Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cancer Res ; 67(20): 9887-93, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17942920

RESUMO

Amplification and overexpression of erbB2 (Her-2/neu) proto-oncogene has been linked to human malignancies including tumors of the breast, ovary, and stomach. It has been implicated in tumor growth, sensitivity to standard chemotherapy, prognosis of patients, and disease-free survival. Although the clinical use of trastuzumab (Herceptin) has prolonged the survival of breast cancer patients with erbB2-overexpressing tumors, there is an urgent need for more potent and orally bioavailable small-molecule inhibitors. CP-724,714 is a potent inhibitor of erbB2 receptor autophosphorylation in intact cells and is currently undergoing phase I clinical trials. Here, we describe the effects of CP-724,714 in vitro and in vivo in human breast cancer models. CP-724,714 is selective for inhibiting growth of HER2-driven cell lines. In addition, we show that it induces G1 cell cycle block in erbB2-overexpressing BT-474 human breast carcinoma cells and inhibits erbB2 autophosphorylation in xenografts when administered p.o. to athymic mice. It induces a marked reduction of extracellular signal-regulated kinase and Akt phosphorylation, tumor cell apoptosis, and release of caspase-3. P.o. administration (q.d. or b.i.d.) of CP-724,714 inhibits the growth of erbB2-overexpressing tumors in athymic mice without overt adverse effects.


Assuntos
Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Camundongos , Camundongos Nus , Células NIH 3T3 , Fosforilação/efeitos dos fármacos , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor ErbB-2/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Radiology ; 243(1): 105-11, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17329684

RESUMO

PURPOSE: To prospectively evaluate the ability of macromolecular contrast medium (MMCM)-enhanced dynamic magnetic resonance (MR) imaging to depict vascular changes in response to cyclooxygenase-2 (COX-2) inhibition of angiogenesis in a human breast cancer model. MATERIALS AND METHODS: The institutional committee for animal research approved this study. A human breast cancer cell line, MDA-MB-231, was implanted in 30 female homozygotous athymic rats that were alternately assigned to either a drug treatment group that received celecoxib on a daily basis for 7 days or a control group that received saline. Each animal underwent MR imaging after intravenous administration of a high-molecular-weight contrast agent at baseline and again 24 hours and 7 days after administration. Eleven rats in each group successfully underwent all three studies and had data sets of sufficient technical quality. A bidirectional two-compartment tissue model was used to estimate transendothelial permeability (K(PS)) and fractional plasma volume (fPV) for each tumor. Microvessel density was also measured to enable histologic assessment of angiogenesis. Repeated-measures analysis of variance and unpaired two-tailed t tests were used to evaluate differences in mean values between MR examinations performed in the same rats and between baseline values in treated and control rats, respectively. RESULTS: MR imaging-assayed microvascular K(PS) decreased significantly after 7 days of treatment with celecoxib (P < .05), but it was not significantly changed after 7 days in the control group. Likewise, microvascular density, a histologic surrogate of angiogenesis, was significantly (P < .05) lower in the treatment group than in the control group. The fPV did not significantly change in either group. CONCLUSION: Dynamic MR imaging revealed microvascular permeability to a high-molecular-weight contrast agent was significantly reduced by treatment with celecoxib.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Neoplasias da Mama/irrigação sanguínea , Inibidores de Ciclo-Oxigenase/uso terapêutico , Imageamento por Ressonância Magnética , Neovascularização Patológica/prevenção & controle , Pirazóis/uso terapêutico , Sulfonamidas/uso terapêutico , Animais , Neoplasias da Mama/metabolismo , Celecoxib , Linhagem Celular Tumoral , Meios de Contraste , Modelos Animais de Doenças , Feminino , Humanos , Estudos Prospectivos , Ratos , Ratos Nus
3.
Bioinformatics ; 22(16): 2028-36, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16809386

RESUMO

MOTIVATION: Although numerous methods have been developed to better capture biological information from microarray data, commonly used single gene-based methods neglect interactions among genes and leave room for other novel approaches. For example, most classification and regression methods for microarray data are based on the whole set of genes and have not made use of pathway information. Pathway-based analysis in microarray studies may lead to more informative and relevant knowledge for biological researchers. RESULTS: In this paper, we describe a pathway-based classification and regression method using Random Forests to analyze gene expression data. The proposed methods allow researchers to rank important pathways from externally available databases, discover important genes, find pathway-based outlying cases and make full use of a continuous outcome variable in the regression setting. We also compared Random Forests with other machine learning methods using several datasets and found that Random Forests classification error rates were either the lowest or the second-lowest. By combining pathway information and novel statistical methods, this procedure represents a promising computational strategy in dissecting pathways and can provide biological insight into the study of microarray data. AVAILABILITY: Source code written in R is available from http://bioinformatics.med.yale.edu/pathway-analysis/rf.htm.


Assuntos
Biologia Computacional/métodos , Perfilação da Expressão Gênica/métodos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Reconhecimento Automatizado de Padrão , Algoritmos , Animais , Simulação por Computador , Interpretação Estatística de Dados , Humanos , Camundongos , Modelos Biológicos , Modelos Estatísticos , Ratos , Análise de Regressão , Software
4.
Toxicol Pathol ; 34(1): 27-32, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16507541

RESUMO

Acute vascular injury that leads to vascular inflammation is a common finding in the preclinical toxicity testing of drugs in rats and dogs. However, the relevance of this finding for risk to humans is unclear. Concern about the safety of these drugs is heightened by the current lack of noninvasive clinical methods to predict the onset of vascular damage in animals or humans. Determining the relevance of this poorly understood preclinical outcome for humans requires a better understanding of the molecular mechanisms of injury in addition to the development of sensitive and specific leading biomarkers for the clinical diagnosis of acute vascular damage. Most molecular research on this toxicity has been performed in rats, but recent development of canine gene expression microarrays makes transcriptomic studies now possible in the dog. In this study, we investigated the molecular mechanisms of drug-induced vascular injury in dogs using gene arrays. After treating Beagles with toxic doses of CI-947, an adenosine receptor agonist, we profiled gene expression in the coronary arteries and correlated those changes with histopathology at 16 and 24 hours after dosing. The results demonstrated that pathobiological processes such as stimulation of the innate immune response, increased extracellular matrix turnover and oxidative stress were active at times of very early injury.


Assuntos
Adenosina/análogos & derivados , Perfilação da Expressão Gênica/métodos , Agonistas do Receptor Purinérgico P1 , Testes de Toxicidade/métodos , Vasculite/induzido quimicamente , Doença Aguda , Adenosina/toxicidade , Administração Oral , Animais , Biomarcadores , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Cães , Relação Dose-Resposta a Droga , Expressão Gênica/efeitos dos fármacos , Genômica , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Vasculite/genética , Vasculite/patologia
5.
Eur Radiol ; 15(11): 2268-75, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16012822

RESUMO

Carboxymethyldextran (CMD)-A2-Gd-DOTA, a slow clearance blood pool contrast agent with a molecular weight of 52.1 kDa, designed to have intravascular residence for more than 1 h, was evaluated for its potential to characterize and differentiate the microvessels of malignant and benign breast tumors. Precontrast single-slice inversion-recovery snapshot FLASH and dynamic contrast-enhanced MRI using an axial T1-weighted three-dimensional spoiled gradient recalled sequence was performed in 30 Sprague-Dawley rats with chemically induced breast tumors. Endothelial transfer coefficient and fractional plasma volume of the breast tumors were estimated from MRI data acquired with CMD-A2-Gd-DOTA enhancement injected at a dose of 0.1 mmol Gd/kg body weight using a two-compartment bidirectional model of the tumor tissue. The correlation between MRI microvessel characteristics and histopathological tumor grade was determined using the Scarff-Bloom-Richardson method. Using CMD-A2-Gd-DOTA, no significant correlations were found between the MR-estimated endothelial transfer coefficient or plasma volumes with histological tumor grade. Analysis of CMD-A2-Gd-DOTA-enhanced MR kinetic data failed to demonstrate feasibility for the differentiation of benign from malignant tumors or for image-based tumor grading.


Assuntos
Imageamento por Ressonância Magnética , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/patologia , Compostos Organometálicos , Animais , Microcirculação , Ratos , Ratos Sprague-Dawley
6.
Invest Radiol ; 40(6): 349-54, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15905721

RESUMO

OBJECTIVES: The objectives of this study were to determine if magnetic resonance (MR) estimates of quantitative tissue microvascular characteristics from regions of interest (ROI) limited to the tumor periphery provided a better correlation with tumor histologic grade than ROI defined for the whole tumor in cross-section. METHODS: A metaanalysis was based on 98 quantitative MR image breast tumor characterizations acquired in 3 separate experimental studies using identical methods for tumor induction and contrast enhancement. RESULTS: The endothelial transfer coefficient (K) of albumin (Gd-DTPA)30 from the tumor periphery correlated (r = 0.784) significantly more strongly (P < 0.001) with the pathologic tumor grade than K derived from the whole tumor (r = 0.604). K estimates, either from the tumor periphery or from the whole tumor, correlated significantly more strongly with histologic grade (P < 0.01) than MR image estimates of fractional plasma volume (fPV) from either tumor periphery (r = 0.368) or whole tumor (r = 0.323). CONCLUSIONS: K estimates from the tumor periphery were the best of these measurable MR image microvascular characteristics for predicting the histologic grade.


Assuntos
Imageamento por Ressonância Magnética/métodos , Neoplasias Mamárias Experimentais/diagnóstico , Animais , Meios de Contraste , Ratos , Ratos Sprague-Dawley , Estudos Retrospectivos
7.
Toxicol Pathol ; 32 Suppl 1: 106-15, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15209410

RESUMO

Successful development and use of biomarkers will improve the productivity of oncology drug development. Recognition of the importance of biomarkers for speeding drug development is reflected in the precise definitions and concepts proposed by an NIH Working Group to standardize terminology and promote a more coherent and systematic approach to the development and use of biomarkers. Potential clinical biomarkers of drug efficacy are often identified through pre-clinical studies or basic research. Identification of potential biomarkers for use in oncology is moving rapidly forward through continuing advances in clinical imaging technologies, especially molecular and functional imaging. Other rapid advances are a product of the growing availability of new scientific reagents for established technologies and of high-throughput genomic and proteomic technologies that can generate hundreds of potential biomarkers for further evaluation. In certain cases, conventional clinical diagnostic techniques or assays can be adapted for use in pre-clinical models to evaluate their ability to serve as biomarkers for predicting clinical responses to new drug candidates. Evaluation (pre-clinical and clinical) of a potential biomarker is often the longest stage of biomarker development, and standards for evaluation or validation depend on the intended use and stage of clinical development. Biomarkers verified for use in preclinical studies can be used to help select appropriate animal models and lead compounds. Biomarkers verified for use in clinical trials can confirm a drug's pharmacological or biological mechanism of action, guide protocol design, aid patient and dose selection, and help to minimize safety risks. Oncology drug development can be optimized by using a tiered set of clinical biomarkers that predict compound efficacy and safety with increasing confidence at each rise in tier thereby aiding corporate decision-making about advancing compounds. In oncology, a special class of extensively evaluated biomarkers of efficacy (surrogate endpoints) that generally correlate with desired clinical outcomes can be used as a basis for corporate decisions as well as for gaining accelerated provisional regulatory approval of a drug.


Assuntos
Anticarcinógenos/uso terapêutico , Biomarcadores Tumorais/genética , Desenho de Fármacos , Neoplasias/tratamento farmacológico , Animais , Genômica , Humanos , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Modelos Biológicos , Proteômica , Tomografia Computadorizada de Emissão
8.
Cancer Res ; 63(21): 7301-9, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14612527

RESUMO

Signaling through vascular endothelial growth factor (VEGF) receptors (VEGFRs) is a key pathway initiating endothelial cell proliferation and migration resulting in angiogenesis, a requirement for human tumor growth and metastasis. Abrogation of signaling through VEGFR by a variety of approaches has been demonstrated to inhibit angiogenesis and tumor growth. Small molecule inhibitors of VEGFR tyrosine kinase have been shown to inhibit angiogenesis, inhibit tumor growth, and prevent metastases. Our goal was to discover and characterize an p.o. active VEGFR-2 small molecule inhibitor. A novel isothiazole, CP-547,632, was identified as a potent inhibitor of the VEGFR-2 and basic fibroblast growth factor (FGF) kinases (IC(50) = 11 and 9 nM, respectively). It is selective relative to epidermal growth factor receptor, platelet-derived growth factor beta, and other related TKs. It also inhibits VEGF-stimulated autophosphorylation of VEGFR-2 in a whole cell assay with an IC(50) value of 6 nM. After oral administration of CP-547,632 to mice bearing NIH3T3/H-ras tumors, VEGFR-2 phosphorylation in tumors was inhibited in a dose-dependent fashion (EC(50) = 590 ng/ml). These plasma concentrations correlated well with the observed concentrations of the compound necessary to inhibit VEGF-induced corneal angiogenesis in BALB/c mice. A sponge angiogenesis assay was used to directly compare the inhibitory activities of CP-547,632 against FGF receptor 2 or VEGFR-2; this compound potently inhibits both basic FGF and VEGF-induced angiogenesis in vivo. The antitumor efficacy of this agent was evaluated after once daily p.o. administration to athymic mice bearing human xenografts and resulted in as much as 85% tumor growth inhibition. CP-547,632 is a well-tolerated, orally-bioavailable inhibitor presently under clinical investigation for the treatment of human malignancies.


Assuntos
Inibidores Enzimáticos/farmacologia , Tiazóis/farmacologia , Ureia/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Inibidores Enzimáticos/farmacocinética , Feminino , Humanos , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Nus , Células NIH 3T3 , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Ureia/análogos & derivados , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Eur J Nucl Med Mol Imaging ; 30(3): 448-55, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12722742

RESUMO

The aim of this study was to evaluate the potential of dynamic magnetic resonance imaging (MRI) enhanced by macromolecular contrast agents to monitor noninvasively the therapeutic effect of an anti-angiogenesis VEGF receptor kinase inhibitor in an experimental cancer model. MDA-MB-435, a poorly differentiated human breast cancer cell line, was implanted into the mammary fat pad in 20 female homozygous athymic rats. Animals were assigned randomly to a control (n=10) or drug treatment group (n=10). Baseline dynamic MRI was performed on sequential days using albumin-(GdDTPA)30 (6.0 nm diameter) and ultrasmall superparamagnetic iron oxide (USPIO) particles (approximately 30 nm diameter). Subjects were treated either with PTK787/ZK 222584, a VEGF receptor tyrosine kinase inhibitor, or saline given orally twice daily for 1 week followed by repeat MRI examinations serially using each contrast agent. Employing a unidirectional kinetic model comprising the plasma and interstitial water compartments, tumor microvessel characteristics including fractional plasma volume and transendothelial permeability (K(PS)) were estimated for each contrast medium. Tumor growth and the microvascular density, a histologic surrogate of angiogenesis, were also measured. Control tumors significantly increased (P<0.05) in size and in microvascular permeability (K(PS)) based on MRI assays using both macromolecular contrast media. In contrast, tumor growth was significantly reduced (P<0.05) in rats treated with PTK787/ZK 222584 and K(PS) values declined slightly. Estimated values for the fractional plasma volume did not differ significantly between treatment groups or contrast agents. Microvascular density counts correlated fairly with the tumor growth rate (r=0.64) and were statistically significant higher (P<0.05) in the control than in the drug-treated group. MRI measurements of tumor microvascular response, particularly transendothelial permeability (K(PS)), using either of two macromolecular contrast media, were able to detect effects of treatment with a VEGF receptor tyrosine kinase inhibitor on tumor vascular permeability. In a clinical setting such quantitative MRI measurements could be used to monitor tumor anti-angiogenesis therapy.


Assuntos
Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/diagnóstico , Imageamento por Ressonância Magnética/métodos , Neovascularização Patológica/diagnóstico , Neovascularização Patológica/tratamento farmacológico , Ftalazinas/uso terapêutico , Piridinas , Inibidores da Angiogênese/uso terapêutico , Animais , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Feminino , Humanos , Transplante de Neoplasias , Valor Preditivo dos Testes , Ratos , Resultado do Tratamento
12.
Toxicol Pathol ; 30(1): 147-56, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11890468

RESUMO

ILSI-HESI sponsored an international consortium for the evaluation of alternative models, including the TrpS3+/- mouse. for use in short-term carcinogenicity testing of pharmaceuticals. Products of the ILSI evaluation included guidance for protocol design and assay interpretation, spontaneous tumor incidences, diagnostic criteria for common proliferative lesions, and results of assays for pharmaceutical agents that are known human and/or rodent carcinogens and non-carcinogens. Based on the ILSI evaluation, recommended protocol elements for this model include: 26-week study duration, groups > or = 15/sex/dose, a positive control group (benzene or p-cresidine), a negative control group and 3 dose groups, the high dose set at MTD or MFD, routine in-life evaluations, and complete necropsies with microscopic evaluation of tissues. Favored statistical analyses are trend tests or pair-wise comparisons, with no adjustments for survival. For an assay to be valid, positive control groups must demonstrate an effect, and the MTD or MFD must be reached in both sexes. Criteria for a negative response include a valid assay, no statistical increase in common tumors, no biologically significant numerical increase in rare tumors, and no tumor incidence above that of historical controls. Positive responses can consist of statistically significant increases in the incidence of a common tumor or numerical increases in a rare tumor, which may not be statistically significant. In either case, the incidence should be clearly above historical control values. Evidence of a dose response or occurrence of hyperplasia in a tissue with a neoplastic response can support interpreting an assay as positive. The two most common spontaneous tumors (> 1 %) in Trp53+/- mice are malignant thymic lymphomas and subcutaneous sarcomas. Use of implanted electronic transponders can increase the incidence of sarcomas. Important rare spontaneous tumors (incidence < or = 1%) are osteosarcomas and pulmonary adenomas. Many other tumor types have been reported to occur sporadically in Trp53+/- mice. Diagnostic challenges for this model include differentiating lymphoma from atypical thymic hyperplasia and recognizing the variable histopathology of subcutaneous sarcomas. In reported bioassays, Trp53+/- mice responded positively to genotoxic carcinogens, negatively to non-genotoxic rodent carcinogens, and negatively to noncarcinogens, indicating that unlike the 2-year mouse assay, this short-term assay is not overly sensitive. Positive responses often elicited an increase in tumors that occur spontaneously. To successfully use this model, pathologists must understand the biology of the Trp53 tumor suppressor gene and the principles of protocol design and data interpretation for short-term bioassays. They must also know the historical response pattern of Trp53+/- mice to test agents and be able to accurately diagnose tumors in this model. Use of the Trp53+/- mouse presents the pharmaceutical industry with several challenges, one of which is managing the uncertainty created by a lack of precedents for regulatory decisions about some possible outcomes for short-term carcinogenicity assays.


Assuntos
Testes de Carcinogenicidade/métodos , Genes Supressores de Tumor , Genes p53/genética , Camundongos Transgênicos/fisiologia , Patologia/métodos , Farmacologia/métodos , Animais , Bioensaio , Indústria Farmacêutica , Camundongos , Neoplasias/induzido quimicamente , Neoplasias/epidemiologia , Neoplasias/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA