Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Lancet ; 396(10254): 839-852, 2020 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-32888407

RESUMEN

BACKGROUND: Lisocabtagene maraleucel (liso-cel) is an autologous, CD19-directed, chimeric antigen receptor (CAR) T-cell product. We aimed to assess the activity and safety of liso-cel in patients with relapsed or refractory large B-cell lymphomas. METHODS: We did a seamless design study at 14 cancer centres in the USA. We enrolled adult patients (aged ≥18 years) with relapsed or refractory large B-cell lymphomas. Eligible histological subgroups included diffuse large B-cell lymphoma, high-grade B-cell lymphoma with rearrangements of MYC and either BCL2, BCL6, or both (double-hit or triple-hit lymphoma), diffuse large B-cell lymphoma transformed from any indolent lymphoma, primary mediastinal B-cell lymphoma, and follicular lymphoma grade 3B. Patients were assigned to one of three target dose levels of liso-cel as they were sequentially tested in the trial (50 × 106 CAR+ T cells [one or two doses], 100 × 106 CAR+ T cells, and 150 × 106 CAR+ T cells), which were administered as a sequential infusion of two components (CD8+ and CD4+ CAR+ T cells) at equal target doses. Primary endpoints were adverse events, dose-limiting toxicities, and the objective response rate (assessed per Lugano criteria); endpoints were assessed by an independent review committee in the efficacy-evaluable set (comprising all patients who had confirmed PET-positive disease and received at least one dose of liso-cel). This trial is registered with ClinicalTrials.gov, NCT02631044. FINDINGS: Between Jan 11, 2016, and July 5, 2019, 344 patients underwent leukapheresis for manufacture of CAR+ T cells (liso-cel), of whom 269 patients received at least one dose of liso-cel. Patients had received a median of three (range 1-8) previous lines of systemic treatment, with 260 (97%) patients having had at least two lines. 112 (42%) patients were aged 65 years or older, 181 (67%) had chemotherapy-refractory disease, and seven (3%) had secondary CNS involvement. Median follow-up for overall survival for all 344 patients who had leukapheresis was 18·8 months (95% CI 15·0-19·3). Overall safety and activity of liso-cel did not differ by dose level. The recommended target dose was 100 × 106 CAR+ T cells (50 × 106 CD8+ and 50 × 106 CD4+ CAR+ T cells). Of 256 patients included in the efficacy-evaluable set, an objective response was achieved by 186 (73%, 95% CI 66·8-78·0) patients and a complete response by 136 (53%, 46·8-59·4). The most common grade 3 or worse adverse events were neutropenia in 161 (60%) patients, anaemia in 101 (37%), and thrombocytopenia in 72 (27%). Cytokine release syndrome and neurological events occurred in 113 (42%) and 80 (30%) patients, respectively; grade 3 or worse cytokine release syndrome and neurological events occurred in six (2%) and 27 (10%) patients, respectively. Nine (6%) patients had a dose-limiting toxicity, including one patient who died from diffuse alveolar damage following a dose of 50 × 106 CAR+ T cells. INTERPRETATION: Use of liso-cel resulted in a high objective response rate, with a low incidence of grade 3 or worse cytokine release syndrome and neurological events in patients with relapsed or refractory large B-cell lymphomas, including those with diverse histological subtypes and high-risk features. Liso-cel is under further evaluation at first relapse in large B-cell lymphomas and as a treatment for other relapsed or refractory B-cell malignancies. FUNDING: Juno Therapeutics, a Bristol-Myers Squibb Company.


Asunto(s)
Antígenos CD19/uso terapéutico , Inmunoterapia Adoptiva/métodos , Linfoma de Células B Grandes Difuso/patología , Linfoma de Células B Grandes Difuso/terapia , Anciano , Anciano de 80 o más Años , Anemia/epidemiología , Antígenos CD19/administración & dosificación , Antígenos CD19/efectos adversos , Productos Biológicos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/trasplante , Síndrome de Liberación de Citoquinas/epidemiología , Femenino , Humanos , Inmunoterapia Adoptiva/efectos adversos , Infusiones Intravenosas , Leucaféresis/métodos , Linfoma de Células B Grandes Difuso/clasificación , Linfoma de Células B Grandes Difuso/inmunología , Masculino , Enfermedades del Sistema Nervioso/epidemiología , Neutropenia/epidemiología , Recurrencia , Seguridad , Análisis de Supervivencia , Trombocitopenia/epidemiología , Resultado del Tratamiento
2.
Proc Natl Acad Sci U S A ; 106(40): 17101-4, 2009 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-19805137

RESUMEN

Organisms require faithful DNA replication to avoid deleterious mutations. In yeast, replicative leading- and lagging-strand DNA polymerases (Pols epsilon and delta, respectively) have intrinsic proofreading exonucleases that cooperate with each other and mismatch repair to limit spontaneous mutation to less than 1 per genome per cell division. The relationship of these pathways in mammals and their functions in vivo are unknown. Here we show that mouse Pol epsilon and delta proofreading suppress discrete mutator and cancer phenotypes. We found that inactivation of Pol epsilon proofreading elevates base-substitution mutations and accelerates a unique spectrum of spontaneous cancers; the types of tumors are entirely different from those triggered by loss of Pol delta proofreading. Intercrosses of Pol epsilon-, Pol delta-, and mismatch repair-mutant mice show that Pol epsilon and delta proofreading act in parallel pathways to prevent spontaneous mutation and cancer. These findings distinguish Pol epsilon and delta functions in vivo and reveal tissue-specific requirements for DNA replication fidelity.


Asunto(s)
ADN Polimerasa III/genética , ADN Polimerasa II/genética , Mutación , Neoplasias/patología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Secuencia de Bases , ADN Polimerasa II/metabolismo , ADN Polimerasa III/metabolismo , Femenino , Perfilación de la Expresión Génica , Frecuencia de los Genes , Genotipo , Estimación de Kaplan-Meier , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos , Datos de Secuencia Molecular , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Semin Cancer Biol ; 20(5): 281-93, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20951805

RESUMEN

Cancer is fueled by mutations and driven by adaptive selection. Normal cells avoid deleterious mutations by replicating their genomes with extraordinary accuracy. Here we review the pathways governing DNA replication fidelity and discuss evidence implicating replication errors (point mutation instability or PIN) in carcinogenesis.


Asunto(s)
Aberraciones Cromosómicas , Reparación del ADN/genética , Replicación del ADN/genética , Mutación , Neoplasias/genética , Animales , Daño del ADN , Reparación de la Incompatibilidad de ADN , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Reordenamiento Génico , Humanos , Inestabilidad de Microsatélites , Mutación Puntual
4.
Toxicol Pathol ; 38(3): 476-85, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20190201

RESUMEN

Acute tumor lysis syndrome (ATLS) is characterized by severe metabolic abnormalities and organ dysfunction resulting from rapid destruction of neoplastic cells. Metabolic disturbances are thought to be the primary cause of clinical ATLS symptoms, which include renal dysfunction, seizures, and cardiac arrhythmias. The histopathologic lesions associated with organ dysfunction are largely unknown because of the low rate of mortality of ATLS in humans and the few cases of ATLS identified in laboratory animals. Here, we describe histologic, immunohistochemical, and electron microscopic analyses of thirty-one ATLS cases from a cohort of 499 mice that are prone to spontaneous lymphoblastic lymphoma owing to genetic defects in DNA replication fidelity. Seventy-three percent of our cohort died with lymphoblastic lymphoma, and 8% of affected mice died with diffuse microthromboemboli consistent with ATLS. Mice with ATLS had a high spontaneous mortality rate (>50%), a large tumor burden with disseminated disease, and evidence of leukemia. Blood vessels in the lung, kidney, and other organs were occluded by microthromboemboli composed of chromatin, cellular debris, fibrin, platelets, and entrapped erythrocytes and malignant cells. This case series suggests that ATLS can occur at high frequency in mice with disseminated lymphoblastic lymphoma and leads to a high rate of spontaneous death from microthromboemboli.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras/complicaciones , Síndrome de Lisis Tumoral/patología , Síndrome de Lisis Tumoral/veterinaria , Animales , Reparación de la Incompatibilidad de ADN/genética , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microscopía Electrónica de Transmisión , Tromboembolia/etiología , Tromboembolia/patología , Síndrome de Lisis Tumoral/genética
5.
Curr Biol ; 16(6): R209-11, 2006 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-16546074

RESUMEN

Proofreading is the primary guardian of DNA polymerase fidelity. New work has revealed that polymerases with intrinsic proofreading activity may cooperate with non-proofreading polymerases to ensure faithful DNA replication.


Asunto(s)
Replicación del ADN/fisiología , ADN Polimerasa Dirigida por ADN/metabolismo , ADN/metabolismo , Animales , Exonucleasas/metabolismo , Modelos Genéticos , Mutación
6.
Contemp Clin Trials ; 48: 153-65, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27109037

RESUMEN

We present a novel Bayesian adaptive phase 1 design to determine the optimal dosing regimen for an adoptive T-cell therapy in a mixed patient population. Our design is motivated by a B-cell Non-Hodgkin Lymphoma trial evaluating multiple dosing regimens within multiple disease subtypes. A utility score is calculated from both safety and efficacy utility functions and used to guide dose-escalation decisions. We pool safety data across disease subtypes and use a single dose-toxicity model while sharing efficacy information between disease subtypes using a hierarchical dose-response model. In addition, an adaptive randomization approach is applied to dynamically assign patients to a regimen when more than one regimen is open for enrollment. We illustrate this study design through a simulated trial example, and we investigate the operating characteristics using simulation studies.


Asunto(s)
Traslado Adoptivo/métodos , Linfoma de Células B Grandes Difuso/terapia , Linfoma de Células del Manto/terapia , Linfocitos T/trasplante , Antígenos CD19/metabolismo , Teorema de Bayes , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Humanos , Linfocitos T/metabolismo , Trasplante Autólogo
7.
DNA Repair (Amst) ; 9(1): 11-22, 2010 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-19896421

RESUMEN

Werner syndrome (WS) is a human autosomal recessive genetic instability and cancer predisposition syndrome with features of premature aging. Several genetically determined mouse models of WS have been generated, however, none develops features of premature aging or an elevated risk of neoplasia unless additional genetic perturbations are introduced. In order to determine whether differences in cellular phenotype could explain the discrepant phenotypes of Wrn-/- mice and WRN-deficient humans, we compared the cellular phenotype of newly derived Wrn-/- mouse primary fibroblasts with previous analyses of primary and transformed fibroblasts from WS patients and with newly derived, WRN-depleted human primary fibroblasts. These analyses confirmed previously reported cellular phenotypes of WRN-mutant and WRN-deficient human fibroblasts, and demonstrated that the human WRN-deficient cellular phenotype can be detected in cells grown in 5% or in 20% oxygen. In contrast, we did not identify prominent cellular phenotypes present in WRN-deficient human cells in Wrn-/- mouse fibroblasts. Our results indicate that human and mouse fibroblasts have different functional requirements for WRN protein, and that the absence of a strong cellular phenotype may in part explain the failure of Wrn-/- mice to develop an organismal phenotype resembling Werner syndrome.


Asunto(s)
Exodesoxirribonucleasas/metabolismo , RecQ Helicasas/metabolismo , Síndrome de Werner/enzimología , Animales , Proliferación Celular , Células Cultivadas , Daño del ADN , Exodesoxirribonucleasas/deficiencia , Histonas/metabolismo , Humanos , Longevidad , Ratones , Ratones Noqueados , Neoplasias/enzimología , Neoplasias/genética , Oxígeno/metabolismo , Fenotipo , RecQ Helicasas/deficiencia , Síndrome de Werner/genética , Síndrome de Werner/patología , Helicasa del Síndrome de Werner
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA