Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Invest New Drugs ; 38(2): 457-467, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31065954

RESUMEN

Background Fibroblast growth factors (FGFs) have a fundamental role in cancer. Sequestering FGFs with GSK3052230 (FP-1039) blocks their ability to activate FGFRs while avoiding toxicities associated with small molecule inhibitors of FGFR, including hyperphosphatemia and retinal, nail, and skin toxicities. Methods A multicenter, open-label, phase Ib study evaluated weekly GSK3052230 added to pemetrexed/cisplatin in patients with treatment-naive, unresectable malignant pleural mesothelioma. Doses were escalated according to a 3 + 3 design, followed by cohort expansion at the maximum tolerated dose (MTD). Endpoints included safety, overall response rate, progression-free survival, and pharmacokinetics. Results 36 patients were dosed at 10, 15, and 20 mg/kg doses of GSK3052230. Three dose-limiting toxicities were observed at 20 mg/kg and one at 15 mg/kg. The MTD was defined as 15 mg/kg and used for cohort expansion. The most common treatment-related adverse events (AEs) were nausea (56%), decreased appetite (36%), infusion reactions (36%), decreased neutrophil counts (36%), and fatigue (33%). The confirmed ORR was 39% (95% CI: 23.1-56.5) (14/36 PRs) and 47% had stable disease (17/36), giving a disease control rate of 86%. At 15 mg/kg GSK3052230 (n = 25), the ORR was 44% (95% CI: 24.4-65.1), and the median PFS was 7.4 months (95% CI: 6.7-13.4). Four patients had disease control for over 1 year, and three were still ongoing. Conclusion At 15 mg/kg weekly, GSK3052230 was well tolerated in combination with pemetrexed/cisplatin and durable responses were observed. Importantly, AEs associated with small molecule inhibitors of FGFR were not observed, as predicted by the unique mechanism of action of this drug.


Asunto(s)
Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Cisplatino/administración & dosificación , Inmunoglobulina G/administración & dosificación , Mesotelioma Maligno/tratamiento farmacológico , Proteínas de Fusión Oncogénica/administración & dosificación , Pemetrexed/administración & dosificación , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Cisplatino/efectos adversos , Femenino , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Inmunoglobulina G/efectos adversos , Ligandos , Masculino , Mesotelioma Maligno/metabolismo , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/efectos adversos , Proteínas de Fusión Oncogénica/farmacocinética , Pemetrexed/efectos adversos , Proteínas Recombinantes de Fusión , Resultado del Tratamiento
2.
Leukemia ; 31(1): 159-169, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27389055

RESUMEN

Granulocyte macrophage-colony-stimulating factor (GM-CSF) signaling regulates hematopoiesis and immune responses. CSF2RA, the gene encoding the α-subunit for GM-CSF, is significantly downregulated in t(8;21) (RUNX1-ETO or RE) leukemia patients, suggesting that it may serve as a tumor suppressor. We previously reported that GM-CSF signaling is inhibitory to RE leukemogenesis. Here we conducted gene expression profiling of primary RE hematopoietic stem/progenitor cells (HSPCs) treated with GM-CSF to elucidate the mechanisms mediating the negative effects of GM on RE leukemogenicity. We observed that GM treatment of RE HSPCs resulted in a unique gene expression profile that resembles primary human cells undergoing myelopoiesis, which was not observed in control HSPCs. Additionally, we discovered that GM-CSF signaling attenuates MYC-associated gene signatures in RE HSPCs. In agreement with this, a functional screen of a subset of GM-CSF-responsive genes demonstrated that a MYC inhibitor, MXI1 (Max interactor 1), reduced the leukemic potential of RE HSPCs and t(8;21) acute myeloid leukemia (AML) cells. Furthermore, MYC knockdown and treatment with the BET (bromodomain and extra terminal domain) inhibitor JQ1 reduced the leukemic potential of t(8;21) cell lines. Altogether, we discovered a novel molecular mechanism mediating the GM-CSF-induced reduction in leukemic potential of RE cells, and our findings support MYC inhibition as an effective strategy for reducing the leukemogenicity of t(8;21) AML.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Células Madre Hematopoyéticas/metabolismo , Leucemia Mieloide Aguda/etiología , Proteínas de Fusión Oncogénica/efectos adversos , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Perfilación de la Expresión Génica , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Ratones , Mielopoyesis/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-myc/fisiología , Proteína 1 Compañera de Translocación de RUNX1 , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/fisiología
3.
Ann Oncol ; 27(3): 526-32, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26646757

RESUMEN

BACKGROUND: Fibroblast growth factors (FGFs) play important roles in multiple cancers by supporting tumor growth and angiogenesis. FP-1039 (GSK3052230) is a FGF ligand trap consisting of the extracellular domain of FGF receptor 1 (FGFR1) fused with the Fc region of IgG1. FP-1039 binds and neutralizes multiple FGFs that normally bind FGFR1. The primary objective of this phase I study was to evaluate the safety and tolerability of FP-1039. PATIENTS AND METHODS: Eligible patients with metastatic or locally advanced solid tumors for which standard treatments were ineffective were treated with weekly doses of FP-1039 for 4 weeks, followed by 2 weeks observation. RESULTS: Thirty-nine subjects received a mean of 6 infusions of FP-1039 at doses ranging from 0.5 to 16 mg/kg weekly, with no maximally tolerated dose identified. Grade 3 or greater treatment emergent adverse events were uncommon. Four dose-limiting toxicities were reported at doses of 0.75 mg/kg (urticaria), 1 mg/kg (intestinal perforation and neutropenia), and 16 mg/kg (muscular weakness). Drug exposure was dose proportional, and the terminal elimination half-life was 2.6-3.9 days following a single dose. Target engagement as measured by low free plasma FGF2 levels was achieved. FGF pathway dysregulation was uncommon. No objective responses were observed. CONCLUSION: In nonselected cancer patients with advanced disease, treatment with FP-1039 was well tolerated and toxicities associated with small molecule drugs that inhibit FGFR tyrosine kinases, including hyperphosphatemia, were not observed. Further studies of FP-1039 in patients selected for FGF pathway dysregulation, who are most likely to benefit, are now underway.


Asunto(s)
Antineoplásicos/uso terapéutico , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Inmunoglobulina G/uso terapéutico , Neoplasias/tratamiento farmacológico , Proteínas de Fusión Oncogénica/uso terapéutico , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/uso terapéutico , Adulto , Anciano , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Inmunoglobulina G/efectos adversos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/efectos adversos , Proteínas de Fusión Oncogénica/farmacocinética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Proteínas Recombinantes de Fusión
4.
Int J Cancer ; 121(12): 2794-800, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17721997

RESUMEN

Persistent infection with human papillomaviruses (HPV) is a prerequisite for the development of cervical cancer. Vaccination with virus-like particles (VLP) has demonstrated efficacy in prophylaxis but lacks therapeutic potential. HPV16 L1E7 chimeric virus-like particles (CVLP) consist of a carboxy-terminally truncated HPV16L1 protein fused to the amino-terminal part of the HPV16 E7 protein and self-assemble by recombinant expression of the fusion protein. The CVLP are able to induce L1- and E7-specific cytotoxic T lymphocytes. We have performed a first clinical trial to gain information about the safety and to generate preliminary data on the therapeutic potential of the CVLP in humans. A randomized, double blind, placebo-controlled clinical trial has been conducted in 39 HPV16 mono-infected high grade cervical intraepithelial neoplasia (CIN) patients (CIN 2/3). Two doses (75 mug or 250 mug) of CVLP were applied. The duration of the study was 24 weeks with 2 optional visits after another 12 and 24 weeks. The vaccine showed a very good safety profile with only minor adverse events attributable to the immunization. Antibodies with high titers against HPV16 L1 and low titers against HPV16 E7 as well as cellular immune responses against both proteins were induced. Responses were equivalent for both vaccine concentrations. A trend for histological improvement to CIN 1 or normal was seen in 39% of the patients receiving the vaccine and only 25% of the placebo recipients. Fifty-six percent of the responders were also HPV16 DNA-negative by the end of the study. Therefore, we demonstrated evidence for safety and a nonsignificant trend for the clinical efficacy of the HPV16 L1E7 CVLP vaccine.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Papillomavirus Humano 16/inmunología , Proteínas de Fusión Oncogénica/uso terapéutico , Proteínas Oncogénicas Virales/uso terapéutico , Vacunas contra Papillomavirus/uso terapéutico , Displasia del Cuello del Útero/tratamiento farmacológico , Displasia del Cuello del Útero/virología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/virología , Adulto , Anciano , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/efectos adversos , ADN Viral/efectos de los fármacos , ADN Viral/aislamiento & purificación , Método Doble Ciego , Esquema de Medicación , Femenino , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/aislamiento & purificación , Humanos , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/administración & dosificación , Proteínas de Fusión Oncogénica/efectos adversos , Proteínas Oncogénicas Virales/administración & dosificación , Proteínas Oncogénicas Virales/efectos adversos , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/tratamiento farmacológico , Infecciones por Papillomavirus/inmunología , Vacunas contra Papillomavirus/administración & dosificación , Vacunas contra Papillomavirus/efectos adversos , Factores de Tiempo , Resultado del Tratamiento , Infecciones Tumorales por Virus/complicaciones , Infecciones Tumorales por Virus/tratamiento farmacológico , Infecciones Tumorales por Virus/inmunología , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/patología , Displasia del Cuello del Útero/inmunología , Displasia del Cuello del Útero/patología
5.
Blood ; 99(12): 4568-77, 2002 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12036890

RESUMEN

Several patients with clinical features of chronic myeloid leukemia (CML) have fusion of the TEL (ETV6) gene on 12p13 with ABL on 9q34 and express a chimeric Tel-Abl protein that contains the same portion of the Abl tyrosine kinase fused to Tel, an Ets family transcription factor, rather than Bcr. In a murine retroviral bone marrow transduction-transplantation model, a Tel (exon 1-5)-Abl fusion protein induced 2 distinct illnesses: a CML-like myeloproliferative disease very similar to that induced by Bcr-Abl but with increased latency and a novel syndrome characterized by small-bowel myeloid cell infiltration and necrosis, increased circulating endotoxin and tumor necrosis factor alpha levels, and fulminant hepatic and renal failure. Induction of both diseases required the Tel pointed homology oligomerization domain and Abl tyrosine kinase activity. Myeloid cells from mice with both diseases expressed Tel-Abl protein. CML-like disease induced by Tel-Abl and Bcr-Abl was polyclonal and originated from cells with multilineage (myeloid, erythroid, and B- and T-lymphoid) repopulating ability and the capacity to generate day-12 spleen colonies in secondary transplantations. In contrast to findings with Bcr-Abl, however, neither Tel-Abl-induced disease could be adoptively transferred to irradiated secondary recipient syngeneic mice. These results show that Tel-Abl has leukemogenic properties from distinct from those of Bcr-Abl and may act in a different bone marrow progenitor.


Asunto(s)
Trastornos Mieloproliferativos/etiología , Trastornos Mieloproliferativos/patología , Proteínas de Fusión Oncogénica/efectos adversos , Proteínas Tirosina Quinasas/efectos adversos , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Cromosomas Humanos Par 12 , Cromosomas Humanos Par 9 , Femenino , Proteínas de Fusión bcr-abl/efectos adversos , Proteínas de Fusión bcr-abl/genética , Humanos , Enfermedades Intestinales/etiología , Enfermedades Intestinales/mortalidad , Leucemia Mielógena Crónica BCR-ABL Positiva/etiología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Ratones Endogámicos BALB C , Trastornos Mieloproliferativos/mortalidad , Trasplante de Neoplasias/efectos adversos , Trasplante de Neoplasias/mortalidad , Trasplante de Neoplasias/patología , Proteínas de Fusión Oncogénica/genética , Proteínas Tirosina Quinasas/genética , Tasa de Supervivencia , Transducción Genética , Translocación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA