Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Infect Dis ; 71(6): 1481-1490, 2020 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-31621832

RESUMEN

BACKGROUND: Chemoprophylaxis vaccination with sporozoites (CVac) with chloroquine induces protection against a homologous Plasmodium falciparum sporozoite (PfSPZ) challenge, but whether blood-stage parasite exposure is required for protection remains unclear. Chloroquine suppresses and clears blood-stage parasitemia, while other antimalarial drugs, such as primaquine, act against liver-stage parasites. Here, we evaluated CVac regimens using primaquine and/or chloroquine as the partner drug to discern whether blood-stage parasite exposure impacts protection against homologous controlled human malaria infection. METHODS: In a Phase I, randomized, partial double-blind, placebo-controlled study of 36 malaria-naive adults, all CVac subjects received chloroquine prophylaxis and bites from 12-15 P. falciparum-infected mosquitoes (CVac-chloroquine arm) at 3 monthly iterations, and some received postexposure primaquine (CVac-primaquine/chloroquine arm). Drug control subjects received primaquine, chloroquine, and uninfected mosquito bites. After a chloroquine washout, subjects, including treatment-naive infectivity controls, underwent homologous, PfSPZ controlled human malaria infection and were monitored for parasitemia for 21 days. RESULTS: No serious adverse events occurred. During CVac, all but 1 subject in the study remained blood-smear negative, while only 1 subject (primaquine/chloroquine arm) remained polymerase chain reaction-negative. Upon challenge, compared to infectivity controls, 3/3 chloroquine arm subjects displayed delayed patent parasitemia (P = .01) but not sterile protection, while 3/11 primaquine/chloroquine subjects remained blood-smear negative. CONCLUSIONS: CVac-primaquine/chloroquine is safe and induces sterile immunity to P. falciparum in some recipients, but a single 45 mg dose of primaquine postexposure does not completely prevent blood-stage parasitemia. Unlike previous studies, CVac-chloroquine did not produce sterile immunity. CLINICAL TRIALS REGISTRATION: NCT01500980.


Asunto(s)
Antimaláricos , Malaria Falciparum , Adulto , Animales , Antimaláricos/uso terapéutico , Quimioprevención , Humanos , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/prevención & control , Plasmodium falciparum , Esporozoítos , Vacunación
2.
Blood ; 134(24): 2149-2158, 2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31697826

RESUMEN

Immunotherapy with the adoptive transfer of T cells redirected with CD19-specific chimeric antigen receptors (CARs) for B-lineage acute lymphoblastic leukemia (ALL) can salvage >80% of patients having relapsed/refractory disease. The therapeutic index of this emerging modality is attenuated by the occurrence of immunologic toxicity syndromes that occur upon CAR T-cell engraftment. Here, we report on the low incidence of severe cytokine release syndrome (CRS) in a subject treated with a CAR T-cell product composed of a defined ratio CD4:CD8 T-cell composition with a 4-1BB:zeta CAR targeting CD19 who also recieved early intervention treatment. We report that early intervention with tocilizumab and/or corticosteroids may reduce the frequency at which subjects transition from mild CRS to severe CRS. Although early intervention doubled the numbers of subjects dosed with tocilizumab and/or corticosteroids, there was no apparent detrimental effect on minimal residual disease-negative complete remission rates or subsequent persistence of functional CAR T cells compared with subjects who did not receive intervention. Moreover, early intervention therapy did not increase the proportion of subjects who experience neurotoxicity or place subjects at risk for infectious sequelae. These data support the contention that early intervention with tocilizumab and/or corticosteroids in subjects with early signs of CRS is without negative impact on the antitumor potency of CD19 CAR T cells. This intervention serves to enhance the therapeutic index in relapsed/refractory patients and provides the rationale to apply CAR T-cell therapy more broadly in ALL therapy. This trial was registered at www.clinicaltrials.gov as #NCT020284.


Asunto(s)
Antígenos CD19/inmunología , Antígenos CD19/metabolismo , Síndrome de Liberación de Citoquinas/etiología , Inmunoterapia Adoptiva , Leucemia-Linfoma Linfoblástico de Células Precursoras B/complicaciones , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Receptores de Antígenos de Linfocitos T/metabolismo , Adolescente , Corticoesteroides/administración & dosificación , Corticoesteroides/farmacología , Adulto , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/farmacología , Niño , Preescolar , Síndrome de Liberación de Citoquinas/metabolismo , Citocinas/sangre , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Incidencia , Lactante , Masculino , Clasificación del Tumor , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Adulto Joven
3.
Ann Neurol ; 86(1): 42-54, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31074527

RESUMEN

OBJECTIVE: To test whether systemic cytokine release is associated with central nervous system inflammatory responses and glial injury in immune effector cell-associated neurotoxicity syndrome (ICANS) after chimeric antigen receptor (CAR)-T cell therapy in children and young adults. METHODS: We performed a prospective cohort study of clinical manifestations as well as imaging, pathology, CSF, and blood biomarkers on 43 subjects ages 1 to 25 who received CD19-directed CAR/T cells for acute lymphoblastic leukemia (ALL). RESULTS: Neurotoxicity occurred in 19 of 43 (44%) subjects. Nine subjects (21%) had CTCAE grade 3 or 4 neurological symptoms, with no neurotoxicity-related deaths. Reversible delirium, headache, decreased level of consciousness, tremor, and seizures were most commonly observed. Cornell Assessment of Pediatric Delirium (CAPD) scores ≥9 had 94% sensitivity and 33% specificity for grade ≥3 neurotoxicity, and 91% sensitivity and 72% specificity for grade ≥2 neurotoxicity. Neurotoxicity correlated with severity of cytokine release syndrome, abnormal past brain magnetic resonance imaging (MRI), and higher peak CAR-T cell numbers in blood, but not cerebrospinal fluid (CSF). CSF levels of S100 calcium-binding protein B and glial fibrillary acidic protein increased during neurotoxicity, indicating astrocyte injury. There were concomitant increases in CSF white blood cells, protein, interferon-γ (IFNγ), interleukin (IL)-6, IL-10, and granzyme B (GzB), with concurrent elevation of serum IFNγ IL-10, GzB, granulocyte macrophage colony-stimulating factor, macrophage inflammatory protein 1 alpha, and tumor necrosis factor alpha, but not IL-6. We did not find direct evidence of endothelial activation. INTERPRETATION: Our data are most consistent with ICANS as a syndrome of systemic inflammation, which affects the brain through compromise of the neurovascular unit and astrocyte injury. ANN NEUROL 2019.


Asunto(s)
Antígenos CD19/efectos adversos , Inmunoterapia Adoptiva/efectos adversos , Neuroglía/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico por imagen , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Adolescente , Adulto , Antígenos CD19/administración & dosificación , Antígenos CD19/inmunología , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Inmunoterapia Adoptiva/tendencias , Lactante , Masculino , Neuroglía/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Estudios Prospectivos , Adulto Joven
4.
J Clin Invest ; 129(5): 2123-2132, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30860496

RESUMEN

BACKGROUND: Chimeric antigen receptor (CAR) T cells can induce remission in highly refractory leukemia and lymphoma subjects, yet the parameters for achieving sustained relapse-free survival are not fully delineated. METHODS: We analyzed 43 pediatric and young adult subjects participating in a Phase I trial of defined composition CD19CAR T cells (NCT02028455). CAR T cell phenotype, function and expansion, as well as starting material T cell repertoire, were analyzed in relation to therapeutic outcome (defined as achieving complete remission within 63 days) and duration of leukemia free survival and B cell aplasia. RESULTS: These analyses reveal that initial therapeutic failures (n = 5) were associated with attenuated CAR T cell expansion and/or rapid attrition of functional CAR effector cells following adoptive transfer. The CAR T products were similar in phenotype and function when compared to products resulting in sustained remissions. However, the initial apheresed peripheral blood T cells could be distinguished by an increased frequency of LAG-3+/TNF-αlow CD8 T cells and, following adoptive transfer, the rapid expression of exhaustion markers. For the 38 subjects who achieved an initial sustained MRD-neg remission, remission durability correlated with therapeutic products having increased frequencies of TNF-α-secreting CAR CD8+ T cells, and was dependent on a sufficiently high CD19+ antigen load at time of infusion to trigger CAR T cell proliferation. CONCLUSION: These parameters have the potential to prospectively identify patients at risk for therapeutic failure and support the development of approaches to boost CAR T cell activation and proliferation in patients with low levels of CD19 antigen. TRIAL REGISTRATION: ClinicalTrials.gov NCT02028455. FUNDING: Partial funding for this study was provided by Stand Up to Cancer & St. Baldrick's Pediatric Dream Team Translational Research Grant (SU2C-AACR-DT1113), RO1 CA136551-05, Alex Lemonade Stand Phase I/II Infrastructure Grant, Conquer Cancer Foundation Career Development Award, Washington State Life Sciences Discovery Fund, Ben Towne Foundation, William Lawrence & Blanche Hughes Foundation, and Juno Therapeutics, Inc., a Celgene Company.


Asunto(s)
Antígenos CD19/metabolismo , Linfocitos T CD8-positivos/citología , Regulación Leucémica de la Expresión Génica , Inmunoterapia Adoptiva , Leucemia/terapia , Adolescente , Adulto , Antígenos CD/metabolismo , Proliferación Celular , Niño , Preescolar , Supervivencia sin Enfermedad , Femenino , Trasplante de Células Madre Hematopoyéticas , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Humanos , Inmunofenotipificación , Lactante , Células K562 , Estimación de Kaplan-Meier , Leucemia/inmunología , Activación de Linfocitos , Masculino , Fenotipo , Receptor de Muerte Celular Programada 1/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Recurrencia , Inducción de Remisión , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven , Proteína del Gen 3 de Activación de Linfocitos
5.
Biol Blood Marrow Transplant ; 25(2): 223-232, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30315942

RESUMEN

Cytotoxic chemotherapy and radiation can render lymphocyte repertoires qualitatively and quantitatively defective. Thus, heavily treated patients are often poor candidates for the manufacture of autologous chimeric antigen receptor (CAR)-T cell products. In the United States and Europe, children with high-risk neuroblastoma undergo apheresis early in the course of treatment to collect peripheral blood stem cells (PBSCs) for cryopreservation in preparation for high-dose chemotherapy followed by autologous stem cell rescue. Here, we investigate whether these cryopreserved chemotherapy and granulocyte colony-stimulating factor (G-CSF)-mobilized PBSCs can serve as starting material for CAR-T cell manufacturing. We evaluated T cell precursor subsets in cryopreserved PBSC units from 8 patients with neuroblastoma using fluorescent activated cell sorting-based analysis. Every cryopreserved unit collected early in treatment contained both CD4 and CD8 precursors with significant numbers of naïve and central memory precursors. Significant numbers of Ki67+/PD1+ T cells were detected, presumably the result of chemotherapy-induced lymphopenia and subsequent homeostatic proliferation. Cryopreserved PBSC units containing 56 to 112 × 106 T cells were amenable to immunomagnetic selection, CD3 × 28 bead activation, lentiviral transduction, and cytokine-driven expansion, provided that CD14 monocytes were depleted before the initiation of cultures. Second- and third-generation CD171 CAR+ CD4 and CD8 effector cells derived from cryopreserved units displayed antineuroblastoma lytic potency and cytokine secretion comparable to those derived from a healthy donor and mediated in vivo antitumor regression in NSG mice. We conclude that cryopreserved PBSCs procured via standard methods during early treatment can serve as an alternative starting source for CAR-T cell manufacturing, extending the options for heavily treated patients.


Asunto(s)
Traslado Adoptivo , Criopreservación , Movilización de Célula Madre Hematopoyética , Neuroblastoma , Células Madre de Sangre Periférica , Receptores Quiméricos de Antígenos/inmunología , Animales , Línea Celular Tumoral , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neuroblastoma/inmunología , Neuroblastoma/patología , Neuroblastoma/terapia , Células Madre de Sangre Periférica/inmunología , Células Madre de Sangre Periférica/patología , Estudios Retrospectivos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
PLoS One ; 9(11): e109654, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25405724

RESUMEN

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.


Asunto(s)
Experimentación Humana , Malaria Falciparum/diagnóstico , Plasmodium falciparum/patogenicidad , Esporozoítos , Adulto , Animales , Anopheles/parasitología , Anopheles/fisiología , Mordeduras y Picaduras/parasitología , Femenino , Humanos , Malaria Falciparum/complicaciones , Malaria Falciparum/etiología , Malaria Falciparum/inmunología , Masculino , Plasmodium falciparum/fisiología
7.
Mol Cell Proteomics ; 13(10): 2646-60, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25023128

RESUMEN

Malaria remains one of the most prevalent and lethal human infectious diseases worldwide. A comprehensive characterization of antibody responses to blood stage malaria is essential to support the development of future vaccines, sero-diagnostic tests, and sero-surveillance methods. We constructed a proteome array containing 4441 recombinant proteins expressed by the blood stages of the two most common human malaria parasites, P. falciparum (Pf) and P. vivax (Pv), and used this array to screen sera of Papua New Guinea children infected with Pf, Pv, or both (Pf/Pv) that were either symptomatic (febrile), or asymptomatic but had parasitemia detectable via microscopy or PCR. We hypothesized that asymptomatic children would develop antigen-specific antibody profiles associated with antidisease immunity, as compared with symptomatic children. The sera from these children recognized hundreds of the arrayed recombinant Pf and Pv proteins. In general, responses in asymptomatic children were highest in those with high parasitemia, suggesting that antibody levels are associated with parasite burden. In contrast, symptomatic children carried fewer antibodies than asymptomatic children with infections detectable by microscopy, particularly in Pv and Pf/Pv groups, suggesting that antibody production may be impaired during symptomatic infections. We used machine-learning algorithms to investigate the relationship between antibody responses and symptoms, and we identified antibody responses to sets of Plasmodium proteins that could predict clinical status of the donors. Several of these antibody responses were identified by multiple comparisons, including those against members of the serine enriched repeat antigen family and merozoite protein 4. Interestingly, both P. falciparum serine enriched repeat antigen-5 and merozoite protein 4 have been previously investigated for use in vaccines. This machine learning approach, never previously applied to proteome arrays, can be used to generate a list of potential seroprotective and/or diagnostic antigens candidates that can be further evaluated in longitudinal studies.


Asunto(s)
Malaria Falciparum/inmunología , Malaria Vivax/inmunología , Análisis por Matrices de Proteínas/métodos , Proteínas Protozoarias/análisis , Inteligencia Artificial , Niño , Preescolar , Humanos , Lactante , Malaria Falciparum/parasitología , Malaria Falciparum/patología , Malaria Vivax/parasitología , Malaria Vivax/patología , Nueva Guinea , Plasmodium falciparum/inmunología , Plasmodium falciparum/metabolismo , Plasmodium vivax/inmunología , Plasmodium vivax/metabolismo , Proteínas Protozoarias/inmunología
8.
Vaccine ; 32(19): 2135-8, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24582635

RESUMEN

Whole-parasite malaria vaccines have shown promise in clinical trials. We recently reported the first human trial of a malaria vaccine based on Plasmodium falciparum genetically attenuated parasites (PfGAP). Herein we report for the first time that PfGAP induces prolonged functional humoral responses in humans. Six volunteers were exposed to 5 bites of PfGAP-infected mosquitoes followed by approximately 200 bites. Plasma collected from all volunteers 3 months after the last exposure efficiently inhibits invasion of hepatocytes by P. falciparum sporozoites. The level of inhibition observed is comparable to that attained using plasma collected after 4-5 intravenously administrations of high numbers of irradiated sporozoites, validating the potential of PfGAP malaria vaccines. Our data highlight the role of antibody responses in pre-erythrocytic stages of human malaria, and suggests that to be protective, malaria vaccines might need to elicit long-lasting functional antibodies in addition to cellular responses.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Hepatocitos/parasitología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control , Línea Celular , Humanos , Inmunidad Humoral , Plasmodium falciparum/genética , Esporozoítos/inmunología , Vacunas Atenuadas/inmunología
9.
Vaccine ; 31(43): 4975-83, 2013 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-24029408

RESUMEN

BACKGROUND: Immunization with genetically engineered, attenuated malaria parasites (GAP) that arrest during liver infection confers sterile protection in mouse malaria models. A first generation Plasmodium falciparum GAP (Pf p52(-)/p36(-) GAP) was previously generated by deletion of two pre-erythrocytic stage-expressed genes (P52 and P36) in the NF54 strain. METHODS: A first-in-human, proof-of-concept, safety and immunogenicity clinical trial in six human volunteers was conducted. Exposure consisted of delivery of Pf p52(-)/p36(-) GAP sporozoites via infected Anopheles mosquito bite with a five-bite/volunteer exposure followed by an approximately 200-bite exposure/volunteer one month later. RESULTS: The exposures were well tolerated with mild to moderate local and systemic reactions. All volunteers remained blood stage negative after low dose exposure. Five volunteers remained blood stage negative after high dose exposure. One volunteer developed peripheral parasitemia twelve days after high dose exposure. Together the findings indicate that Pf p52(-)/p36(-) GAP was severely but not completely attenuated. All six volunteers developed antibodies to CSP. Furthermore, IFN-γ responses to whole sporozoites and multiple antigens were elicited in 5 of 6 volunteers, with both CD4 and CD8 cell cytokine production detected. CONCLUSION: Severe attenuation and favorable immune responses following administration of a first generation Pf p52(-)/p36(-) GAP suggests that further development of live-attenuated strains using genetic engineering should be pursued.


Asunto(s)
Anopheles/parasitología , Inmunización/métodos , Vacunas contra la Malaria/inmunología , Malaria/prevención & control , Plasmodium falciparum/inmunología , Esporozoítos/inmunología , Adolescente , Adulto , Animales , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Femenino , Eliminación de Gen , Genes Protozoarios , Voluntarios Sanos , Humanos , Inmunización/efectos adversos , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/efectos adversos , Vacunas contra la Malaria/genética , Masculino , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidad , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Adulto Joven
10.
Am J Trop Med Hyg ; 87(3): 460-9, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22826492

RESUMEN

The development of pre-erythrocytic Plasmodium vivax vaccines is hindered by the lack of in vitro culture systems or experimental rodent models. To help bypass these roadblocks, we exploited the fact that naturally exposed Fy- individuals who lack the Duffy blood antigen (Fy) receptor are less likely to develop blood-stage infections; therefore, they preferentially develop immune responses to pre-erythrocytic-stage parasites, whereas Fy+ individuals experience both liver- and blood-stage infections and develop immune responses to both pre-erythrocytic and erythrocytic parasites. We screened 60 endemic sera from P. vivax-exposed Fy+ or Fy- donors against a protein microarray containing 91 P. vivax proteins with P. falciparum orthologs that were up-regulated in sporozoites. Antibodies against 10 P. vivax antigens were identified in sera from P. vivax-exposed individuals but not unexposed controls. This technology has promising implications in the discovery of potential vaccine candidates against P. vivax malaria.


Asunto(s)
Antígenos de Protozoos/aislamiento & purificación , Sistema del Grupo Sanguíneo Duffy/inmunología , Eritrocitos/parasitología , Malaria Vivax/parasitología , Plasmodium vivax/inmunología , Anticuerpos Antiprotozoarios/sangre , Anticuerpos Antiprotozoarios/inmunología , Formación de Anticuerpos , Antígenos de Protozoos/genética , Colombia , Biología Computacional , Eritrocitos/inmunología , Perfilación de la Expresión Génica , Ensayos Analíticos de Alto Rendimiento , Humanos , Hígado/parasitología , Hígado/patología , Vacunas contra la Malaria/inmunología , Plasmodium vivax/genética , Plasmodium vivax/crecimiento & desarrollo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Esporozoítos/inmunología , Esporozoítos/metabolismo , Regulación hacia Arriba
11.
PLoS One ; 7(6): e39000, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22745697

RESUMEN

Co-infection with HIV and P. falciparum worsens the prognosis of both infections; however, the mechanisms driving this adverse interaction are not fully delineated. To evaluate this, we studied HIV-1 and P. falciparum interactions in vitro using peripheral blood mononuclear cells (PBMCs) from human malaria naïve volunteers experimentally infected with P. falciparum in a malaria challenge trial. PBMCs collected before the malaria challenge and at several time points post-infection were infected with HIV-1 and co-cultured with either P. falciparum infected (iRBCs) or uninfected (uRBCs) red blood cells. HIV p24Ag and TNF-α, IFN-γ, IL-4, IL-6, IL-10, IL-17, and MIP-1α were quantified in the co-culture supernatants. In general, iRBCs stimulated more HIV p24Ag production by PBMCs than did uRBCs. HIV p24Ag production by PBMCs in the presence of iRBCs (but not uRBCs) further increased during convalescence (days 35, 56, and 90 post-challenge). In parallel, iRBCs induced higher secretion of pro-inflammatory cytokines (TNF-α, IFN-γ, and MIP-1α) than uRBCs, and production increased further during convalescence. Because the increase in p24Ag production occurred after parasitemia and generalized immune activation had resolved, our results suggest that enhanced HIV production is related to the development of anti-malaria immunity and may be mediated by pro-inflammatory cytokines.


Asunto(s)
Infecciones por VIH/fisiopatología , Malaria/fisiopatología , Células Cultivadas , Quimiocina CCL3/metabolismo , Coinfección , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , Humanos , Interferón gamma/metabolismo , Malaria/inmunología , Malaria/metabolismo , Plasmodium falciparum/patogenicidad , Factor de Necrosis Tumoral alfa/metabolismo
12.
Eur J Immunol ; 42(7): 1767-77, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22585585

RESUMEN

In addition to naturally occurring regulatory T (nTreg) cells derived from the thymus, functionally competent Treg cells can be induced in vitro from peripheral blood lymphocytes in response to TCR stimulation with cytokine costimulation. Using these artificial stimulation conditions, both naïve as well as memory CD4(+) T cells can be converted into induced Treg (iTreg) cells, but the cellular origin of such iTreg cells in vivo or in response to more physiologic stimulation with pathogen-derived antigens is less clear. Here, we demonstrate that a freeze/thaw lysate of Plasmodium falciparum schizont extract (PfSE) can induce functionally competent Treg cells from peripheral lymphocytes in a time- and dose-dependent manner without the addition of exogenous costimulatory factors. The PfSE-mediated induction of Treg cells required the presence of nTreg cells in the starting culture. Further experiments mixing either memory or naïve T cells with antigen presenting cells and CFSE-labeled Treg cells identified CD4(+) CD45RO(+) CD25(-) memory T cells rather than Treg cells as the primary source of PfSE-induced Treg cells. Taken together, these data suggest that in the presence of nTreg cells, PfSE induces memory T cells to convert into iTreg cells that subsequently expand alongside PfSE-induced effector T cells.


Asunto(s)
Eritrocitos/parasitología , Malaria Falciparum/sangre , Malaria Falciparum/parasitología , Plasmodium falciparum/inmunología , Linfocitos T Reguladores/parasitología , Animales , Diferenciación Celular/inmunología , Células Cultivadas , Eritrocitos/citología , Eritrocitos/inmunología , Citometría de Flujo , Humanos , Memoria Inmunológica/inmunología , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Malaria Falciparum/inmunología , Estadísticas no Paramétricas , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología
13.
Trends Immunol ; 31(2): 63-70, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20056484

RESUMEN

T cell-mediated inflammatory immune responses contribute to both the clearance and pathology of malaria infections; the host's ability to down-regulate inflammation once parasitemia is controlled is crucial to avoid immune-mediated pathology but remains poorly understood. Various regulatory populations of T lymphocytes can modulate inflammatory immune responses and there has been considerable recent interest in the potential for regulatory T cells to modify the outcome of both murine and human malaria infections. Here, we review these studies, focussing in particular on recent studies in humans, propose a model by which different regulatory T cell populations might contribute to the control of inflammation at different stages of infection and discuss the implications for the design of safe and effective malaria vaccines.


Asunto(s)
Malaria/inmunología , Linfocitos T Reguladores/inmunología , Animales , Proliferación Celular , Susceptibilidad a Enfermedades , Humanos , Memoria Inmunológica , Vacunas contra la Malaria/inmunología , Linfocitos T Reguladores/citología
14.
Eur J Immunol ; 40(1): 47-60, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19877016

RESUMEN

Regulatory T cells (Treg) play crucial roles in regulating autoimmune responses and immunity to tumors and infectious diseases. However, numerous subpopulations of Treg are now being described and the utility of various Treg markers is being reassessed. Here we report the results of a detailed phenotypic comparison of two supposedly regulatory human T-cell populations, namely CD4+FOXP3+ T cells and CD4+CD25hi T cells. We find that CD4+FOXP3+ cells are extremely heterogeneous with respect to CD25 expression and that FOXP3+ and CD25hi CD4+ T cells differ in their expression of chemokine receptors (CCR), CD95 and Bcl-2, suggestive of distinct migration characteristics and susceptibility to apoptosis. Further, we propose that CD25 expression should be regarded as an activation marker rather than as a defining marker of Treg. Lastly, CD4+FOXP3+ T cells activated in vitro with malaria antigen expressed the highest levels of CCR4 and CD95, and the lowest levels of CCR7, indicating that they are most likely generated from effector memory cells during an immune response and rapidly succumb to apoptosis at the end of the response.


Asunto(s)
Antígenos de Protozoos/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Anciano , Apoptosis , Biomarcadores , Factores de Transcripción Forkhead/inmunología , Humanos , Memoria Inmunológica , Subunidad alfa del Receptor de Interleucina-7/inmunología , Activación de Linfocitos , Persona de Mediana Edad , Fenotipo , Receptores de Quimiocina/inmunología , Adulto Joven
15.
Eur J Immunol ; 39(5): 1288-300, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19338000

RESUMEN

An important aspect of clinical immunity to malaria is the ability to down-regulate inflammatory responses, once parasitaemia is under control, in order to avoid immune-mediated pathology. The role of classical (CD4(+)CD25(+)CD127(lo/-)FOXP3(+)) Treg in this process, however, remains controversial. Thus, we have characterized the frequency, phenotype and function of Treg populations, over time, in healthy individuals in The Gambia. We observed that both the percentage and the absolute number of CD4(+)FOXP3(+)CD127(lo/-) T cells were higher among individuals living in a rural village with highly seasonal malaria transmission than among individuals living in an urban area where malaria rarely occurs. These CD4(+)FOXP3(+)CD127(lo/-) T cells exhibited an effector memory and apoptosis-prone phenotype and suppressed cytokine production in response to malaria antigen. Cells from individuals exposed to malaria expressed significantly higher levels of mRNA for forkhead box P3 and T-box 21 (T-BET) at the end of the malaria transmission season than at the end of the non-transmission season. Importantly, the ratio of T-BET to forkhead box P3 was remarkably consistent between populations and over time, indicating that in healthy individuals, a transient increase in Th1 responses during the malaria transmission season is balanced by a commensurate Treg response, ensuring that immune homeostasis is maintained.


Asunto(s)
Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Adolescente , Adulto , Anciano , Animales , Niño , Estudios de Cohortes , Femenino , Citometría de Flujo , Factores de Transcripción Forkhead/sangre , Factores de Transcripción Forkhead/inmunología , Gambia/epidemiología , Humanos , Memoria Inmunológica/inmunología , Malaria Falciparum/epidemiología , Malaria Falciparum/parasitología , Malaria Falciparum/transmisión , Masculino , Persona de Mediana Edad , Estaciones del Año , Estudios Seroepidemiológicos , Proteínas de Dominio T Box/sangre , Proteínas de Dominio T Box/inmunología , Linfocitos T Colaboradores-Inductores/parasitología , Linfocitos T Reguladores/parasitología
16.
PLoS Pathog ; 5(4): e1000364, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19343213

RESUMEN

Failure to establish an appropriate balance between pro- and anti-inflammatory immune responses is believed to contribute to pathogenesis of severe malaria. To determine whether this balance is maintained by classical regulatory T cells (CD4(+) FOXP3(+) CD127(-/low); Tregs) we compared cellular responses between Gambian children (n = 124) with severe Plasmodium falciparum malaria or uncomplicated malaria infections. Although no significant differences in Treg numbers or function were observed between the groups, Treg activity during acute disease was inversely correlated with malaria-specific memory responses detectable 28 days later. Thus, while Tregs may not regulate acute malarial inflammation, they may limit memory responses to levels that subsequently facilitate parasite clearance without causing immunopathology. Importantly, we identified a population of FOXP3(-), CD45RO(+) CD4(+) T cells which coproduce IL-10 and IFN-gamma. These cells are more prevalent in children with uncomplicated malaria than in those with severe disease, suggesting that they may be the regulators of acute malarial inflammation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Factores de Transcripción Forkhead/metabolismo , Inmunidad Celular , Malaria Falciparum/inmunología , Enfermedad Aguda , Linfocitos T CD4-Positivos/metabolismo , Distribución de Chi-Cuadrado , Niño , Preescolar , Femenino , Factores de Transcripción Forkhead/genética , Gambia , Humanos , Memoria Inmunológica , Inflamación , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Modelos Lineales , Masculino , Estadísticas no Paramétricas , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Células TH1/metabolismo
17.
Int J Parasitol ; 34(13-14): 1517-28, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15582528

RESUMEN

Natural killer (NK) cells are lymphoid cells that mediate significant cytotoxic activity and produce high levels of pro-inflammatory cytokines in response to infection. During viral infection, NK cell cytotoxicity and cytokine production is induced principally by monocyte-macrophage- and dendritic cell-derived cytokines but virally encoded ligands for NK cells are also beginning to be described. NK derived interferon-gamma (IFN-gamma) production is also essential for control of several protozoal infections including toxoplasmosis, trypanosomiasis, leishmaniasis and malaria. The activation of NK cells by protozoan pathogens is also believed to be cytokine-mediated although some recent studies suggest that direct recognition of parasites by NK cells also occurs. Both indirect signalling via accessory cell-derived cytokines and direct signalling, presumably through NK receptors, are needed in order for human malaria parasites (Plasmodium falciparum) to optimally stimulate NK activity.


Asunto(s)
Células Asesinas Naturales/inmunología , Infecciones por Protozoos/inmunología , Humanos , Inmunidad Innata , Activación de Linfocitos , Malaria/inmunología , Receptores Inmunológicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...