosquito in control feeds gave optimal reproducibility (44%) between experiments. We recommend that (1) sera are compared within an experiment, or (2) assays are only analysed where controls have at least 35 oocysts per mosquito. The SMFA is under the recommended conditions appropriate for the study of factors that may influence TRA, e.g. transmission blocking vaccines.


Asunto(s)
Anopheles/parasitología , Conducta Alimentaria/fisiología , Malaria Falciparum/sangre , Malaria Falciparum/transmisión , Membranas Artificiales , Animales , Anopheles/fisiología , Simulación por Computador , Humanos , Insectos Vectores , Malaria Falciparum/prevención & control , Modelos Biológicos , Oocistos , Plasmodium falciparum/crecimiento & desarrollo , Reproducibilidad de los Resultados , Estudios Retrospectivos
20.
Clin Exp Immunol ; 132(3): 467-72, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12780694

RESUMEN

Release of soluble Granzymes (sGranzymes) is considered to reflect activation of cytotoxic T lymphocytes and NK cells. sGranzymes and a number of pro-inflammatory cytokines were measured in plasma of malaria patients with natural or experimentally induced Plasmodium falciparum infections. Concentrations of sGranzyme A and B, IL-10, IL-12p70 and CRP were significantly increased in African children presenting with clinical malaria; IL-10 and CRP concentrations were significantly correlated with disease severity. In nonimmune Dutch volunteers which were experimentally infected by P. falciparum-infected mosquitoes, sGranzyme A increment started 1-2 days prior to clinical symptoms and microscopically detectable parasitaemia. This coincided with increases in IFNgamma, IL-12p40 and IL-8, while sGranzyme B and IL-10 levels increased 24-48 h later. The elevation of sGranzyme A and IFNgamma in nonimmune volunteers suggests that NK cells are activated upon release of parasites by infected liver cells and subsequently during blood stage infection; thus, NK cells are likely involved innate immune human host resistance in the early phase of a malaria infection.


Asunto(s)
Malaria Falciparum/enzimología , Serina Endopeptidasas/sangre , Adolescente , Proteína C-Reactiva/análisis , Niño , Preescolar , Granzimas , Humanos , Lactante , Interferón gamma/sangre , Interleucina-10/sangre , Interleucina-12/sangre , Células Asesinas Naturales/inmunología , Malaria Falciparum/inmunología , Parasitemia/enzimología , Parasitemia/inmunología , Solubilidad , Linfocitos T Citotóxicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...