Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Cancer Ther ; 20(7): 1270-1282, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33879555

RESUMO

The cell surface glycoprotein P-cadherin is highly expressed in a number of malignancies, including those arising in the epithelium of the bladder, breast, esophagus, lung, and upper aerodigestive system. PCA062 is a P-cadherin specific antibody-drug conjugate that utilizes the clinically validated SMCC-DM1 linker payload to mediate potent cytotoxicity in cell lines expressing high levels of P-cadherin in vitro, while displaying no specific activity in P-cadherin-negative cell lines. High cell surface P-cadherin is necessary, but not sufficient, to mediate PCA062 cytotoxicity. In vivo, PCA062 demonstrated high serum stability and a potent ability to induce mitotic arrest. In addition, PCA062 was efficacious in clinically relevant models of P-cadherin-expressing cancers, including breast, esophageal, and head and neck. Preclinical non-human primate toxicology studies demonstrated a favorable safety profile that supports clinical development. Genome-wide CRISPR screens reveal that expression of the multidrug-resistant gene ABCC1 and the lysosomal transporter SLC46A3 differentially impact tumor cell sensitivity to PCA062. The preclinical data presented here suggest that PCA062 may have clinical value for treating patients with multiple cancer types including basal-like breast cancer.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Biomarcadores Tumorais , Caderinas/genética , Imunoconjugados/farmacologia , Neoplasias/genética , Sequência de Aminoácidos , Animais , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antineoplásicos Imunológicos/química , Antineoplásicos Imunológicos/farmacocinética , Sítios de Ligação , Caderinas/química , Caderinas/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos , Expressão Gênica , Humanos , Imunoconjugados/química , Imunoconjugados/farmacocinética , Imuno-Histoquímica , Macaca fascicularis , Camundongos , Modelos Moleculares , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Ligação Proteica , Transporte Proteico , Ratos , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
2.
J Pathol ; 232(2): 103-11, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24122209

RESUMO

We need improved, translatable and predictive tumour models for the evaluation of response and the evolution of resistance to targeted therapeutics. We provide a review of the use of different types of preclinical tumour models to evaluate novel anticancer agents, and model the rapidly evolving landscape of resistance to targeted therapy. We focus on describing the various preclinical models available for candidate drug development and design considerations for preclinical experiments, depending on the aspect of drug action being interrogated. We discuss selected examples of how experimental findings have translated into clinical outcomes for targeted agents, predicted mechanisms that drive resistance and strategies to overcome the evolution thereof. We discuss challenges in preclinical experimental design and interpretation and possible improvements in animal models of therapeutic response and resistance, with an emphasis on improved translation of experimental research into clinical practice.


Assuntos
Antineoplásicos/farmacologia , Descoberta de Drogas/métodos , Resistencia a Medicamentos Antineoplásicos , Neoplasias Experimentais/tratamento farmacológico , Animais , Biomarcadores Tumorais/metabolismo , Humanos , Camundongos , Terapia de Alvo Molecular , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Transdução de Sinais/efeitos dos fármacos , Pesquisa Translacional Biomédica , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Nature ; 494(7436): 251-5, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-23302800

RESUMO

Mutational activation of BRAF is the most prevalent genetic alteration in human melanoma, with ≥50% of tumours expressing the BRAF(V600E) oncoprotein. Moreover, the marked tumour regression and improved survival of late-stage BRAF-mutated melanoma patients in response to treatment with vemurafenib demonstrates the essential role of oncogenic BRAF in melanoma maintenance. However, as most patients relapse with lethal drug-resistant disease, understanding and preventing mechanism(s) of resistance is critical to providing improved therapy. Here we investigate the cause and consequences of vemurafenib resistance using two independently derived primary human melanoma xenograft models in which drug resistance is selected by continuous vemurafenib administration. In one of these models, resistant tumours show continued dependency on BRAF(V600E)→MEK→ERK signalling owing to elevated BRAF(V600E) expression. Most importantly, we demonstrate that vemurafenib-resistant melanomas become drug dependent for their continued proliferation, such that cessation of drug administration leads to regression of established drug-resistant tumours. We further demonstrate that a discontinuous dosing strategy, which exploits the fitness disadvantage displayed by drug-resistant cells in the absence of the drug, forestalls the onset of lethal drug-resistant disease. These data highlight the concept that drug-resistant cells may also display drug dependency, such that altered dosing may prevent the emergence of lethal drug resistance. Such observations may contribute to sustaining the durability of the vemurafenib response with the ultimate goal of curative therapy for the subset of melanoma patients with BRAF mutations.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Indóis/administração & dosagem , Indóis/efeitos adversos , Melanoma/tratamento farmacológico , Melanoma/patologia , Sulfonamidas/administração & dosagem , Sulfonamidas/efeitos adversos , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Esquema de Medicação , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Indóis/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melanoma/genética , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mutação , Transplante de Neoplasias , Proteínas Proto-Oncogênicas B-raf/química , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Tela Subcutânea , Sulfonamidas/farmacologia , Fatores de Tempo , Vemurafenib , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Mol Cancer Ther ; 11(3): 730-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22246440

RESUMO

A novel oral Hsp90 inhibitor, NVP-HSP990, has been developed and characterized in vitro and in vivo. In vitro, NVP-HSP990 exhibits single digit nanomolar IC(50) values on three of the Hsp90 isoforms (Hsp90α, Hsp90ß, and GRP94) and 320 nanomolar IC(50) value on the fourth (TRAP-1), with selectivity against unrelated enzymes, receptors, and kinases. In c-Met amplified GTL-16 gastric tumor cells, NVP-HSP990 dissociated the Hsp90-p23 complex, depleted client protein c-Met, and induced Hsp70. NVP-HSP990 potently inhibited the growth of human cell lines and primary patient samples from a variety of tumor types. In vivo, NVP-HSP990 exhibits drug-like pharmaceutical and pharmacologic properties with high oral bioavailability. In the GTL-16 xenograft model, a single oral administration of 15 mg/kg of NVP-HSP990 induced sustained downregulation of c-Met and upregulation of Hsp70. In repeat dosing studies, NVP-HSP990 treatment resulted in tumor growth inhibition of GTL-16 and other human tumor xenograft models driven by well-defined oncogenic Hsp90 client proteins. On the basis of its pharmacologic profile and broad-spectrum antitumor activities, clinical trials have been initiated to evaluate NVP-HSP990 in advanced solid tumors.


Assuntos
Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Piridonas/farmacologia , Pirimidinas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Administração Oral , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Área Sob a Curva , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Concentração Inibidora 50 , Taxa de Depuração Metabólica , Camundongos , Camundongos Nus , Camundongos SCID , Estrutura Molecular , Neoplasias/metabolismo , Neoplasias/patologia , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/metabolismo , Piridonas/administração & dosagem , Piridonas/farmacocinética , Pirimidinas/administração & dosagem , Pirimidinas/farmacocinética , Carga Tumoral/efeitos dos fármacos
5.
Clin Cancer Res ; 12(16): 4908-15, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16914579

RESUMO

PURPOSE: The ectopically expressed and deregulated fibroblast growth factor receptor 3 (FGFR3) results from a t(4;14) chromosomal translocation that occurs in approximately 15% of multiple myeloma (MM) patients and confers a particularly poor prognosis. This study assesses the antimyeloma activity of CHIR-258, a small-molecule inhibitor of multiple receptor tyrosine kinases that is currently in phase I trials, in a newly developed FGFR3-driven preclinical MM animal model. EXPERIMENTAL DESIGN: We developed an orthotopic MM model in mice using a luciferase-expressing human KMS-11-luc line that expresses mutant FGFR3 (Y373C). The antimyeloma activity of CHIR-258 was evaluated at doses that inhibited FGFR3 signaling in vivo in this FGFR3-driven animal model. RESULTS: Noninvasive bioluminescence imaging detected MM lesions in nearly all mice injected with KMS-11-luc cells, which were mainly localized in the spine, skull, and pelvis, resulting in frequent development of paralysis. Daily oral administration of CHIR-258 at doses that inhibited FGFR3 signaling in KMS-11-luc tumors in vivo resulted in a significant inhibition of KMS-11-luc tumor growth, which translated into a significant improvement in animal survival. CONCLUSIONS: Our data provide a relevant preclinical basis for clinical trials of CHIR-258 in FGFR3-positive MM patients.


Assuntos
Benzimidazóis/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Quinolonas/farmacologia , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Humanos , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Camundongos , Camundongos SCID , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mieloma Múltiplo/enzimologia , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Quinases/antagonistas & inibidores , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/sangue , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Mol Cancer Ther ; 3(11): 1427-38, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15542782

RESUMO

PD 0332991 is a highly specific inhibitor of cyclin-dependent kinase 4 (Cdk4) (IC50, 0.011 micromol/L) and Cdk6 (IC50, 0.016 micromol/L), having no activity against a panel of 36 additional protein kinases. It is a potent antiproliferative agent against retinoblastoma (Rb)-positive tumor cells in vitro, inducing an exclusive G1 arrest, with a concomitant reduction of phospho-Ser780/Ser795 on the Rb protein. Oral administration of PD 0332991 to mice bearing the Colo-205 human colon carcinoma produces marked tumor regression. Therapeutic doses of PD 0332991 cause elimination of phospho-Rb and the proliferative marker Ki-67 in tumor tissue and down-regulation of genes under the transcriptional control of E2F. The results indicate that inhibition of Cdk4/6 alone is sufficient to cause tumor regression and a net reduction in tumor burden in some tumors.


Assuntos
Antineoplásicos/farmacologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Piridinas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quinase 4 Dependente de Ciclina , Quinase 6 Dependente de Ciclina , Quinases Ciclina-Dependentes/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Fase G1/efeitos dos fármacos , Humanos , Camundongos , Estrutura Molecular , Transplante de Neoplasias/patologia , Neoplasias/metabolismo , Neoplasias/patologia , Fosforilação/efeitos dos fármacos , Piperazinas/química , Inibidores de Proteínas Quinases/química , Proteínas Proto-Oncogênicas/metabolismo , Piridinas/química , Proteína do Retinoblastoma/metabolismo , Especificidade por Substrato
7.
Clin Cancer Res ; 9(15): 5729-34, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14654558

RESUMO

PURPOSE: The purpose of this study was to evaluate the effect of the receptor tyrosine kinase inhibitor SU11654 on the activity of its molecular target KIT in canine mast cell tumors (MCT) and correlate target inhibition with mutational status of the c-kit juxtamembrane domain and SU11654 plasma concentration. EXPERIMENTAL DESIGN: Tumor biopsies were obtained from dogs with advanced MCTs before and 8 h after administration of a single oral dose of SU11654, previously shown to be active in dogs with MCTs. Blood samples were taken to determine the plasma concentration of SU11654. Levels of phosphorylated KIT and ERK1/2 were assessed in tumor biopsies by Western blot. Tumors were analyzed by PCR for the presence or absence of an internal tandem duplication (ITD) in the juxtamembrane domain of c-kit. RESULTS: Fourteen dogs with advanced MCTs were enrolled in the study; 11 of these were evaluable for KIT target modulation (the remaining tumor specimens had inevaluable amounts of total KIT protein). Of these, eight MCTs showed reduced levels of phosphorylated KIT relative to total KIT after treatment with SU11654, compared with pretreatment biopsies. All four evaluable MCTs expressing ITD mutant c-kitshowed modulation of KIT phosphorylation, as did four of seven tumors expressing non-ITD c-kit. Phosphorylated ERK1/2 was modulated in seven tumors; this did not correlate with inhibition of KIT phosphorylation CONCLUSION: SU11654 treatment at the efficacious dose results in inhibition of KIT phosphorylation in canine MCTs.


Assuntos
Doenças do Cão/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Indóis/uso terapêutico , Sarcoma de Mastócitos/veterinária , Proteínas Proto-Oncogênicas c-kit/metabolismo , Pirróis/uso terapêutico , Animais , Doenças do Cão/patologia , Cães , Inibidores Enzimáticos/sangue , Inibidores Enzimáticos/farmacocinética , Indóis/sangue , Indóis/farmacocinética , Mastócitos , Sarcoma de Mastócitos/tratamento farmacológico , Sarcoma de Mastócitos/patologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Metástase Neoplásica , Fosforilação , Pirróis/sangue , Pirróis/farmacocinética , Recidiva
8.
Mol Cancer Ther ; 2(10): 1011-21, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14578466

RESUMO

SU11248 is an oral multitargeted tyrosine kinase inhibitor with antitumor and antiangiogenic activities through targeting platelet-derived growth factor receptor, vascular endothelial growth factor receptor, KIT, and FLT3, the first three of which are expressed in human breast cancer and/or its supporting tissues. The purpose of the present studies was to demonstrate the potent anticancer activity of SU11248 alone or in combination with conventional cytotoxic agents against several distinct preclinical models of breast cancer. SU11248 was administered as a monotherapy to (1) mouse mammary tumor virus-v-Ha-ras mice and 7,12-dimethylbenz(a)anthracene-treated rats bearing mammary tumors and (2) mice bearing human breast cancer xenografts of s.c. MX-1 tumors and osseous metastasis of a MDA-MB-435-derived cell line (435/HAL-Luc). SU11248 was also administered in combination with docetaxel both in xenograft models and in combination with 5-fluorouracil and doxorubicin in the MX-1 model. SU11248 treatment potently regressed growth of mammary cancers in mouse mammary tumor virus-v-Ha-ras transgenic mice (82% regression) and 7,12-dimethylbenz(a)anthracene-induced mammary tumors in rats (99% regression at the highest dose; P < 0.05 for both). This agent also inhibited MX-1 tumor growth by 52%, with markedly enhanced anticancer effects when administered in combination with docetaxel, 5-fluorouracil, or doxorubicin compared with either agent alone (P < 0.05). SU11248 treatment in combination with docetaxel effectively prolonged survival of mice, with 435/HAL-Luc cancer xenografts established in bone compared with either agent alone (P < 0.05). These results demonstrate that SU11248 is effective in preclinical breast cancer models and suggest that it may be useful in the treatment of breast cancer in the clinic.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Indóis/farmacologia , Pirróis/farmacologia , Animais , Antibióticos Antineoplásicos/farmacologia , Antibióticos Antineoplásicos/uso terapêutico , Neoplasias Ósseas/secundário , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Feminino , Fluoruracila/uso terapêutico , Humanos , Camundongos , Camundongos Nus , Camundongos Transgênicos , Transplante de Neoplasias , Fosforilação , Ratos , Ratos Sprague-Dawley , Sunitinibe , Fatores de Tempo
9.
Mol Cancer Ther ; 2(5): 471-8, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12748309

RESUMO

The purpose of this study was to evaluate the activity of the indolinone kinase inhibitor SU11248 against the receptor tyrosine kinase KIT in vitro and in vivo, examine the role of KIT in small cell lung cancer (SCLC), and anticipate clinical utility of SU11248 in SCLC. SU11248 is an oral, multitargeted tyrosine kinase inhibitor with direct antitumor and antiangiogenic activity through targeting platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor receptor, KIT, and FLT3 receptors. Treatment of the KIT-expressing SCLC-derived NCI-H526 cell line in vitro with SU11248 resulted in dose-dependent inhibition of stem cell factor-stimulated KIT phosphotyrosine levels and proliferation. The biological significance of KIT inhibition was evaluated in vivo by treating mice bearing s.c. NCI-H526 tumors with SU11248 or another structurally unrelated KIT inhibitor, STI571 (Gleevec), which is also known to inhibit Bcr-Abl and PDGFRbeta. SU11248 treatment resulted in significant tumor growth inhibition, whereas inhibition from STI571 treatment was less dramatic. Both compounds reduced phospho-KIT levels in NCI-H526 tumors, with a greater reduction by SU11248, correlating with efficacy. Likewise, phospho-PDGFRbeta levels contributed by tumor stroma and with known involvement in angiogenesis were strongly inhibited by SU11248 and less so by STI571. Because platinum-based chemotherapy is part of the standard of care for SCLC, SU11248 was combined with cisplatin, and significant tumor growth delay was measured compared with either agent alone. These results expand the profile of SU11248 as a KIT signaling inhibitor and suggest that SU11248 may have clinical potential in the treatment of SCLC via direct antitumor activity mediated via KIT as well as tumor angiogenesis via vascular endothelial growth factor receptor FLK1/KDR and PDGFRbeta.


Assuntos
Carcinoma de Células Pequenas/metabolismo , Indóis/farmacologia , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas c-kit/efeitos dos fármacos , Pirróis/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Benzamidas , Carcinoma de Células Pequenas/patologia , Divisão Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Mesilato de Imatinib , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Fosforilação , Fosfotirosina/metabolismo , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Pirimidinas/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de Células-Tronco/fisiologia , Sunitinibe , Células Tumorais Cultivadas/transplante
10.
Clin Exp Metastasis ; 20(8): 757-66, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14713109

RESUMO

The aim of the study was to investigate inhibitory effects of the receptor tyrosine kinase (RTK) inhibitor SU11248 against CSF-1R and osteoclast (OC) formation. We developed an in vivo model of breast cancer metastasis to evaluate efficacy of SU11248 against tumor growth and tumor-induced osteolysis in bone. The in vitro effects of SU11248 on CSF-1R phosphorylation, OC formation and function were evaluated. Effects on 435/HAL-Luc tumor growth in bone were monitored by in vivo bioluminescence imaging (BLI), and inhibition of osteolysis was evaluated by measurement of serum pyridinoline (PYD) concentration and histology. Phosphorylation of the receptor for M-CSF (CSF-1R) expressed by NIH3T3 cells was inhibited by SU11248 with an IC50 of 50-100 nM, consistent with CSF-1R belonging to the class III split kinase domain RTK family. The early M-CSF-dependent phase of in vitro murine OC development and function were inhibited by SU11248 at 10-100 nM. In vivo inhibition of osteolysis was confirmed by significant lowering of serum PYD levels following SU11248 treatment of tumor-bearing mice (P = 0.047). Using BLI, SU11248 treatment at 40 mg/kg/day for 21 days showed 64% inhibition of tumor growth in bone (P = 0.006), and at 80 mg/kg/day showed 89% inhibition (P = 0.001). Collectively, these data suggest that SU11248 may be an effective and tolerated therapy to inhibit growth of breast cancer bone metastases, with the additional advantage of inhibiting tumor-associated osteolysis.


Assuntos
Neoplasias Ósseas/patologia , Neoplasias Ósseas/fisiopatologia , Indóis/farmacologia , Osteólise , Pirróis/farmacologia , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Animais , Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Adesão Celular , Camundongos , Camundongos Nus , Transplante de Neoplasias , Osteoclastos , Fosforilação , Sunitinibe , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...