Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Immunother ; 41(3): 151-157, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29517616

RESUMEN

Interleukin-18 (IL-18) is an immunostimulatory cytokine that augments antibody-dependent cellular cytotoxicity mediated by human natural killer cells against antibody-coated lymphoma cells in vitro and that has antitumor activity in animal models. Ofatumumab is a CD20 monoclonal antibody with activity against human B-cell lymphomas. A phase I study of recombinant human (rh) IL-18 given with ofatumumab was undertaken in patients with CD20 lymphoma who had undergone high-dose chemotherapy and autologous peripheral blood stem cell transplantation. Cohorts of 3 patients were given intravenous infusions of ofatumumab 1000 mg weekly for 4 weeks with escalating doses of rhIL-18 as a intravenous infusion weekly for 8 consecutive weeks. Nine male patients with CD20 lymphomas were given ofatumumab in combination with rhIL-18 at doses of 3, 10, and 30 µg/kg. No unexpected or dose-limiting toxicities were observed. The mean reduction from predose levels in the number of peripheral blood natural killer cells after the first rhIL-18 infusion was 91%, 96%, and 97% for the 3, 10, and 30 µg/kg cohorts, respectively. Serum concentrations of interferon-γ and chemokines transiently increased following IL-18 dosing. rhIL-18 can be given in biologically active doses by weekly infusions in combination with ofatumumab after peripheral blood stem cell transplantation to patients with lymphoma. A maximum tolerated dose of rhIL-18 plus ofatumumab was not determined. Further studies of rhIL-18 and CD20 monoclonal antibodies in B-cell malignancies are warranted.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Linfoma/terapia , Adulto , Anciano , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Terapia Combinada , Citocinas/sangre , Citocinas/metabolismo , Femenino , Humanos , Interleucina-18/administración & dosificación , Interleucina-18/farmacocinética , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Linfoma/mortalidad , Linfoma/patología , Masculino , Persona de Mediana Edad , Trasplante de Células Madre de Sangre Periférica/métodos , Análisis de Supervivencia , Resultado del Tratamiento
2.
Cancer Immunol Immunother ; 63(3): 283-95, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24363024

RESUMEN

Immunostimulatory cytokines can enhance anti-tumor immunity and are part of the therapeutic armamentarium for cancer treatment. We have previously reported that post-transplant lymphoma patients have an acquired deficiency of signal transducer and activator of transcription 4, which results in defective IFNγ production during clinical immunotherapy. With the goal of further improving cytokine-based immunotherapy, we examined the effects of a soybean peptide called lunasin that synergistically works with cytokines on natural killer (NK) cells. Peripheral blood mononuclear cells of healthy donors and post-transplant lymphoma patients were stimulated with or without lunasin in the presence of IL-12 or IL-2. NK activation was evaluated, and its tumoricidal activity was assessed using in vitro and in vivo tumor models. Chromatin immunoprecipitation assay was performed to evaluate the histone modification of gene loci that are regulated by lunasin and cytokine. Adding lunasin to IL-12- or IL-2-stimulated NK cells demonstrated synergistic effects in the induction of IFNG and GZMB involved in cytotoxicity. The combination of lunasin and cytokines (IL-12 plus IL-2) was capable of restoring IFNγ production by NK cells from post-transplant lymphoma patients. In addition, NK cells stimulated with lunasin plus cytokines displayed higher tumoricidal activity than those stimulated with cytokines alone using in vitro and in vivo tumor models. The underlying mechanism responsible for the effects of lunasin on NK cells is likely due to epigenetic modulation on target gene loci. Lunasin represents a different class of immune modulating agent that may augment the therapeutic responses mediated by cytokine-based immunotherapy.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Inmunoterapia/métodos , Células Asesinas Naturales/efectos de los fármacos , Linfoma/terapia , Fragmentos de Péptidos/administración & dosificación , Proteínas de Soja/administración & dosificación , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Citotoxicidad Inmunológica/efectos de los fármacos , Citotoxicidad Inmunológica/genética , Metilación de ADN/efectos de los fármacos , Sinergismo Farmacológico , Granzimas/genética , Granzimas/metabolismo , Humanos , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-12/administración & dosificación , Interleucina-12/inmunología , Interleucina-2/administración & dosificación , Interleucina-2/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Linfoma/genética , Linfoma/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Datos de Secuencia Molecular , Factor de Transcripción STAT4/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cancer Immunol Immunother ; 62(6): 1073-82, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23604103

RESUMEN

The antitumor activity of monoclonal antibodies is mediated by effector cells, such as natural killer (NK) cells, that express Fc receptors for immunoglobulin. Efficacy of monoclonal antibodies, including the CD20 antibody rituximab, could be improved by agents that augment the function of NK cells. Interleukin (IL)-18 is an immunostimulatory cytokine that has antitumor activity in preclinical models. The effects of IL-18 on NK cell function mediated through Fcγ receptors were examined. Human NK cells stimulated with immobilized IgG in vitro secreted IFN-γ as expected; such IFN-γ production was partially inhibited by blocking CD16 with monoclonal antibodies. IL-18 augmented IFN-γ production by NK cells stimulated with immobilized IgG or CD16 antibodies. NK cell IFN-γ production in response to immobilized IgG and/or IL-18 was inhibited by chemical inhibitors of Syk and several other kinases involved in CD16 signaling pathways. IL-18 augmented antibody-dependent cellular cytotoxicity (ADCC) of human NK cells against rituximab-coated Raji cells in vitro. IL-18 and rituximab acted synergistically to promote regression of human lymphoma xenografts in SCID mice. Inasmuch as IL-18 costimulates IFN-γ production and ADCC of NK cells activated through Fc receptors in vitro and augments antitumor activity of rituximab in vivo, it is an attractive cytokine to combine with monoclonal antibodies for treatment of human cancer.


Asunto(s)
Interleucina-18/farmacología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales de Origen Murino/administración & dosificación , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulinas/metabolismo , Interferón gamma/biosíntesis , Interleucina-18/administración & dosificación , Células Asesinas Naturales/metabolismo , Linfoma/tratamiento farmacológico , Linfoma/inmunología , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/metabolismo , Receptores Fc/metabolismo , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Rituximab , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Blood ; 118(23): 6097-106, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21998209

RESUMEN

Signal Transducer and Activator of Transcription 4 (STAT4) is a transcription factor that is activated by IL-12 signaling and promotes Th1-cell differentiation and IFN-γ production. Defective IFN-γ production because of STAT4 mRNA and protein deficiency occurs after autologous stem cell transplantation for lymphoma. In the present study, we investigated the mechanisms of STAT4 deficiency in lymphoma patients. The tumor-bearing state is not responsible, because STAT4 levels were not significantly different in PBMCs obtained from healthy control subjects compared with those from lymphoma patients before treatment. STAT4 protein levels were significantly decreased in PBMCs and T cells obtained from lymphoma patients after standard-dose chemotherapy. Furthermore, treatment of control PBMC cultures or a natural killer cell line with chemotherapy drugs in vitro also resulted in reduced STAT4 protein and diminished, IL-12-induced IFN-γ production. Translation of STAT4 protein was not impaired in chemotherapy-treated cells, whereas the STAT4 protein half-life was significantly reduced. Chemotherapy drugs promoted the ubiquitination and proteasomal degradation of STAT4. Treatment with the proteasome inhibitor bortezomib reversed chemotherapy-induced STAT4 deficiency and defective IFN-γ production. We conclude that acquired STAT4 deficiency in lymphoma patients is a consequence of treatment with chemotherapy, results that have important implications for the design of optimal immunotherapy for lymphoma.


Asunto(s)
Etopósido/efectos adversos , Linfoma/tratamiento farmacológico , Linfoma/genética , Factor de Transcripción STAT4/genética , Factor de Transcripción STAT4/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos Alquilantes/efectos adversos , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Fitogénicos/efectos adversos , Antineoplásicos Fitogénicos/farmacología , Ácidos Borónicos/farmacología , Bortezomib , Carmustina/efectos adversos , Carmustina/farmacología , Células Cultivadas , Interacciones Farmacológicas , Etopósido/farmacología , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Humanos , Interleucina-12/genética , Interleucina-12/metabolismo , Interleucina-2/genética , Interleucina-2/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/fisiología , Linfoma/patología , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Biosíntesis de Proteínas/efectos de los fármacos , Pirazinas/farmacología , Estabilidad del ARN/efectos de los fármacos , Factor de Transcripción STAT4/deficiencia , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Ubiquitina/metabolismo
5.
Blood ; 113(23): 5887-90, 2009 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-19359411

RESUMEN

IL-12 activates STAT4, which is a critical regulator of inflammation and T helper type I (Th1) lineage development in murine systems. The requirement for STAT4 in the generation of human Th1 cells has not been examined thoroughly. Compared with control Th1 cultures, expression of the Th1 genes IFNgamma, IL-12Rbeta2, and TNFalpha is greatly reduced in Th1 cultures of CD4 T cells isolated from lymphoma patients after autologous stem cell transplantation who have acquired STAT4 deficiency. Moreover, IL-4 and IL-5 production is increased in patient Th1 cultures though there are no defects in the development of Th2 cells. Reconstitution of STAT4 in patient T cells allowed recovery of IFNgamma and IL-12Rbeta2 expression, whereas ectopic expression of IL-12Rbeta2 did not rescue STAT4 expression, and increased IFNgamma production only to levels intermediate between control and patient samples. These results demonstrate that, as in murine systems, STAT4 is required for optimal human Th1 lineage development.


Asunto(s)
Diferenciación Celular/inmunología , Factor de Transcripción STAT4/deficiencia , Factor de Transcripción STAT4/metabolismo , Células TH1/citología , Células TH1/metabolismo , Células Cultivadas , Humanos , Linfoma/genética , Linfoma/inmunología , Linfoma/metabolismo , Receptores de Interleucina-12/metabolismo , Factor de Transcripción STAT4/genética , Células TH1/inmunología
6.
Blood ; 106(3): 963-70, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15817683

RESUMEN

Production of interferon gamma (IFN-gamma) is critical for optimal antitumor immunotherapy in several preclinical animal models. Interleukin-12 (IL-12)-induced IFN-gamma production is markedly defective after autologous stem cell transplantation. Quantitative deficiency in CD4 T cells, relative increase in CD25+CD4+ T cells, and bias toward T helper 2 (Th2) differentiation are not the primary mechanisms of defective IFN-gamma production. IL-12 receptor beta1 (IL-12Rbeta1) and IL-12Rbeta2 are expressed at equivalent or higher levels on posttransplantation patient peripheral blood mononuclear cells (PBMCs) as compared with control PBMCs. IL-12-induced tyrosine phosphorylation of signal transducer and activator of transcription 4 (STAT4) was undetectable or barely detectable in posttransplantation patient PBMCs, whereas IL-4-induced tyrosine phosphorylation of STAT6 did not differ in posttransplantation patient and control PBMCs. Levels of STAT4 protein were decreased by 97% in posttransplantation patient PBMCs. Levels of STAT4 mRNA were also significantly decreased in posttransplantation patient PBMCs. Incubation with IL-12 and IL-18 in combination partially reversed the defective IFN-gamma production by posttransplantation patient PBMCs. IFN-gamma production in response to IL-12 plus IL-18 did not require increased expression of STAT4 but was dependent on the activity of p38 mitogen-activated protein kinase (MAPK). These results indicate that defective IFN-gamma production is due to an intrinsic deficiency in STAT4 expression by posttransplantation patient lymphocytes and suggest strategies for circumventing this deficiency in cancer immunotherapy.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Interferón gamma/biosíntesis , Linfoma/terapia , Transactivadores/deficiencia , Estudios de Casos y Controles , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Interferón gamma/deficiencia , Interleucina-12/farmacología , Interleucina-18/farmacología , Leucocitos Mononucleares/química , Linfoma/inmunología , Fosforilación , ARN Mensajero/análisis , Factor de Transcripción STAT4 , Transactivadores/genética , Transactivadores/metabolismo , Trasplante Autólogo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Clin Cancer Res ; 10(6): 1935-42, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15041709

RESUMEN

PURPOSE: The purpose is to determine the immunological effects of recombinant human interleukin (rhIL)-12 therapy after autologous stem cell transplantation. EXPERIMENTAL DESIGN: Twelve patients (8 non-Hodgkin's lymphoma, 2 Hodgkin's disease, and 2 plasma cell myeloma) were treated with rhIL-12 by bolus i.v. injection in doses of 30, 100, or 250 ng/kg starting at a median of 66 days posttransplant. Immunological assays were performed using serum and peripheral blood mononuclear cell (PBMC) samples obtained on study. RESULTS: Dose-dependent increases in the total lymphocyte count occurred during rhIL-12 therapy. The absolute number of peripheral blood CD4 T cells increased up to 16.3-fold, CD8 T cells up to 20.5-fold, B cells up to 11-fold, and natural killer (NK) cells up to 12.3-fold during rhIL-12 administration and returned to pretreatment baseline levels after discontinuation of rhIL-12. CD56(bright) NK cells expanded dramatically in the blood of a patient with baseline lymphopenia before rhIL-12 therapy. In vitro proliferation of patient PBMCs in response to IL-12 was indistinguishable from that of PBMCs obtained from healthy control sub-jects. Moreover, spontaneous in vitro proliferation of patient PBMCs increased significantly during rhIL-12 therapy. Increased levels of IFN-gamma and IL-18 were detected in the serum of patients treated in the 100 and 250 ng/kg dose cohorts during the first multiple dose cycle. CONCLUSIONS: Expansion of T, B, and NK cells occurs in vivo during rhIL-12 therapy after autologous stem cell transplantation for hematological malignancies. In contrast to their striking defect in IL-12-induced IFN-gamma production, posttransplant patient PBMCs exhibit normal proliferative responses to IL-12 in vitro. Additional investigation of rhIL-12 for posttransplantation immunotherapy is warranted.


Asunto(s)
Interleucina-12/uso terapéutico , Recuento de Linfocitos , Trasplante de Células Madre , Trasplante Autólogo/inmunología , Antígenos CD/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Humanos , Interleucina-12/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Subgrupos Linfocitarios/inmunología , Linfoma/inmunología , Linfoma/terapia , Mieloma Múltiple/inmunología , Mieloma Múltiple/terapia , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/uso terapéutico
8.
Clin Cancer Res ; 8(11): 3383-93, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12429625

RESUMEN

PURPOSE: To determine the safety, maximum tolerated dose,and biological effects of recombinant human IL-12 after autologous stem cell transplantation for cancer. EXPERIMENTAL DESIGN: Twelve patients with hematological malignancies (8 non-Hodgkin's lymphoma, 2 Hodgkin's disease, and 2 plasma cell myeloma) began interleukin (IL)-12 therapy at a median of 66 days after transplantation. Recombinant human IL-12 was given by bolus i.v. injection in doses of 30, 100, or 250 ng/kg once as an inpatient and then, after a 2-week hiatus, once daily for 5 consecutive days every 3 weeks on an outpatient basis. RESULTS: Common side effects included fever, chills, fatigue, nausea or vomiting, and asymptomatic elevation in serum liver function tests. Transient neutropenia and thrombocytopenia were also common, but no patient required platelet transfusion or had a neutropenic fever. Dose-limiting toxicities (diarrhea and elevated liver function tests) occurred in 2 of 3 patients treated in the 250 ng/kg cohort. Biological effects, including increases in serum IFN-gamma levels and transient lymphopenia involving CD4 T cells, CD8 T cells, B cells, and NK cells, were seen at all three dose levels. CONCLUSIONS: Biologically active doses of IL-12 can be given safely to patients after autologous stem cell transplantation for high-risk hematological malignancies. Further studies are indicated to assess the efficacy of IL-12 in this setting.


Asunto(s)
Neoplasias Hematológicas/terapia , Inmunoterapia/métodos , Interleucina-12/uso terapéutico , Trasplante de Células Madre , Adulto , Anciano , Linfocitos T CD4-Positivos/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Enfermedad de Hodgkin/terapia , Humanos , Interferón gamma/sangre , Interleucina-12/sangre , Interleucina-12/farmacocinética , Cinética , Linfoma no Hodgkin/terapia , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Plasmacitoma/terapia , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapéutico , Factores de Tiempo , Trasplante Autólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...