Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cancer Med ; 11(2): 358-370, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34921527

RESUMEN

Filanesib is a first-in-class kinesin spindle protein inhibitor which demonstrated safety and encouraging activity in combination with bortezomib and dexamethasone in relapsed/refractory multiple myeloma in a preliminary analysis of dose-escalation phase results. This multicenter study included first a dose-escalation phase to determine maximum tolerated dose of two schedules of filanesib, bortezomib, and dexamethasone and a subsequent dose-expansion phase using the maximum tolerated doses. In the dose-expansion phase, 28 patients were evaluable for safety and efficacy. The most common grade ≥3 adverse events were neutropenia (21%) and anemia (18%), which were noncumulative and reversible, and hypertension (18%). The overall response rate was 43% with median duration of response not yet reached (range, 2.8-23.7+ months) with median follow-up of 6.3 months. A post hoc analysis incorporated 29 dose-escalation phase patients who received therapeutic filanesib doses, with an overall response rate of 39% and median duration of response of 18.0 months among the 57 total patients with median progression-free survival of 8.5 months. Notably, the PFS of high risk patients was comparable at 8.5 months, driven by the patients with 1q21 gain, characterized by increased MCL-1 expression, with a PFS of 9.1 months versus 3.5 months for the remainder of high risk patients. Patients with t(11;14) also had an encouraging PFS of 15.0 months. The combination of filanesib, bortezomib, and dexamethasone continues to show safety and encouraging activity in relapsed/refractory multiple myeloma, particularly in those patients with 1q21 gain and t(11;14).


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bortezomib/administración & dosificación , Dexametasona/administración & dosificación , Mieloma Múltiple/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Tiadiazoles/administración & dosificación , Adulto , Anciano , Aberraciones Cromosómicas , Cromosomas Humanos Par 1/genética , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Recurrencia Local de Neoplasia/patología , Supervivencia sin Progresión
2.
Leuk Res ; 74: 130-136, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30055822

RESUMEN

BACKGROUND: Exposures to DNA-damaging drugs and ionizing radiations increase risks of acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). METHODS: 9028 recipients of hematopoietic cell autotransplants (1995-2010) for Hodgkin lymphoma (HL; n = 916), non-Hodgkin lymphoma (NHL; n = 3546) and plasma cell myeloma (PCM; n = 4566), reported to the CIBMTR, were analyzed for risk of subsequent AML or MDS. RESULTS: 335 MDS/AML cases were diagnosed posttransplant (3.7%). Variables associated with an increased risk for AML or MDS in multivariate analyses were: (1) conditioning with total body radiation versus chemotherapy alone for HL (HR = 4.0; 95% confidence interval [1.4, 11.6]) and NHL (HR = 2.5 [1.1, 2.5]); (2) ≥3 versus 1 line of chemotherapy for NHL (HR = 1.9 [1.3, 2.8]); and (3) subjects with NHL transplanted in 2005-2010 versus 1995-1999 (HR = 2.1 [1.5, 3.1]). Using Surveillance, Epidemiology and End Results (SEER) data, we found risks for AML/MDS in HL, NHL and PCM to be 5-10 times the background rate. In contrast, relative risks were 10-50 for AML and approximately 100 for MDS in the autotransplant cohort. CONCLUSIONS: There are substantial risks of AML and MDS after autotransplants for HL, NHL and PCM.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda , Leucemia de Células Plasmáticas , Síndromes Mielodisplásicos , Neoplasias Primarias Secundarias , Adolescente , Adulto , Anciano , Femenino , Humanos , Leucemia Mieloide Aguda/epidemiología , Leucemia Mieloide Aguda/etiología , Leucemia de Células Plasmáticas/epidemiología , Leucemia de Células Plasmáticas/terapia , Linfoma/epidemiología , Linfoma/terapia , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/epidemiología , Síndromes Mielodisplásicos/etiología , Factores de Riesgo , Trasplante Autólogo
3.
Blood ; 130(8): 974-981, 2017 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-28637662

RESUMEN

Daratumumab plus pomalidomide and dexamethasone (pom-dex) was evaluated in patients with relapsed/refractory multiple myeloma with ≥2 prior lines of therapy who were refractory to their last treatment. Patients received daratumumab 16 mg/kg at the recommended dosing schedule, pomalidomide 4 mg daily for 21 days of each 28-day cycle, and dexamethasone 40 mg weekly. Safety was the primary end point. Overall response rate (ORR) and minimal residual disease (MRD) by next-generation sequencing were secondary end points. Patients (N = 103) received a median (range) of 4 (1-13) prior therapies; 76% received ≥3 prior therapies. The safety profile of daratumumab plus pom-dex was similar to that of pom-dex alone, with the exception of daratumumab-specific infusion-related reactions (50%) and a higher incidence of neutropenia, although without an increase in infection rate. Common grade ≥3 adverse events were neutropenia (78%), anemia (28%), and leukopenia (24%). ORR was 60% and was generally consistent across subgroups (58% in double-refractory patients). Among patients with a complete response or better, 29% were MRD negative at a threshold of 10-5 Among the 62 responders, median duration of response was not estimable (NE; 95% confidence interval [CI], 13.6-NE). At a median follow-up of 13.1 months, the median progression-free survival was 8.8 (95% CI, 4.6-15.4) months and median overall survival was 17.5 (95% CI, 13.3-NE) months. The estimated 12-month survival rate was 66% (95% CI, 55.6-74.8). Aside from increased neutropenia, the safety profile of daratumumab plus pom-dex was consistent with that of the individual therapies. Deep, durable responses were observed in heavily treated patients. The study was registered at www.clinicaltrials.gov as #NCT01998971.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Dexametasona/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Talidomida/análogos & derivados , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Demografía , Dexametasona/efectos adversos , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Recurrencia , Talidomida/efectos adversos , Talidomida/uso terapéutico , Resultado del Tratamiento
4.
Cancer ; 122(21): 3327-3335, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27433944

RESUMEN

BACKGROUND: Filanesib is a kinesin spindle protein inhibitor that has demonstrated encouraging activity in patients with recurrent/refractory multiple myeloma. Preclinical synergy with bortezomib was the rationale for the current phase 1 study. METHODS: The current study was a multicenter study with an initial dose-escalation phase to determine the maximum tolerated dose of 2 schedules of filanesib plus bortezomib with and without dexamethasone, followed by a dose-expansion phase. RESULTS: With the addition of prophylactic filgastrim, the maximum planned dose was attained: 1.3 mg/m2 /day of bortezomib plus 40 mg of dexamethasone on days 1, 8, and 15 of a 28-day cycle, with filanesib given intravenously either at a dose of 1.5 mg/m2 /day (schedule 1: days 1, 2, 15, and 16) or 3 mg/m2 /day (schedule 2: days 1 and 15). The most common adverse events (assessed for severity using version 4.0 of the National Cancer Institute Common Terminology Criteria for Adverse Events) were transient, noncumulative neutropenia and thrombocytopenia with grade 3/4 events reported in 44% (16% in cycle 1 with filgastrim) and 29% of patients, respectively. A low (≤11%) overall rate of nonhematological grade 3/4 toxicity was observed. With a median of 3 prior lines of therapy and 56% of patients with disease that was refractory to proteasome inhibitors, the overall response rate was 20% (55 patients), and was 29% in 14 patients with proteasome inhibitors-refractory disease receiving filanesib at a dose of ≥1.25 mg/m2 (duration of response, 5.2 to ≥21.2 months). CONCLUSIONS: The current phase 1 study established a dosing schedule for the combination of these agents that demonstrated a favorable safety profile with a low incidence of nonhematologic toxicity and manageable hematologic toxicity. The combination of filanesib, bortezomib, and dexamethasone appears to have durable activity in patients with recurrent/refractory multiple myeloma. Cancer 2016;122:3327-3335. © 2016 American Cancer Society.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Terapia Recuperativa , Adulto , Anciano , Bortezomib/administración & dosificación , Dexametasona/administración & dosificación , Relación Dosis-Respuesta a Droga , Femenino , Estudios de Seguimiento , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Mieloma Múltiple/patología , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Pronóstico , Tiadiazoles/administración & dosificación
5.
Biol Blood Marrow Transplant ; 22(7): 1197-1205, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27040394

RESUMEN

Busulfan, cyclophosphamide, and etoposide (BuCyE) is a commonly used conditioning regimen for autologous stem cell transplantation (ASCT). This multicenter, phase II study examined the safety and efficacy of BuCyE with individually adjusted busulfan based on preconditioning pharmacokinetics. The study initially enrolled Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL) patients ages 18 to 80 years but was amended due to high early treatment-related mortality (TRM) in patients > 65 years. BuCyE outcomes were compared with contemporaneous recipients of carmustine, etoposide, cytarabine, and melphalan (BEAM) from the Center for International Blood and Marrow Transplant Research. Two hundred seven subjects with HL (n = 66) or NHL (n = 141) were enrolled from 32 centers in North America, and 203 underwent ASCT. Day 100 TRM for all subjects (n = 203), patients > 65 years (n = 17), and patients ≤ 65 years (n = 186) were 4.5%, 23.5%, and 2.7%, respectively. The estimated rates of 2-year progression-free survival (PFS) were 33% for HL and 58%, 77%, and 43% for diffuse large B cell lymphoma (DLBCL; n = 63), mantle cell lymphoma (MCL; n = 29), and follicular lymphoma (FL; n = 23), respectively. The estimated rates of 2-year overall survival (OS) were 76% for HL and 65%, 89%, and 89% for DLBCL, MCL, and FL, respectively. In the matched analysis rates of 2-year TRM were 3.3% for BuCyE and 3.9% for BEAM, and there were no differences in outcomes for NHL. Patients with HL had lower rates of 2-year PFS with BuCyE, 33% (95% CI, 21% to 46%), than with BEAM, 59% (95% CI, 52% to 66%), with no differences in TRM or OS. BuCyE provided adequate disease control and safety in B cell NHL patients ≤ 65 years but produced worse PFS in HL patients when compared with BEAM.


Asunto(s)
Busulfano/administración & dosificación , Ciclofosfamida/uso terapéutico , Etopósido/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/métodos , Linfoma/terapia , Acondicionamiento Pretrasplante/métodos , Adulto , Anciano , Busulfano/farmacocinética , Busulfano/uso terapéutico , Carmustina/uso terapéutico , Citarabina/uso terapéutico , Combinación de Medicamentos , Trasplante de Células Madre Hematopoyéticas/mortalidad , Enfermedad de Hodgkin/mortalidad , Enfermedad de Hodgkin/terapia , Humanos , Linfoma/mortalidad , Linfoma no Hodgkin/mortalidad , Linfoma no Hodgkin/terapia , Melfalán/uso terapéutico , Persona de Mediana Edad , América del Norte , Análisis de Supervivencia , Acondicionamiento Pretrasplante/mortalidad , Trasplante Autólogo
6.
J Clin Oncol ; 31(33): 4199-206, 2013 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-24127452

RESUMEN

PURPOSE: The Programmed Death-1 (PD-1) immune checkpoint pathway may be usurped by tumors, including diffuse large B-cell lymphoma (DLBCL), to evade immune surveillance. The reconstituting immune landscape after autologous hematopoietic stem-cell transplantation (AHSCT) may be particularly favorable for breaking immune tolerance through PD-1 blockade. PATIENTS AND METHODS: We conducted an international phase II study of pidilizumab, an anti-PD-1 monoclonal antibody, in patients with DLBCL undergoing AHSCT, with correlative studies of lymphocyte subsets. Patients received three doses of pidilizumab beginning 1 to 3 months after AHSCT. RESULTS: Sixty-six eligible patients were treated. Toxicity was mild. At 16 months after the first treatment, progression-free survival (PFS) was 0.72 (90% CI, 0.60 to 0.82), meeting the primary end point. Among the 24 high-risk patients who remained positive on positron emission tomography after salvage chemotherapy, the 16-month PFS was 0.70 (90% CI, 0.51 to 0.82). Among the 35 patients with measurable disease after AHSCT, the overall response rate after pidilizumab treatment was 51%. Treatment was associated with increases in circulating lymphocyte subsets including PD-L1E-bearing lymphocytes, suggesting an on-target in vivo effect of pidilizumab. CONCLUSION: This is the first demonstration of clinical activity of PD-1 blockade in DLBCL. Given these results, PD-1 blockade after AHSCT using pidilizumab may represent a promising therapeutic strategy in this disease.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/métodos , Tolerancia Inmunológica/efectos de los fármacos , Linfoma de Células B Grandes Difuso/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/inmunología , Chile , Terapia Combinada , Supervivencia sin Enfermedad , Esquema de Medicación , Fatiga/etiología , Femenino , Humanos , India , Israel , Recuento de Linfocitos , Linfoma de Células B Grandes Difuso/sangre , Linfoma de Células B Grandes Difuso/inmunología , Masculino , Persona de Mediana Edad , Neutropenia/etiología , Receptor de Muerte Celular Programada 1/inmunología , Trasplante Autólogo , Resultado del Tratamiento , Estados Unidos , Adulto Joven
7.
Biol Blood Marrow Transplant ; 15(5): 554-63, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19361747

RESUMEN

We analyzed the outcomes of 283 patients receiving unrelated donor allogeneic hematopoietic cell transplantation for non-Hodgkin lymphoma (NHL) facilitated by the Center for International Blood and Marrow Transplant Research/National Marrow Donor Program (CIBMTR/NMDP) between 1991 and 2004. All patients received myeloablative conditioning regimens. The median follow-up of survivors is 5 years. Seventy-three (26%) patients are alive. The day 100 probability of death from all causes is estimated at 39%. The cumulative incidence of developing grade III-IV acute graft-versus-host disease (aGVHD) at day 100 is 25%. The estimated 5-year survival and failure free survival are 24% (95% confidence interval [CI]: 19-30) and 22% (95% CI: 17-28), respectively. Factors adversely associated with overall survival (OS) included increasing age, decreased performance status, and refractory disease. Follicular lymphoma (FL) and peripheral T cell lymphoma had improved survival compared to aggressive B cell lymphomas. Factors adversely associated with progression-free survival (PFS) included performance status, histology, and disease status at transplant. Long-term failure-free survival is possible following unrelated donor transplantation for NHL, although early mortality was high in this large cohort.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Linfoma no Hodgkin/terapia , Adolescente , Adulto , Distribución por Edad , Anciano , Niño , Preescolar , Femenino , Estudios de Seguimiento , Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/mortalidad , Humanos , Linfoma no Hodgkin/complicaciones , Linfoma no Hodgkin/mortalidad , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Factores de Riesgo , Análisis de Supervivencia , Donantes de Tejidos , Acondicionamiento Pretrasplante/métodos , Trasplante Homólogo , Resultado del Tratamiento , Adulto Joven
8.
Blood ; 113(21): 5074-82, 2009 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-19270260

RESUMEN

We conducted a double-blind, randomized multicenter trial to determine whether the addition of mycophenolate mofetil (MMF) improves the efficacy of initial systemic treatment of chronic graft-versus-host disease (GVHD). The primary endpoint was resolution of chronic GVHD and withdrawal of all systemic treatment within 2 years, without secondary treatment. Enrollment of 230 patients was planned, providing 90% power to observe a 20% difference in success rates between the 2 arms. The study was closed after 4 years because the interim estimated cumulative incidence of success for the primary endpoint was 23% among 74 patients in the MMF arm and 18% among 77 patients in the control arm, indicating a low probability of positive results for the primary endpoint after completing the study as originally planned. Analysis of secondary endpoints showed no evidence of benefit from adding MMF to the systemic regimen first used for treatment of chronic GVHD. The estimated hazard ratio of death was 1.99 (95% confidence interval, 0.9-4.3) among patients in the MMF arm compared with the control arm. MMF should not be added to the initial systemic treatment regimen for chronic GVHD. This trial was registered at www.clinicaltrials.gov as #NCT00089141 on August 4, 2004.


Asunto(s)
Enfermedad Injerto contra Huésped/tratamiento farmacológico , Ácido Micofenólico/análogos & derivados , Causas de Muerte , Enfermedad Crónica , Método Doble Ciego , Femenino , Enfermedad Injerto contra Huésped/mortalidad , Enfermedad Injerto contra Huésped/patología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Estado de Ejecución de Karnofsky , Masculino , Persona de Mediana Edad , Ácido Micofenólico/administración & dosificación , Tasa de Supervivencia , Resultado del Tratamiento
9.
Proc Natl Acad Sci U S A ; 104(52): 20884-9, 2007 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-18093940

RESUMEN

Although it is accepted that regulatory T cells (T regs) contribute to cancer progression, most studies in the field consider nonantigen-specific suppression. Here, we show the presence of tumor antigen-specific CD4(+) T regs in the blood of patients with metastatic melanoma. These CD4(+) T regs recognize a broad range of tumor antigens, including gp100 and TRP1 (melanoma tissue differentiation antigens), NY-ESO-1 (cancer/testis antigen) and survivin (inhibitor of apoptosis protein (IAP) family antigen). These tumor antigen-specific T regs proliferate in peripheral blood mononuclear cells (PBMC) cultures in response to specific 15-mer peptides, produce preferentially IL-10 and express high levels of FoxP3. They suppress autologous CD4(+)CD25(-) T cell responses in a cell contact-dependent manner and thus share properties of both naturally occurring regulatory T cells and type 1 regulatory T cells. Such tumor antigen-specific T regs were not detected in healthy individuals. These tumor antigen-specific T regs might thus represent another target for immunotherapy of metastatic melanoma.


Asunto(s)
Antígenos de Neoplasias/sangre , Melanoma/sangre , Melanoma/patología , Linfocitos T Reguladores/metabolismo , Adulto , Anciano , Antígenos de Neoplasias/biosíntesis , Linfocitos T CD4-Positivos/metabolismo , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Proteínas Inhibidoras de la Apoptosis , Interleucina-10/metabolismo , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Leucocitos Mononucleares/metabolismo , Masculino , Glicoproteínas de Membrana/biosíntesis , Proteínas de la Membrana/biosíntesis , Proteínas Asociadas a Microtúbulos/metabolismo , Persona de Mediana Edad , Metástasis de la Neoplasia , Proteínas de Neoplasias/metabolismo , Survivin , Tripsina , Tripsinógeno/biosíntesis , Tripsinógeno/metabolismo , Antígeno gp100 del Melanoma
10.
Immunol Rev ; 220: 129-50, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17979844

RESUMEN

Immunotherapy seeks to mobilize a patient's immune system for therapeutic benefit. It can be passive, i.e. transfer of immune effector cells (T cells) or proteins (antibodies), or active, i.e. vaccination. In cancer, passive immunotherapy can lead to some objective clinical responses, thus demonstrating that the immune system can reject tumors. However, passive immunotherapy is not expected to yield long-lived memory T cells that might control tumor outgrowth. Active immunotherapy with dendritic cell (DC)-based vaccines has the potential to induce both tumor-specific effector and memory T cells. Early clinical trials testing vaccination with ex vivo-generated DCs pulsed with tumor antigens provide a proof-of-principle that therapeutic immunity can be elicited. Yet, there is a need to improve their efficacy. The next generation of DC vaccines is expected to generate large numbers of high-avidity effector CD8(+) T cells and to overcome regulatory T cells. Therapeutic vaccination protocols will combine improved ex vivo DC vaccines with therapies that offset the suppressive environment established by tumors.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/trasplante , Neoplasias/terapia , Humanos
11.
Immunol Rev ; 219: 118-42, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17850486

RESUMEN

The dendritic cell (DC) system of antigen-presenting cells controls immunity and tolerance. DCs initiate and regulate immune responses in a manner that depends on signals they receive from microbes and their cellular environment. They allow the immune system to make qualitatively distinct responses against different microbial infections. DCs are composed of subsets that express different microbial receptors and express different surface molecules and cytokines. Our studies lead us to propose that interstitial (dermal) DCs preferentially activate humoral immunity, whereas Langerhans cells preferentially induce cellular immunity. Alterations of the DC system result in diseases such as autoimmunity, allergy, and cancer. Conversely, DCs can be exploited for vaccination, and novel vaccines that directly target DCs in vivo are being designed.


Asunto(s)
Autoinmunidad , Citocinas/metabolismo , Células Dendríticas/inmunología , Inmunidad , Infecciones/inmunología , Neoplasias/inmunología , Animales , Presentación de Antígeno , Citocinas/inmunología , Células Dendríticas/metabolismo , Humanos , Inmunidad Innata , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Proteínas Adaptadoras de Señalización NOD/inmunología , Proteínas Adaptadoras de Señalización NOD/metabolismo , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo
12.
Eur J Immunol ; 37(9): 2657-67, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17683111

RESUMEN

Dendritic cells (DC) have a unique capacity to present external antigens to CD8(+) T cells, i.e. cross-presentation. However, it is not fully established whether the ability to cross-presentation is restricted to a unique subset of DC in humans. Here, we show that two major myeloid DC subsets, i.e. Langerhans cells (LC) and interstitial DC (Int-DC), have the ability to cross-present antigens to CD8(+) T cells in vitro. LC and Int-DC were obtained from DC generated by culturing human CD34(+)-hematopoietic progenitor cells with GM-CSF, FLT3-L, and TNF-alpha (CD34-DC). Both DC subsets were able to capture necrotic/apoptotic allogeneic melanoma cells and present antigens to CD8(+) T cells, resulting in efficient priming of naive CD8(+) T cells into CTL capable of killing melanoma cells. Strikingly, a single stimulation with either subset (LC or Int-DC) or total CD34-DC loaded with necrotic/apoptotic melanoma cells was sufficient to activate melanoma-specific memory CD8(+) T cells obtained from patients with metastatic melanoma to become effective CTL. Thus, this study provides the rationale to use CD34-DC loaded with necrotic/apoptotic allogeneic melanoma cells in a clinical trial.


Asunto(s)
Antígenos de Neoplasias/inmunología , Células Dendríticas/inmunología , Células de Langerhans/inmunología , Melanoma/inmunología , Linfocitos T Citotóxicos/inmunología , Antígenos CD34/inmunología , Antígenos CD34/metabolismo , Apoptosis , Diferenciación Celular , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/metabolismo , Perfilación de la Expresión Génica , Humanos , Activación de Linfocitos , Melanoma/patología , Necrosis/patología
13.
Cancer Immunol Immunother ; 55(10): 1209-18, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16331519

RESUMEN

Between March 1999 and May 2000, 18 HLA-A*0201(+) patients with metastatic melanoma were enrolled in a phase I trial using a dendritic cell (DC) vaccine generated by culturing CD34(+) hematopoietic progenitors. This vaccine includes Langerhans cells. The DC vaccine was loaded with four melanoma peptides (MART-1/MelanA, tyrosinase, MAGE-3, and gp100), Influenza matrix peptide (Flu-MP), and keyhole limpet hemocyanin (KLH). Ten patients received eight vaccinations, one patient received six vaccinations, one patient received five vaccinations, and six patients received four vaccinations. Peptide-specific immunity was measured by IFN-gamma production and tetramer staining in blood mononuclear cells. The estimated median overall survival was 20 months (range: 2-83), and the median event-free survival was 7 months (range: 2-83). As of August 2005, four patients are alive (three patients had M1a disease and one patient had M1c disease). Three of them have had no additional therapy since trial completion; two of them had solitary lymph node metastasis, and one patient had liver metastasis. Patients who survived longer were those who mounted melanoma peptide-specific immunity to at least two melanoma peptides. The present results therefore justify the design of larger follow-up studies to assess the immunological and clinical outcomes in patients with metastatic melanoma vaccinated with peptide-pulsed CD34-derived DCs.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/inmunología , Melanoma/terapia , Proteínas de Neoplasias/inmunología , Células Madre/inmunología , Adulto , Anciano , Antígenos CD34/inmunología , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Citometría de Flujo , Humanos , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Activación de Linfocitos , Antígeno MART-1 , Melanoma/inmunología , Melanoma/mortalidad , Glicoproteínas de Membrana/inmunología , Persona de Mediana Edad , Monofenol Monooxigenasa/inmunología , Análisis de Supervivencia , Tiempo , Resultado del Tratamiento , Antígeno gp100 del Melanoma
14.
Blood ; 105(4): 1408-16, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15486071

RESUMEN

The association of ethnicity with the incidence of graft-versus-host disease (GVHD) and other clinical outcomes after transplantation is controversial. We compared the results of HLA-identical sibling bone marrow transplantations for leukemia, performed between 1990 and 1999, among different ethnic populations, including 562 Japanese, 829 white Americans, 71 African Americans, 195 Scandinavians, and 95 Irish. Results for adults and children were analyzed separately. Multivariate analyses of adult patients showed that white Americans, African Americans, and Irish cohorts were at significantly higher risk for acute GVHD than Japanese or Scandinavian cohorts (relative risk [RR] = 1.77, P < .001; RR = 1.84, P < .006; RR = 2.22, P < .001, respectively). White Americans, African Americans, and Irish, but not Scandinavians, were at significantly higher risk for early (within 3 months of transplantation) transplant-related mortality (TRM) compared with Japanese (RR = 2.99, P < .001; RR = 5.88, P < .001; RR = 2.66, P < .009, respectively). No differences in the risk for chronic GVHD, relapse, and overall survival were noted. In the pediatric cohort (limited to Japanese and white Americans), white Americans were at significantly higher risk for acute (RR = 1.93; P = .04) and chronic (RR = 3.16; P = .002) GVHD. No differences in other clinical outcomes were noted. Our findings suggest that ethnicity may influence the risk for GVHD, though overall survival rates after transplantation remain similar.


Asunto(s)
Trasplante de Médula Ósea/etnología , Trasplante de Médula Ósea/mortalidad , Enfermedad Injerto contra Huésped/etnología , Enfermedad Injerto contra Huésped/mortalidad , Antígenos HLA/análisis , Prueba de Histocompatibilidad , Hermanos , Adulto , Factores de Edad , Alelos , Trasplante de Médula Ósea/inmunología , Niño , Preescolar , Supervivencia sin Enfermedad , Femenino , Frecuencia de los Genes , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Antígenos HLA/genética , Humanos , Lactante , Recién Nacido , Leucemia/etiología , Leucemia/mortalidad , Masculino , Recurrencia , Resultado del Tratamiento
15.
Blood ; 101(5): 2015-23, 2003 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-12393427

RESUMEN

Although numerous reports indicate that patients receiving autotransplants for lymphoma are at increased risk for myelodysplastic syndrome (MDS)/acute myeloid leukemia (AML), the separate contributions of pretransplantation- and transplantation-related therapy are not well characterized. We conducted a case-control study of 56 patients with MDS/AML and 168 matched controls within a cohort of 2 739 patients receiving autotransplants for Hodgkin disease or non-Hodgkin lymphoma at 12 institutions (1989-1995). Detailed abstraction of medical records was undertaken to determine all pre- and posttransplantation therapy, and transplantation-related procedures. In multivariate analyses, risks of MDS/AML significantly increased with the intensity of pretransplantation chemotherapy with mechlorethamine (relative risks [RRs] = 2.0 and 4.3 for cumulative doses < 50 mg/m2 and > or = 50 mg/m,2 respectively; trend over dose categories, P =.04) or chlorambucil (RRs = 3.8 and 8.4 for duration < 10 months or > or = 10 months, respectively; trend, P =.009), compared with cyclophosphamide-based therapy. Transplantation-conditioning regimens including total-body irradiation (TBI) at doses 12 Gy or less did not appear to elevate leukemia risk (RR = 1.3; P =.48) compared with non-TBI regimens; however, a statistically significant increased risk was found for TBI doses of 13.2 Gy (RR = 4.6; P =.03). Peripheral blood stem cells were associated with a nonsignificant increased risk of MDS/AML (RR = 1.8; P =.12) compared with bone marrow grafts. Our data show that type and intensity of pretransplantation chemotherapy with alkylating agents are important risk factors of MDS/AML following autotransplantation. Transplantation-related factors may also modulate this risk; however, the apparent contribution of high-dose TBI requires confirmation.


Asunto(s)
Trasplante de Médula Ósea/efectos adversos , Leucemia Mieloide/epidemiología , Linfoma/terapia , Síndromes Mielodisplásicos/epidemiología , Neoplasias Primarias Secundarias/epidemiología , Trasplante de Células Madre de Sangre Periférica/efectos adversos , Enfermedad Aguda , Adolescente , Adulto , Antineoplásicos Alquilantes/administración & dosificación , Antineoplásicos Alquilantes/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Estudios de Casos y Controles , Niño , Clorambucilo/administración & dosificación , Clorambucilo/efectos adversos , Estudios de Cohortes , Ciclofosfamida/administración & dosificación , Relación Dosis-Respuesta en la Radiación , Femenino , Humanos , Leucemia Mieloide/etiología , Leucemia Inducida por Radiación/epidemiología , Leucemia Inducida por Radiación/etiología , Masculino , Mecloretamina/administración & dosificación , Mecloretamina/efectos adversos , Persona de Mediana Edad , Análisis Multivariante , Síndromes Mielodisplásicos/etiología , Neoplasias Primarias Secundarias/etiología , Prednisona/administración & dosificación , Procarbazina/administración & dosificación , Riesgo , Acondicionamiento Pretrasplante/efectos adversos , Trasplante Autólogo , Vincristina/administración & dosificación , Irradiación Corporal Total/efectos adversos
17.
Curr Opin Hematol ; 9(3): 202-6, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11953665

RESUMEN

Hematopoietic growth factors have made it possible to collect, manufacture, and engineer dendritic cells ex vivo for clinical use and expand dendritic cell subsets in vivo when administered to patients. Dendritic cells are important vectors in the induction of an effective immune response against infection and neoplastic disease. Antigens alone, even those preprocessed to bind to antigen-presenting major histocompatibility complex class I and II molecules, are insufficient to regulate effective T-cell-mediated immunity. Activated dendritic cells are essential to this task. Studies of dendritic cell biology in the laboratory and preclinical studies have facilitated the implementation of clinical trials using dendritic cells in the treatment of melanoma and other cancers. Dendritic cell subset functional differences, effective tumor target antigen loading of dendritic cells for presentation to immune effector cells, dendritic cell maturation, the route of dendritic cell administration to humans, and immunologic monitoring are parameters that require vigorous study in the context of dendritic cell immunotherapy of cancer.


Asunto(s)
Vacunas contra el Cáncer , Células Dendríticas/fisiología , Factores de Crecimiento de Célula Hematopoyética , Animales , Células Dendríticas/inmunología , Factores de Crecimiento de Célula Hematopoyética/inmunología , Humanos , Neoplasias/inmunología , Neoplasias/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA