Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Clin Oncol ; 42(9): 1067-1076, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-37963313

RESUMEN

PURPOSE: GemPred, a transcriptomic signature predictive of the efficacy of adjuvant gemcitabine (GEM), was developed from cell lines and organoids and validated retrospectively. The phase III PRODIGE-24/CCTG PA6 trial has demonstrated the superiority of modified folinic acid, fluorouracil, irinotecan, and oxaliplatin (mFOLFIRINOX) over GEM as adjuvant therapy in patients with resected pancreatic ductal adenocarcinoma at the expense of higher toxicity. We evaluated the potential predictive value of GemPred in this population. PATIENTS AND METHODS: Routine formalin-fixed paraffin-embedded surgical specimens of 350 patients were retrieved for RNA sequencing and GemPred prediction (167 in the GEM arm and 183 in the mFOLFIRINOX [mFFX] arm). Survival analyses were stratified by resection margins, lymph node status, and cancer antigen 19-9 level. RESULTS: Eighty-nine patients' tumors (25.5%) were GemPred+ and were thus predicted to be gemcitabine-sensitive. In the GEM arm, GemPred+ patients (n = 50, 30%) had a significantly longer disease-free survival (DFS) than GemPred- patients (n = 117, 70%; median 27.3 v 10.2 months, hazard ratio [HR], 0.43 [95% CI, 0.29 to 0.65]; P < .001) and cancer-specific survival (CSS; median 68.4 v 28.6 months, HR, 0.42 [95% CI, 0.27 to 0.66]; P < .001). GemPred had no prognostic value in the mFFX arm. DFS and CSS were similar in GemPred+ patients who received adjuvant GEM and mFFX (median 27.3 v 24.0 months, and 68.4 v 51.4 months, respectively). The statistical interaction between GEM and GemPred+ status was significant for DFS (P = .008) and CSS (P = .004). GemPred+ patients had significantly more adverse events of grade ≥3 in the mFFX arm (76%) compared with those in the GEM arm (40%; P = .001). CONCLUSION: This ancillary study of a phase III randomized trial demonstrates that among the quarter of patients with a GemPred-positive transcriptomic signature, survival was comparable with that of mFOLFIRINOX, whereas those receiving adjuvant gemcitabine had fewer adverse events.


Asunto(s)
Adenocarcinoma , Neoplasias Pancreáticas , Humanos , Gemcitabina , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/cirugía , Desoxicitidina/efectos adversos , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/cirugía , Estudios Retrospectivos , Fluorouracilo/efectos adversos , Adyuvantes Inmunológicos/uso terapéutico , ARN/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos
2.
JTO Clin Res Rep ; 1(3): 100068, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34589950

RESUMEN

INTRODUCTION: Molecular profiling is considered a standard of care in advanced NSCLC. A comprehensive next-generation sequencing panel can discover somatic or germline BRCA1/2 mutations that are new druggable molecular alterations. However, the phenotypic and potential therapeutic relevance of BRCA1/2 mutation in NSCLC remains poorly defined. METHODS: From April 2014 to March 2017, 600 newly diagnosed, EGFR/ALK negative patients with advanced NSCLC were enrolled in the SAFIR02-Lung trial. Molecular profiling was done at study entry on archival tissue or frozen tissue collected from a new biopsy specimen before the third cycle of platinum-based chemotherapy. The prevalence of BRCA1/2 variants and its biological relevance were assessed. A homologous recombinant deficiency (HRD) score was based on the copy number variation data, and the germline status was determined by blood analysis. The BRCA Share database and the French CGG consortium were the references for the variant classification. RESULTS: Of 379 patients with a molecular profile discussed in a tumor molecular board, BRCA1/2 variants were identified in 20 patients (5.3%), including eight patients (2.1%) with a confirmed pathogenic BRCA mutation. Two patients (0.5%) harbored a germline BRCA2 mutation, and for six others, a somatic BRCA mutation was identified (1.6%). All were men and mainly smokers (88%). The overall response rate to chemotherapy was 13%. BRCA variants of unknown significance were detected in 12 patients (3.2%), achieving an 8.3% overall response rate with chemotherapy. One-third of tumors carrying pathogenic BRCA mutations or variants of unknown significance had biallelic inactivation and high HRD score. Overall survival of this cohort was 12.8 months. CONCLUSIONS: Pathogenic BRCA1/2 mutations occur in 2.1% of patients with advanced NSCLC. The predictive role of BRCA mutation for making treatment decisions in NSCLC seems limited based on clinical response (low platinum sensitivity) and molecular features (discrepancy between biallelic inactivation and high HRD score).

3.
Cell Rep ; 10(7): 1055-65, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25704810

RESUMEN

The innate immune system senses cytosolic dsDNA and bacterial cyclic dinucleotides and initiates signaling via the adaptor STING to induce type 1 interferon (IFN) response. We demonstrate here that BTK-deficient cells have impaired IFN-ß production and TBK1/IRF3 activation when stimulated with agonists or infected with pathogens that activate STING signaling. BTK interacts with STING and DDX41 helicase. The kinase and SH3/SH2 interaction domains of BTK bind, respectively, the DEAD-box domain of DDX41 and transmembrane region of STING. BTK phosphorylates DDX41, and its kinase activities are critical for STING-mediated IFN-ß production. We show that Tyr364 and Tyr414 of DDX41 are critical for its recognition of AT-rich DNA and binding to STING, and tandem mass spectrometry identifies Tyr414 as the BTK phosphorylation site. Modeling studies further indicate that phospho-Tyr414 strengthens DDX41's interaction with STING. Hence, BTK plays a critical role in the activation of DDX41 helicase and STING signaling.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , ADN/metabolismo , Interferón beta/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Agammaglobulinemia Tirosina Quinasa , Animales , Sitios de Unión , Línea Celular , ARN Helicasas DEAD-box/química , ARN Helicasas DEAD-box/genética , ADN/química , Células HEK293 , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/genética , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Simulación de Dinámica Molecular , Parasitemia/mortalidad , Parasitemia/patología , Parasitemia/veterinaria , Fosfopéptidos/análisis , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/deficiencia , Proteínas Tirosina Quinasas/genética , Transducción de Señal , Tasa de Supervivencia
4.
PLoS Negl Trop Dis ; 8(7): e3043, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25078474

RESUMEN

Tropical pathogens often cause febrile illnesses in humans and are responsible for considerable morbidity and mortality. The similarities in clinical symptoms provoked by these pathogens make diagnosis difficult. Thus, early, rapid and accurate diagnosis will be crucial in patient management and in the control of these diseases. In this study, a microfluidic lab-on-chip integrating multiplex molecular amplification and DNA microarray hybridization was developed for simultaneous detection and species differentiation of 26 globally important tropical pathogens. The analytical performance of the lab-on-chip for each pathogen ranged from 102 to 103 DNA or RNA copies. Assay performance was further verified with human whole blood spiked with Plasmodium falciparum and Chikungunya virus that yielded a range of detection from 200 to 4×105 parasites, and from 250 to 4×107 PFU respectively. This lab-on-chip was subsequently assessed and evaluated using 170 retrospective patient specimens in Singapore and Thailand. The lab-on-chip had a detection sensitivity of 83.1% and a specificity of 100% for P. falciparum; a sensitivity of 91.3% and a specificity of 99.3% for P. vivax; a positive 90.0% agreement and a specificity of 100% for Chikungunya virus; and a positive 85.0% agreement and a specificity of 100% for Dengue virus serotype 3 with reference methods conducted on the samples. Results suggested the practicality of an amplification microarray-based approach in a field setting for high-throughput detection and identification of tropical pathogens.


Asunto(s)
Enfermedades Transmisibles/diagnóstico , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas/métodos , Técnicas de Diagnóstico Molecular/métodos , Medicina Tropical/métodos , Humanos , Técnicas Analíticas Microfluídicas/instrumentación , Técnicas de Diagnóstico Molecular/instrumentación , Sensibilidad y Especificidad , Singapur , Tailandia , Medicina Tropical/instrumentación
5.
Blood ; 123(18): e100-9, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24652986

RESUMEN

Rosetting phenomenon has been linked to malaria pathogenesis. Although rosetting occurs in all causes of human malaria, most data on this subject has been derived from Plasmodium falciparum. Here, we investigate the function and factors affecting rosette formation in Plasmodium vivax. To achieve this, we used a range of novel ex vivo protocols to study fresh and cryopreserved P vivax (n = 135) and P falciparum (n = 77) isolates from Thailand. Rosetting is more common in vivax than falciparum malaria, both in terms of incidence in patient samples and percentage of infected erythrocytes forming rosettes. Rosetting to P vivax asexual and sexual stages was evident 20 hours postreticulocyte invasion, reaching a plateau after 30 hours. Host ABO blood group, reticulocyte count, and parasitemia were not correlated with P vivax rosetting. Importantly, mature erythrocytes (normocytes), rather than reticulocytes, preferentially form rosetting complexes, indicating that this process is unlikely to directly facilitate merozoite invasion. Although antibodies against host erythrocyte receptors CD235a and CD35 had no effect, Ag-binding fragment against the BRIC 4 region of CD236R significantly inhibited rosette formation. Rosetting assays using CD236R knockdown normocytes derived from hematopoietic stem cells further supports the role of glycophorin C as a receptor in P vivax rosette formation.


Asunto(s)
Eritrocitos/metabolismo , Eritrocitos/parasitología , Glicoforinas/metabolismo , Malaria Vivax/metabolismo , Plasmodium vivax/inmunología , Formación de Roseta/métodos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Criopreservación/métodos , Eritrocitos/patología , Técnicas de Silenciamiento del Gen , Glicoforinas/genética , Glicoforinas/inmunología , Humanos , Malaria Vivax/diagnóstico , Malaria Vivax/parasitología , Plasmodium vivax/aislamiento & purificación , Receptores de Complemento 3b/antagonistas & inhibidores , Flujo de Trabajo
6.
Methods Mol Biol ; 923: 567-76, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22990805

RESUMEN

Sterile immunity against malaria has been obtained in mammalian hosts exclusively through vaccination with whole parasite preparations. Induction of complete protection against challenge was obtained using sporozoites attenuated by irradiation or genetic manipulations. It has been demonstrated recently that immunization with normal sporozoites under chloroquine cover confers sterile protection in mice and humans, using substantially fewer parasites and injections than with irradiated sporozoite immunization. Subsequently, it was shown that other drugs can substitute for chloroquine. We describe the immunization protocol using live sporozoites under chloroquine cover, which confers sterile immunity in rodents.


Asunto(s)
Antimaláricos/uso terapéutico , Vacunas contra la Malaria/inmunología , Malaria/terapia , Plasmodium/inmunología , Esporozoítos/inmunología , Animales , Antimaláricos/administración & dosificación , Cloroquina/administración & dosificación , Cloroquina/uso terapéutico , Culicidae/parasitología , Eritrocitos/parasitología , Hígado/inmunología , Hígado/parasitología , Malaria/tratamiento farmacológico , Malaria/prevención & control , Vacunas contra la Malaria/administración & dosificación , Ratones , Plasmodium/crecimiento & desarrollo
7.
Immunol Res ; 53(1-3): 115-26, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22418726

RESUMEN

Malaria remains one of the main infectious diseases in intertropical regions. The malaria parasite has a complex life cycle in its mammalian host, switching between variable forms as it traverses through different tissues and anatomic locations, either intra- or intercellularly. During its journey, the parasite encounters and interacts with the host immune system, which functions to prevent infections and limit ensuing pathologies. One important component of the host immune system is the dendritic cells (DC) network. DC form a heterogeneous group of pathogen-sensing and antigen-presenting cells that play a crucial role in the initiation of adaptive immunity. Here, we review the known and unknown interactions between the malaria parasites and the DC system, starting from the inoculation of the parasite in the skin up to its exit from the liver, also known as the pre-erythrocytic stage of the infection, and discuss how deciphering these interactions may contribute to our understanding of the Plasmodium parasite biology as well as to the induction of immune protection via vaccination.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/parasitología , Vacunas contra la Malaria/inmunología , Malaria/inmunología , Plasmodium/inmunología , Inmunidad Adaptativa , Animales , Eritrocitos/inmunología , Humanos , Inmunoterapia , Malaria/prevención & control , Vacunas contra la Malaria/administración & dosificación , Terapia Molecular Dirigida
8.
PLoS One ; 6(3): e17464, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21394207

RESUMEN

BACKGROUND: The liver stages of malaria parasites are inhibited by cytokines such as interferon-γ or Interleukin (IL)-6. Binding of these cytokines to their receptors at the surface of the infected hepatocytes leads to the production of nitric oxide (NO) and radical oxygen intermediates (ROI), which kill hepatic parasites. However, conflicting results were obtained with TNF-α possibly because of differences in the models used. We have reassessed the role of TNF-α in the different cellular systems used to study the Plasmodium pre-erythrocytic stages. METHODS AND FINDINGS: Human or mouse TNF-α were tested against human and rodent malaria parasites grown in vitro in human or rodent primary hepatocytes, or in hepatoma cell lines. Our data demonstrated that TNF-α treatment prevents the development of malaria pre-erythrocytic stages. This inhibitory effect however varies with the infecting parasite species and with the nature and origin of the cytokine and hepatocytes. Inhibition was only observed for all parasite species tested when hepatocytes were pre-incubated 24 or 48 hrs before infection and activity was directed only against early hepatic parasite. We further showed that TNF-α inhibition was mediated by a soluble factor present in the supernatant of TNF-α stimulated hepatocytes but it was not related to NO or ROI. Treatment TNF-α prevents the development of human and rodent malaria pre-erythrocytic stages through the activity of a mediator that remains to be identified. CONCLUSIONS: Treatment TNF-α prevents the development of human and rodent malaria pre-erythrocytic stages through the activity of a mediator that remains to be identified. However, the nature of the cytokine-host cell-parasite combination must be carefully considered for extrapolation to the human infection.


Asunto(s)
Hepatocitos/parasitología , Interacciones Huésped-Parásitos/efectos de los fármacos , Estadios del Ciclo de Vida/efectos de los fármacos , Malaria/parasitología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/crecimiento & desarrollo , Factor de Necrosis Tumoral alfa/farmacología , Animales , Antígenos CD/metabolismo , Línea Celular Tumoral , Eritrocitos/efectos de los fármacos , Eritrocitos/parasitología , Hepatocitos/efectos de los fármacos , Humanos , Ratones , Óxido Nítrico/farmacología , Especies Reactivas de Oxígeno/farmacología , Solubilidad/efectos de los fármacos , Tetraspanina 28
9.
Infect Immun ; 78(5): 2182-8, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20194600

RESUMEN

Immunization with live Plasmodium sporozoites under chloroquine prophylaxis (Spz plus CQ) induces sterile immunity against sporozoite challenge in rodents and, more importantly, in humans. Full protection is obtained with substantially fewer parasites than with the classic immunization with radiation-attenuated sporozoites. The sterile protection observed comprised a massive reduction in the hepatic parasite load and an additional effect at the blood stage level. Differences in the immune responses induced by the two protocols occur but are as yet little characterized. We have previously demonstrated that in mice immunized with irradiated sporozoites, immune responses against the circumsporozoite protein (CSP), the major component of the sporozoite's surface and the leading malaria vaccine candidate, were not essential for sterile protection. Here, we have employed transgenic Plasmodium berghei parasites in which the endogenous CSP was replaced by that of Plasmodium yoelii, another rodent malaria species, to assess the role of CSP in the sterile protection induced by the Spz-plus-CQ protocol. The data demonstrated that this role was minor because sterile immunity was obtained irrespective of the origin of CSP expressed by the parasites in this model of protection. The immunity was obtained through a single transient exposure of the host to the immunizing parasites (preerythrocytic and erythrocytic), a dose much smaller than that required for immunization with radiation-attenuated sporozoites.


Asunto(s)
Vacunas contra la Malaria/inmunología , Malaria/prevención & control , Plasmodium berghei/inmunología , Plasmodium yoelii/inmunología , Proteínas Protozoarias/inmunología , Animales , Femenino , Interferón gamma/metabolismo , Leucocitos Mononucleares/inmunología , Hígado/parasitología , Ratones , Ratones Endogámicos BALB C , Plasmodium berghei/genética , Plasmodium yoelii/genética , Bazo/inmunología , Esporozoítos/inmunología
10.
PLoS One ; 4(11): e7717, 2009 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-19890387

RESUMEN

Immunization with irradiated Plasmodium sporozoites induces sterile immunity in rodents, monkeys and humans. The major surface component of the sporozoite the circumsporozoite protein (CS) long considered as the antigen predominantly responsible for this immunity, thus remains the leading candidate antigen for vaccines targeting the parasite's pre-erythrocytic (PE) stages. However, this role for CS was questioned when we recently showed that immunization with irradiated sporozoites (IrrSpz) of a P. berghei line whose endogenous CS was replaced by that of P. falciparum still conferred sterile protection against challenge with wild type P. berghei sporozoites. In order to investigate the involvement of CS in the cross-species protection recently observed between the two rodent parasites P. berghei and P. yoelii, we adopted our gene replacement approach for the P. yoelii CS and exploited the ability to conduct reciprocal challenges. Overall, we found that immunization led to sterile immunity irrespective of the origin of the CS in the immunizing or challenge sporozoites. However, for some combinations, immune responses to CS contributed to the acquisition of protective immunity and were dependent on the immunizing IrrSpz dose. Nonetheless, when data from all the cross-species immunization/challenges were considered, the immune responses directed against non-CS parasite antigens shared by the two parasite species played a major role in the sterile protection induced by immunization with IrrSpz. This opens the perspective to develop a single vaccine formulation that could protect against multiple parasite species.


Asunto(s)
Vacunas contra la Malaria/inmunología , Plasmodium berghei/inmunología , Plasmodium yoelii/inmunología , Proteínas Protozoarias/fisiología , Animales , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Femenino , Sistema Inmunológico , Inmunoglobulina G/metabolismo , Inmunoglobulina M/metabolismo , Malaria/inmunología , Ratones , Ratones Endogámicos BALB C , Plasmodium berghei/metabolismo , Plasmodium yoelii/metabolismo , Proteínas Protozoarias/inmunología , Esporozoítos/inmunología
11.
Blood ; 114(27): 5522-31, 2009 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-19837977

RESUMEN

Host responses controlling blood-stage malaria include both innate and acquired immune effector mechanisms. During Plasmodium chabaudi infection in mice, a population of CD11b(high)Ly6C(+) monocytes are generated in bone marrow, most of which depend on the chemokine receptor CCR2 for migration from bone marrow to the spleen. In the absence of this receptor mice harbor higher parasitemias. Most importantly, splenic CD11b(high)Ly6C(+) cells from P chabaudi-infected wild-type mice significantly reduce acute-stage parasitemia in CCR2(-/-) mice. The CD11b(high)Ly6C(+) cells in this malaria infection display effector functions such as production of inducible nitric oxide synthase and reactive oxygen intermediates, and phagocytose P chabaudi parasites in vitro, and in a proportion of the cells, in vivo in the spleen, suggesting possible mechanisms of parasite killing. In contrast to monocyte-derived dendritic cells, CD11b(high)Ly6C(+) cells isolated from malaria-infected mice express low levels of major histocompatibility complex II and have limited ability to present the P chabaudi antigen, merozoite surface protein-1, to specific T-cell receptor transgenic CD4 T cells and fail to activate these T cells. We propose that these monocytes, which are rapidly produced in the bone marrow as part of the early defense mechanism against invading pathogens, are important for controlling blood-stage malaria parasites.


Asunto(s)
Movimiento Celular/fisiología , Monocitos/parasitología , Plasmodium chabaudi/fisiología , Bazo/parasitología , Animales , Células Presentadoras de Antígenos/metabolismo , Células Presentadoras de Antígenos/parasitología , Células Presentadoras de Antígenos/patología , Antígenos Ly/metabolismo , Antígeno CD11b/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/parasitología , Linfocitos T CD4-Positivos/patología , Citometría de Flujo , Interacciones Huésped-Parásitos , Malaria/sangre , Malaria/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Monocitos/patología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Parasitemia/metabolismo , Fagocitosis/fisiología , Receptores CCR2/genética , Receptores CCR2/metabolismo , Bazo/metabolismo , Bazo/patología , Linfocitos T/metabolismo , Linfocitos T/parasitología , Linfocitos T/patología , Factor de Necrosis Tumoral alfa/metabolismo
12.
J Immunol ; 181(12): 8552-8, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19050274

RESUMEN

Immunity to malaria has long been thought to be stage-specific. In this study we show that immunization of BALB/c mice with live erythrocytes infected with nonlethal strains of Plasmodium yoelii under curative chloroquine cover conferred protection not only against challenge by blood stage parasites but also against sporozoite challenge. This cross-stage protection was dose-dependent and long lasting. CD4(+) and CD8(+) T cells inhibited malaria liver but not blood stage. Their effect was mediated partially by IFN-gamma, and was completely dependent of NO. Abs against both pre-erythrocytic and blood parasites were elicited and were essential for protection against blood stage and liver stage parasites. Our results suggest that Ags shared by liver and blood stage parasites can be the foundation for a malaria vaccine that would provide effective protection against both pre-erythrocytic and erythrocytic asexual parasites found in the mammalian host.


Asunto(s)
Antimaláricos/administración & dosificación , Cloroquina/administración & dosificación , Eritrocitos/inmunología , Eritrocitos/parasitología , Parasitosis Hepáticas/prevención & control , Malaria/prevención & control , Plasmodium yoelii/crecimiento & desarrollo , Plasmodium yoelii/inmunología , Animales , Transfusión de Eritrocitos , Eritrocitos/efectos de los fármacos , Femenino , Inmunidad Innata/efectos de los fármacos , Parasitosis Hepáticas/tratamiento farmacológico , Parasitosis Hepáticas/inmunología , Malaria/sangre , Malaria/tratamiento farmacológico , Malaria/inmunología , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/inmunología , Ratones , Ratones Endogámicos BALB C , Plasmodium yoelii/efectos de los fármacos , Esporozoítos/efectos de los fármacos , Esporozoítos/crecimiento & desarrollo , Esporozoítos/inmunología
13.
PLoS One ; 2(12): e1371, 2007 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-18159254

RESUMEN

BACKGROUND: Research aimed at developing vaccines against infectious diseases generally seeks to induce robust immune responses to immunodominant antigens. This approach has led to a number of efficient bacterial and viral vaccines, but it has yet to do so for parasitic pathogens. For malaria, a disease of global importance due to infection by Plasmodium protozoa, immunization with radiation-attenuated sporozoites uniquely leads to long lasting sterile immunity against infection. The circumsporozoite protein (CSP), an important component of the sporozoite's surface, remains the leading candidate antigen for vaccines targeting the parasite's pre-erythrocytic stages. Difficulties in developing CSP-based vaccines that reproduce the levels of protection afforded by radiation-attenuated sporozoites have led us to question the role of CSP in the acquisition of sterile immunity. We have used a parasite transgenic for the CSP because it allowed us to test whether a major immunodominant Plasmodium antigen is indeed needed for the induction of sterile protective immunity against infection. METHODOLOGY/MAIN FINDINGS: We employed a P. berghei parasite line that expresses a heterologous CSP from P. falciparum in order to assess the role of the CSP in the protection conferred by vaccination with radiation-attenuated P. berghei parasites. Our data demonstrated that sterile immunity could be obtained despite the absence of immune responses specific to the CSP expressed by the parasite used for challenge. CONCLUSIONS: We conclude that other pre-erythrocytic parasite antigens, possibly hitherto uncharacterised, can be targeted to induce sterile immunity against malaria. From a broader perspective, our results raise the question as to whether immunodominant parasite antigens should be the favoured targets for vaccine development.


Asunto(s)
Malaria Falciparum/inmunología , Proteínas Protozoarias/inmunología , Secuencia de Aminoácidos , Animales , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Proteínas Protozoarias/química , Homología de Secuencia de Aminoácido
14.
J Immunol ; 178(10): 6416-25, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17475871

RESUMEN

Most C57BL/6 mice infected i.p. with Plasmodium berghei ANKA (PbA) die between 7 and 14 days with neurologic signs, and the remainder die later (>15 days) with severe anemia. Daily i.p. injections of a recombinant human IFN-alpha (active on mouse cells) prevented death by cerebral malaria (87% deaths in the control mice vs 6% in IFN-alpha-treated mice). The mechanisms of this IFN-alpha protective effect were multiple. IFN-alpha-treated, PbA-infected mice showed 1) a marked decrease in the number of PbA parasites in the blood mediated by IFN-gamma, 2) less sequestered parasites in cerebral vessels, 3) reduced up-regulation of ICAM-1 expression in brain endothelial cells, 4) milder rise of blood levels of TNF, 5) increased levels of IFN-gamma in the blood resulting from an increased production by splenic CD8+ T cells, and 6) fewer leukocytes (especially CD8+ T cells) sequestered in cerebral vessels. On the other hand, IFN-alpha treatment did not affect the marked anemia observed in PbA-infected mice. Survival time in IFN-alpha-treated mice was further increased by performing three blood transfusions over consecutive days.


Asunto(s)
Interferón Tipo I/administración & dosificación , Malaria Cerebral/inmunología , Malaria Cerebral/prevención & control , Parasitemia/tratamiento farmacológico , Anemia/inmunología , Anemia/parasitología , Anemia/patología , Animales , Femenino , Humanos , Inyecciones Intraperitoneales , Interferón Tipo I/uso terapéutico , Malaria Cerebral/parasitología , Malaria Cerebral/patología , Ratones , Ratones Endogámicos C57BL , Parasitemia/inmunología , Parasitemia/patología , Plasmodium berghei/efectos de los fármacos , Plasmodium berghei/inmunología , Proteínas Recombinantes
15.
Expert Rev Vaccines ; 5(4): 473-81, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16989628

RESUMEN

Despite nearly 80 years of vaccine research and control efforts, malaria remains one of the most prevalent of all infectious diseases. The fact that people living in regions in which malaria is endemic eventually develop immunity to the parasite and the disease suggest that it might be possible to develop vaccines against malaria. Although few vaccination trials were conducted with whole parasites, the only protocol that leads to the induction of sterile immunity in humans relies on immunization with attenuated parasites. This observation has spurred the search for subunit vaccines that aim to reproduce this protection. As yet, none of the current candidate subunit vaccines have achieved complete protection reproducibly. This failure, coupled with the recent advent of the genetically modified Plasmodium parasites, has led to a renewed interest in the use of live parasites for vaccination. This article reviews past studies, summarizes recent developments in this field and discusses the challenges to be overcome before mass immunization with live parasites could be envisaged.


Asunto(s)
Vacunas contra la Malaria , Malaria/inmunología , Plasmodium/inmunología , Esporozoítos/inmunología , Animales , Humanos
16.
Int J Parasitol ; 36(5): 547-54, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16600241

RESUMEN

Malaria remains a major global health problem and cerebral malaria (CM) is one of the most serious complications of this disease. Recent years have seen important advances in our understanding of the pathogenesis of cerebral malaria. Parasite sequestration, a hallmark of this syndrome, is thought to be solely responsible for the pathological process. However, this phenomenon cannot explain all aspects of the pathogenesis of CM. The use of an animal model, Plasmodium berghei ANKA in mice, has allowed the identification of specific pathological components of CM. Although multiple pathways may lead to CM, an important role for CD8+ T cells has been clarified. Other cells, including platelets, and mediators such as cytokines also have an important role. In this review we have focused on the role of T cells, and discuss what remains to be studied to understand the pathways by which these cells mediate CM.


Asunto(s)
Malaria Cerebral/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Encéfalo/inmunología , Quimiotaxis de Leucocito , Modelos Animales de Enfermedad , Interacciones Huésped-Parásitos , Ratones , Plasmodium berghei/patogenicidad , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...