Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Nature ; 615(7954): 920-924, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36922593

RESUMEN

Targeting critical epigenetic regulators reverses aberrant transcription in cancer, thereby restoring normal tissue function1-3. The interaction of menin with lysine methyltransferase 2A (KMT2A), an epigenetic regulator, is a dependence in acute leukaemia caused by either rearrangement of KMT2A or mutation of the nucleophosmin 1 gene (NPM1)4-6. KMT2A rearrangements occur in up to 10% of acute leukaemias and have an adverse prognosis, whereas NPM1 mutations occur in up to 30%, forming the most common genetic alteration in acute myeloid leukaemia7,8. Here, we describe the results of the first-in-human phase 1 clinical trial investigating revumenib (SNDX-5613), a potent and selective oral inhibitor of the menin-KMT2A interaction, in patients with relapsed or refractory acute leukaemia (ClinicalTrials.gov, NCT04065399). We show that therapy with revumenib was associated with a low frequency of grade 3 or higher treatment-related adverse events and a 30% rate of complete remission or complete remission with partial haematologic recovery (CR/CRh) in the efficacy analysis population. Asymptomatic prolongation of the QT interval on electrocardiography was identified as the only dose-limiting toxicity. Remissions occurred in leukaemias refractory to multiple previous lines of therapy. We demonstrate clearance of residual disease using sensitive clinical assays and identify hallmarks of differentiation into normal haematopoietic cells, including differentiation syndrome. These data establish menin inhibition as a therapeutic strategy for susceptible acute leukaemia subtypes.


Asunto(s)
Antineoplásicos , N-Metiltransferasa de Histona-Lisina , Leucemia Mieloide Aguda , Nucleofosmina , Proteínas Proto-Oncogénicas , Humanos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Neoplasia Residual/tratamiento farmacológico , Nucleofosmina/genética , Pronóstico , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Inducción de Remisión
2.
Clin Genitourin Cancer ; 16(2): 117-129, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29325739

RESUMEN

Immunotherapy with programmed cell death 1/ligand 1 (PD-1/PD-L1) checkpoint inhibitors has expanded a previously limited pool of effective treatment options for patients with metastatic urothelial carcinoma, particularly those with recurring or refractory disease and those who are ineligible for cisplatin. This review reports key findings from completed and ongoing clinical trials that highlight the potential of PD-1/PD-L1 blockade in urothelial carcinoma. A literature search was performed of PubMed, Embase, ClinicalTrials.gov, and selected annual congress abstracts. Prospective studies, reviews, editorials, and descriptions of ongoing anti-PD-1/PD-L1 studies in bladder cancer were included. Anti-PD-1/PD-L1 monoclonal antibodies have shown efficacy and safety across patient subgroups with urothelial carcinoma, including those with poor prognostic factors. Efficacy was similar across different anti-PD-1/PD-L1 agents. Although these antibodies have demonstrated durable responses in a subset of patients with urothelial carcinoma, clinicians are currently unable to predict which patients may derive benefit from immune checkpoint blockade. Anti-PD-1/PD-L1 antibodies have shown favorable clinical activity and tolerability in patients with metastatic urothelial carcinoma refractory to platinum-based therapy or who are ineligible for cisplatin. The activity of PD-1/PD-L1 inhibitors is now also being studied as first-line monotherapy in cisplatin-eligible patients in combination with chemotherapy as maintenance therapy after first-line chemotherapy, and in earlier disease states, such as muscle-invasive and non-muscle-invasive bladder cancer. Better predictive tools to define target patient populations are needed, as are further investigations to define optimal combinations or sequencing of treatments.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Carcinoma de Células Transicionales/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Antineoplásicos Inmunológicos/efectos adversos , Antígeno B7-H1/antagonistas & inhibidores , Carcinoma de Células Transicionales/inmunología , Ensayos Clínicos como Asunto , Humanos , Inmunoterapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Resultado del Tratamiento , Neoplasias de la Vejiga Urinaria/inmunología
3.
Lancet Oncol ; 19(1): 51-64, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29217288

RESUMEN

BACKGROUND: The approval of anti-programmed death ligand 1 (PD-L1) and anti-programmed death 1 agents has expanded treatment options for patients with locally advanced or metastatic urothelial carcinoma. Avelumab, a human monoclonal anti-PD-L1 antibody, has shown promising antitumour activity and safety in this disease. We aimed to assess the safety profile in patients (both post-platinum therapy and cisplatin-naive) treated with avelumab and to assess antitumour activity of this drug in post-platinum patients. METHODS: In this pooled analysis of two cohorts from the phase 1 dose-expansion JAVELIN Solid Tumor study, patients aged 18 years and older with histologically or cytologically confirmed locally advanced or metastatic urothelial carcinoma that had progressed after at least one previous platinum-based chemotherapy were enrolled from 80 cancer treatment centres or hospitals in the USA, Europe, and Asia. Eligible patients had adequate end-organ function, an Eastern Cooperative Oncology Group performance status of 0 or 1, life expectancy of at least 3 months, and at least one measurable lesion. Cisplatin-ineligible patients who might have been previously treated in the perioperative setting, including platinum-naive patients, were also eligible. Patients unselected for PD-L1 expression received avelumab (10 mg/kg, 1 h intravenous infusion) every 2 weeks until confirmed disease progression, unacceptable toxicity, or other criterion for withdrawal. The primary endpoint for this efficacy expansion cohort was confirmed best overall response (according to RECIST version 1.1), adjudicated by independent review. Safety analysis was done in all patients who received at least one dose of avelumab. Antitumour activity was assessed in post-platinum patients who received at least one dose of avelumab. This trial is registered with ClinicalTrials.gov, number NCT01772004; enrolment in this cohort of patients with metastatic urothelial carcinoma is closed and the trial is ongoing. FINDINGS: Between Sept 3, 2014, and March 15, 2016, 329 patients with advanced metastatic urothelial carcinoma were screened for enrolment into this study; 249 patients were eligible and received treatment with avelumab for a median of 12 weeks (IQR 6·0-19·7) and followed up for a median of 9·9 months (4·3-12·1). Safety and antitumour activity were evaluated at data cutoff on June 9, 2016. In 161 post-platinum patients with at least 6 months of follow-up, a best overall response of complete or partial response was recorded in 27 patients (17%; 95% CI 11-24), including nine (6%) complete responses and 18 (11%) partial responses. The most frequent treatment-related adverse events (any grade in ≥10% patients) were infusion-related reaction (73 [29%]; all grade 1-2) and fatigue (40 [16%]). Grade 3 or worse treatment-related adverse events occurred in 21 (8%) of 249 patients, the most common of which were fatigue (four [2%]), and asthenia, elevated lipase, hypophosphataemia, and pneumonitis in two (1%) patients each. 19 (8%) of 249 patients had a serious adverse event related to treatment with avelumab, and one treatment-related death occurred (pneumonitis). INTERPRETATION: Avelumab showed antitumour activity in the treatment of patients with platinum-refractory metastatic urothelial carcinoma; a manageable safety profile was reported in all avelumab-treated patients. These data provide the rationale for therapeutic use of avelumab in metastatic urothelial carcinoma and it has received accelerated US FDA approval in this setting on this basis. FUNDING: Merck KGaA, and Pfizer Inc.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Carcinoma/tratamiento farmacológico , Compuestos Organoplatinos/uso terapéutico , Compuestos de Platino/uso terapéutico , Neoplasias Urológicas/tratamiento farmacológico , Urotelio/efectos de los fármacos , Anciano , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales Humanizados , Antineoplásicos Inmunológicos/efectos adversos , Asia , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Carcinoma/inmunología , Carcinoma/mortalidad , Carcinoma/secundario , Supervivencia sin Enfermedad , Europa (Continente) , Femenino , Humanos , Masculino , Persona de Mediana Edad , Compuestos Organoplatinos/efectos adversos , Compuestos de Platino/efectos adversos , Factores de Tiempo , Insuficiencia del Tratamiento , Estados Unidos , Neoplasias Urológicas/inmunología , Neoplasias Urológicas/mortalidad , Neoplasias Urológicas/patología , Urotelio/inmunología , Urotelio/patología
4.
Haematologica ; 100(8): 1064-75, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26088929

RESUMEN

Acute promyelocytic leukemia is an aggressive malignancy characterized by the accumulation of promyelocytes in the bone marrow. PML/RARA is the primary abnormality implicated in this pathology, but the mechanisms by which this chimeric fusion protein initiates disease are incompletely understood. Identifying PML/RARA targets in vivo is critical for comprehending the road to pathogenesis. Utilizing a novel sorting strategy, we isolated highly purified promyelocyte populations from normal and young preleukemic animals, carried out microarray and methylation profiling analyses, and compared the results from the two groups of animals. Surprisingly, in the absence of secondary lesions, PML/RARA had an overall limited impact on both the transcriptome and methylome. Of interest, we did identify down-regulation of secondary and tertiary granule genes as the first step engaging the myeloid maturation block. Although initially not sufficient to arrest terminal granulopoiesis in vivo, such alterations set the stage for the later, complete differentiation block seen in leukemia. Further, gene set enrichment analysis revealed that PML/RARA promyelocytes exhibit a subtle increase in expression of cell cycle genes, and we show that this leads to both increased proliferation of these cells and expansion of the promyelocyte compartment. Importantly, this proliferation signature was absent from the poorly leukemogenic p50/RARA fusion model, implying a critical role for PML in the altered cell-cycle kinetics and ability to initiate leukemia. Thus, our findings challenge the predominant model in the field and we propose that PML/RARA initiates leukemia by subtly shifting cell fate decisions within the promyelocyte compartment.


Asunto(s)
Metilación de ADN , Células Precursoras de Granulocitos/metabolismo , Leucemia Promielocítica Aguda/genética , Proteínas de Fusión Oncogénica/genética , Transcripción Genética , Animales , Antígenos CD34/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Análisis por Conglomerados , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Células Precursoras de Granulocitos/patología , Humanos , Inmunofenotipificación , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/patología , Ratones , Ratones Transgénicos , Células Madre Neoplásicas/metabolismo , Proteínas de Fusión Oncogénica/metabolismo
5.
Infect Dis (Lond) ; 47(4): 218-24, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25650728

RESUMEN

BACKGROUND: Pediatric hematopoietic stem cell transplant (HSCT) recipients are at high risk of invasive fungal infections (IFIs). METHODS: To characterize IFIs and changes in fungal organisms over time in pediatric HSCT patients, we performed a retrospective cohort study of all HSCTs performed in pediatric patients at UCLA between 1991 and 2006. RESULTS: In all, 318 patients underwent 324 HSCT transplants over the 15-year period and 69 unique fungal infections were identified in 47 transplant patients. The overall incidence of fungal infections in HSCT recipients was 14.5%, with predominant organisms including Candida species (51%) and Aspergillus species (26%), with Candida albicans accounting for 18.8% of all fungal species. The distribution of organisms over time demonstrated a strong trend towards an increase in rare molds in more recent years. The respiratory tract was the main site of infection (52.6%), with urine and blood also noted as significant sites. Of all deaths in the patients with IFIs, fungal-related mortality accounted for 67.6% of deaths. CONCLUSIONS: HSCT patients have a much higher risk of fungal infections with rarer organisms becoming more prevalent, a finding likely linked to evolving antifungal practices over time. This emphasizes the need for the development and implementation of improved diagnostic, prophylactic, and therapeutic strategies to improve patient survival.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/estadística & datos numéricos , Micosis/epidemiología , Adolescente , Antifúngicos/uso terapéutico , Niño , Preescolar , Humanos , Incidencia , Micosis/tratamiento farmacológico , Micosis/microbiología , Estudios Retrospectivos
6.
Pediatr Blood Cancer ; 60(12): 1922-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23913590

RESUMEN

Pediatric cancer survivors have increased risk of obesity, hypertension, dyslipidemia, and type 2 diabetes, leading to premature cardiovascular disease (CVD). Multiple tissues that are involved in glucose homeostasis and lipid metabolism are adversely affected by chemotherapy. This review highlights the relevant tissue and molecular end-organ effects of therapy exposures and synthesizes the current understanding of the mechanisms underlying CVD risk in this vulnerable population. The review also approaches the topic from a developmental perspective, with the goal of providing a translational approach to identifying the antecedents of overt CVD among survivors of pediatric cancer.


Asunto(s)
Síndrome Metabólico/etiología , Síndrome Metabólico/fisiopatología , Neoplasias/complicaciones , Sobrevivientes , Antineoplásicos/efectos adversos , Trastornos Cerebrovasculares/etiología , Trastornos Cerebrovasculares/fisiopatología , Humanos , Neoplasias/terapia
7.
Pediatr Blood Cancer ; 60(6): 949-52, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23024106

RESUMEN

BACKGROUND: The aim of this study was to evaluate serum 25-hydroxyvitamin D (25OHD) concentrations in survivors of childhood cancer and compare levels by underlying diagnosis and as a function of time. PROCEDURE: A retrospective review of 201 pediatric cancer survivors enrolled in a hospital-based cancer survivor registry. Demographic characteristics and 25OHD levels were extracted from the registry. Vitamin D status was determined during routine clinical care and was categorized as normal, insufficient, or deficient. RESULTS: 25OHD levels differed significantly across diagnoses (P = 0.017), with the lowest levels found in patients treated for osteosarcoma, retinoblastoma, hepatoblastoma, and myeloid leukemias. Age was inversely correlated with 25OHD levels (P = 0.03). Average 25OHD level at study entry was 29.8 ng/ml (range: 5-79.7), with 14.4% vitamin D deficient, 39.3% insufficient, and 46.3% normal. 25OHD concentrations decreased 11.4% over time (P < 0.00001). CONCLUSION: Fewer than half of childhood cancer survivors have normal 25OHD concentrations, which further declined over time. Patients with solid tumors were the most affected, despite their lack of routine exposure to glucocorticoids. Future investigations should focus on why vitamin D level varies by diagnosis and how best to replete in this population.


Asunto(s)
Neoplasias/sangre , Sobrevivientes , Vitamina D/sangre , Niño , Femenino , Humanos , Masculino , Neoplasias/patología , Estudios Retrospectivos , Factores de Tiempo
8.
J Pediatr Hematol Oncol ; 27(3): 135-40, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15750444

RESUMEN

The purpose of this study was to determine the incidence of fungal infections in pediatric hematology and oncology (PHO) patients and to describe variations regarding site of infection, organisms, and mortality. The records of 1,052 patients presenting to the UCLA PHO service with various malignancies from 1991 to 2001 were retrospectively reviewed. No patient received invasive antifungal prophylaxis. Transplant patients were excluded. The 11-year incidence of fungal infections in this pediatric oncology cohort was 4.9%. There was a linear increase in the incidence of fungal infections from 2.9% to 7.8% between 1996 and 2001 (P = 0.001). Patients with acute leukemia represented 36% of the population but had a disproportionate incidence (67%) of fungal infections. Adolescents had twice the expected incidence of infection (P < 0.0001). Overall, Candida sp. was the major pathogen. Over time, a trend of fewer infections caused by Candida and more due to Aspergillus was noted. Blood-borne infections decreased over time, while those in the urinary and respiratory tracts increased (P = 0.04). Sixty-two percent of infections occurred in neutropenic patients. PHO patients had an overall mortality of 21%, but those with fungal infections experienced a 2.6-fold higher mortality that was not attributable to infections alone. Empiric antifungal therapy had no effect on mortality rates. Concurrent nonfungal infections did not increase mortality rates. The incidence of fungal infections increased over time, possibly as a result of advances in antibacterial and chemotherapeutic regimens. Adolescents and patients with leukemia were especially at risk. Fungal infections are a poor prognostic factor, independent of fungal-related mortality. New diagnostic methods allowing for early detection and treatment as well as more effective therapies are needed.


Asunto(s)
Neoplasias Hematológicas/complicaciones , Micosis/complicaciones , Micosis/epidemiología , Adolescente , Adulto , Antifúngicos/uso terapéutico , Niño , Preescolar , Femenino , Neoplasias Hematológicas/mortalidad , Humanos , Incidencia , Masculino , Micosis/terapia , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...