Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cancers (Basel) ; 15(2)2023 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-36672409

RESUMO

Drug resistance is a long-standing impediment to effective systemic cancer therapy and acquired drug resistance is a growing problem for molecularly-targeted therapeutics that otherwise have shown unprecedented successes in disease control. The hepatocyte growth factor (HGF)/Met receptor pathway signaling is frequently involved in cancer and has been a subject of targeted drug development for nearly 30 years. To anticipate and study specific resistance mechanisms associated with targeting this pathway, we engineered resistance to the HGF-neutralizing antibody rilotumumab in glioblastoma cells harboring autocrine HGF/Met signaling, a frequent abnormality of this brain cancer in humans. We found that rilotumumab resistance was acquired through an unusual mechanism comprising dramatic HGF overproduction and misfolding, endoplasmic reticulum (ER) stress-response signaling and redirected vesicular trafficking that effectively sequestered rilotumumab and misfolded HGF from native HGF and activated Met. Amplification of MET and HGF genes, with evidence of rapidly acquired intron-less, reverse-transcribed copies in DNA, was also observed. These changes enabled persistent Met pathway activation and improved cell survival under stress conditions. Point mutations in the HGF pathway or other complementary or downstream growth regulatory cascades that are frequently associated with targeted drug resistance in other prevalent cancer types were not observed. Although resistant cells were significantly more malignant, they retained sensitivity to Met kinase inhibition and acquired sensitivity to inhibition of ER stress signaling and cholesterol biosynthesis. Defining this mechanism reveals details of a rapidly acquired yet highly-orchestrated multisystem route of resistance to a selective molecularly-targeted agent and suggests strategies for early detection and effective intervention.

2.
Cancers (Basel) ; 14(24)2022 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-36551660

RESUMO

Two vascular endothelial growth factor (VEGF) receptors, FLT-1 and KDR, are expressed preferentially in proliferating endothelium. There is increasing evidence that recombinant, soluble VEGF receptor domains interfering with VEGF signaling may inhibit in vivo neoangiogenesis, tumor growth and metastatic spread. We hypothesized that a soluble form of FLT-1 receptor (sFLT-1) could inhibit the growth of pre-established tumors via an anti-angiogenic mechanism. A replication-deficient adenovirus (Ad) vector carrying the sflt-1 cDNA (Adsflt) was used to overexpress the sFLT-1 receptor in a breast cancer animal model. MCF-7 cells, which produce VEGF, were used to establish solid tumors in the mammary fat pads of female nude mice. After six weeks, tumors were injected either with Adsflt or a negative control virus (AdCMV.ßgal). After six months, average tumor volume in the Adsflt-infected group (33 ± 22 mm3) decreased by 91% relative to that of the negative control group (388 ± 94 mm3; p < 0.05). Moreover, 10 of 15 Adsflt-infected tumors exhibited complete regression. The vascular density of Adsflt-infected tumors was reduced by 50% relative to that of negative controls (p < 0.05), which is consistent with sFLT-1-mediated tumor regression through an anti-angiogenic mechanism. Moreover, cell necrosis and fibrosis associated with long-term regression of Adsflt−infected tumors were preceded by apoptosis of tumor vascular endothelial cells. Mice treated with Adsflt intratumorally showed no delay in the healing of cutaneous wounds, providing preliminary evidence that Ad-mediated sFLT-1 overexpression may be an effective anti-angiogenic therapy for cancer without the risk of systemic anti-angiogenic effects.

3.
PLoS One ; 16(7): e0241766, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34292953

RESUMO

Comprehensive characterizations of bladder cancer (BCa) have established molecular phenotype classes with distinct alterations and survival trends. Extending these studies within the tyrosine kinase (TK) family to identify disease drivers could improve our use of TK inhibitors to treat specific patient groups or individuals. We examined the expression distribution of TKs as a class (n = 89) in The Cancer Genome Atlas (TCGA) muscle invasive BCa data set (n >400). Patient profiles of potentially oncogenic alterations (overexpression and/or amplification) clustered TKs into 3 groups; alterations of group 1 and 3 TKs were associated with significantly worse patient survival relative to those without alterations. Many TK pathways induce epithelial-to-mesenchymal transition (EMT), which promotes tumor invasiveness and metastasis. Overexpression and/or amplification among 9 EMT transcriptional activators occurred in 43% of TCGA cases. Co-occurring alterations of TKs and EMT transcriptional activators involved most group 1 TKs; 24% of these events were associated with significantly worse patient survival. Co-occurring alterations of receptor TKs and their cognate ligands occurred in 16% of TCGA cases and several BCa-derived cell lines. Suppression of GAS6, MST1 or CSF1, or their respective receptors (AXL, MST1R and CSF1R), in BCa cell lines was associated with decreased receptor activation, cell migration, cell proliferation and anchorage independent cell growth. These studies reveal the patterns and prevalence of potentially oncogenic TK pathway-related alterations in BCa and identify specific alterations associated with reduced BCa patient survival. Detection of these features in BCa patients could better inform TK inhibitor use and improve clinical outcomes.


Assuntos
Comunicação Autócrina , Receptores Proteína Tirosina Quinases/metabolismo , Neoplasias da Bexiga Urinária/patologia , Animais , Linhagem Celular Tumoral , Bases de Dados Factuais , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Estimativa de Kaplan-Meier , Camundongos , Invasividade Neoplásica , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Transplante Heterólogo , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/mortalidade , Receptor Tirosina Quinase Axl
4.
Invest New Drugs ; 39(6): 1577-1586, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34180036

RESUMO

The vascular endothelial growth factor (VEGF)/VEGFR and hepatocyte growth factor (HGF)/c-MET signaling pathways act synergistically to promote angiogenesis. Studies indicate VEGF inhibition leads to increased levels of phosphorylated c-MET, bypassing VEGF-mediated angiogenesis and leading to chemoresistance. We conducted a phase 1 clinical trial with 32 patients with refractory solid tumors to evaluate the safety, pharmacokinetics, and pharmacodynamics of combinations of VEGF-targeting pazopanib and the putative c-MET inhibitor ARQ197 (tivantinib) at 5 dose levels (DLs). Patients either took pazopanib and tivantinib from treatment initiation (escalation phase) or pazopanib alone for 7 days, with paired tumor sampling, prior to starting combination treatment (expansion phase). Hypertension was the most common adverse event. No more than 1 dose limiting toxicity (DLT) occurred at any DL, so the maximum tolerated dose (MTD) was not determined; DL5 (800 mg pazopanib daily and 360 mg tivantinib BID) was used during the expansion phase. Twenty of 31 evaluable patients achieved stable disease lasting up to 22 cycles. Circulating VEGF, VEGFR2, HGF, and c-MET levels were assessed, and only VEGF levels increased. Tumor c-MET levels (total and phosphorylated) were determined in paired biopsies before and after 7 days of pazopanib treatment. Total intact c-MET decreased in 6 of 7 biopsy pairs, in contrast to previously reported c-MET elevation in response to VEGF inhibition. These results are discussed in the context of our previously reported analysis of epithelial-mesenchymal transition in these tumors.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Indazóis/uso terapêutico , Neoplasias/tratamento farmacológico , Pirimidinas/uso terapêutico , Pirrolidinonas/uso terapêutico , Quinolinas/uso terapêutico , Sulfonamidas/uso terapêutico , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/efeitos adversos , Inibidores da Angiogênese/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica , Área Sob a Curva , Relação Dose-Resposta a Droga , Esquema de Medicação , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Indazóis/administração & dosagem , Indazóis/efeitos adversos , Indazóis/farmacocinética , Dose Máxima Tolerável , Pessoa de Meia-Idade , Neoplasias/patologia , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Pirimidinas/administração & dosagem , Pirimidinas/efeitos adversos , Pirimidinas/farmacocinética , Pirrolidinonas/administração & dosagem , Pirrolidinonas/efeitos adversos , Pirrolidinonas/farmacocinética , Quinolinas/administração & dosagem , Quinolinas/efeitos adversos , Quinolinas/farmacocinética , Sulfonamidas/administração & dosagem , Sulfonamidas/efeitos adversos , Sulfonamidas/farmacocinética , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos
5.
Genes Chromosomes Cancer ; 60(6): 434-446, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33527590

RESUMO

Renal cell carcinoma (RCC) is not a single disease but is made up of several different histologically defined subtypes that are associated with distinct genetic alterations which require subtype specific management and treatment. Papillary renal cell carcinoma (pRCC) is the second most common subtype after conventional/clear cell RCC (ccRCC), representing ~20% of cases, and is subcategorized into type 1 and type 2 pRCC. It is important for preclinical studies to have cell lines that accurately represent each specific RCC subtype. This study characterizes seven cell lines derived from both primary and metastatic sites of type 1 pRCC, including the first cell line derived from a hereditary papillary renal carcinoma (HPRC)-associated tumor. Complete or partial gain of chromosome 7 was observed in all cell lines and other common gains of chromosomes 16, 17, or 20 were seen in several cell lines. Activating mutations of MET were present in three cell lines that all demonstrated increased MET phosphorylation in response to HGF and abrogation of MET phosphorylation in response to MET inhibitors. CDKN2A loss due to mutation or gene deletion, associated with poor outcomes in type 1 pRCC patients, was observed in all cell line models. Six cell lines formed tumor xenografts in athymic nude mice and thus provide in vivo models of type 1 pRCC. These type 1 pRCC cell lines provide a comprehensive representation of the genetic alterations associated with pRCC that will give insight into the biology of this disease and be ideal preclinical models for therapeutic studies.


Assuntos
Carcinoma de Células Renais/genética , Autenticação de Linhagem Celular/métodos , Neoplasias Renais/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Instabilidade Cromossômica , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Humanos , Neoplasias Renais/patologia , Camundongos , Mutação , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo
6.
Sci Rep ; 9(1): 2198, 2019 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-30778109

RESUMO

The increasing potential for accidental radiation exposure from either nuclear accidents or terrorist activities has escalated the need for radiation countermeasure development. We previously showed that a 30-day course of high-dose captopril, an ACE inhibitor, initiated 1-4 h after total body irradiation (TBI), improved Hematopoietic Acute Radiation Syndrome (H-ARS) and increased survival in mice. However, because of the time likely required for the deployment of a stockpiled radiation countermeasure to a radiation mass casualty site, there is a need for therapies that can be administered 24-48 hours after initial exposure. Using C57BL/6 mice exposed to an LD50-80/30 of 60Co TBI (7.75-7.9 Gy, 0.615 Gy/min), we show that low-dose captopril administration, initiated as late as 48 h post-TBI and continued for 14 days, significantly enhanced overall survival similarly to high-dose, rapid administration. Captopril treatment did not affect radiation-induced cell cycle arrest genes or the immediate loss of hematopoietic precursors. Reduced mortality was associated with the recovery of bone marrow cellularity and mature blood cell recovery at 21-30 days post-irradiation. Captopril reduced radiation-induced cytokines EPO, G-CSF, and SAA in the plasma. Finally, delayed captopril administration mitigated brain micro-hemorrhage at 21 days post-irradiation. These data indicate that low dose captopril administered as late as 48 h post-TBI for only two weeks improves survival that is associated with hematopoietic recovery and reduced inflammatory response. These data suggest that captopril may be an ideal countermeasure to mitigate H-ARS following accidental radiation exposure.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/administração & dosagem , Captopril/administração & dosagem , Hematopoese/efeitos dos fármacos , Hematopoese/efeitos da radiação , Protetores contra Radiação/administração & dosagem , Irradiação Corporal Total , Síndrome Aguda da Radiação/sangue , Síndrome Aguda da Radiação/etiologia , Síndrome Aguda da Radiação/mortalidade , Síndrome Aguda da Radiação/prevenção & controle , Animais , Contagem de Células Sanguíneas , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Citocinas/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/efeitos da radiação , Mediadores da Inflamação/metabolismo , Camundongos , Doses de Radiação , Exposição à Radiação , Tempo para o Tratamento , Irradiação Corporal Total/efeitos adversos
7.
PLoS One ; 13(6): e0199361, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29928062

RESUMO

The presence of cancer stem cells (CSCs) and the induction of epithelial-to-mesenchymal transition (EMT) in tumors are associated with tumor aggressiveness, metastasis, drug resistance, and poor prognosis, necessitating the development of reagents for unambiguous detection of CSC- and EMT-associated proteins in tumor specimens. To this end, we generated novel antibodies to EMT- and CSC-associated proteins, including Goosecoid, Sox9, Slug, Snail, and CD133. Importantly, unlike several widely used antibodies to CD133, the anti-CD133 antibodies we generated recognize epitopes distal to known glycosylation sites, enabling analyses that are not confounded by differences in CD133 glycosylation. For all target proteins, we selected antibodies that yielded the expected target protein molecular weights by Western analysis and the correct subcellular localization patterns by immunofluorescence microscopy assay (IFA); binding selectivity was verified by immunoprecipitation-mass spectrometry and by immunohistochemistry and IFA peptide blocking experiments. Finally, we applied these reagents to assess modulation of the respective markers of EMT and CSCs in xenograft tumor models by IFA. We observed that the constitutive presence of human hepatocyte growth factor (hHGF) in the tumor microenvironment of H596 non-small cell lung cancer tumors implanted in homozygous hHGF knock-in transgenic mice induced a more mesenchymal-like tumor state (relative to the epithelial-like state when implanted in control SCID mice), as evidenced by the elevated expression of EMT-associated transcription factors detected by our novel antibodies. Similarly, our new anti-CD133 antibody enabled detection and quantitation of drug-induced reductions in CD133-positive tumor cells following treatment of SUM149PT triple-negative breast cancer xenograft models with the CSC/focal adhesion kinase (FAK) inhibitor VS-6063. Thus, our novel antibodies to CSC- and EMT-associated factors exhibit sufficient sensitivity and selectivity for immunofluorescence microscopy studies of these processes in preclinical xenograft tumor specimens and the potential for application with clinical samples.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Microambiente Tumoral/efeitos dos fármacos , Antígeno AC133/metabolismo , Animais , Anticorpos Monoclonais/biossíntese , Antineoplásicos/uso terapêutico , Benzamidas/farmacologia , Benzamidas/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Feminino , Técnicas de Introdução de Genes , Fator de Crescimento de Hepatócito/genética , Humanos , Indicadores e Reagentes , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos Transgênicos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Fenótipo , Pirazinas/farmacologia , Pirazinas/uso terapêutico , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Med Chem ; 14(8): 851-862, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29669502

RESUMO

BACKGROUND: The Hantzsch ester, diethyl 2,6-dimethyl-1,4-dihydropyridine-3,5- dicarboxylate, has been used as a hydride donor and its various biological effects have been reported. To identify chemotherapeutic agents with apoptotic effects, 21 diethyl 2,6-dimethyl-1,4- dihydropyridine-3,5-dicarboxylates were designed and synthesized; they have not been reported as apoptosis inducers thus far. Their structure-cytotoxicity relationships were investigated. Further biological experiments were performed on the title compound. METHODS: The cytotoxicities of the current synthetic compounds were measured using a clonogenic assay in HCT116 human colon cancer cells. An annexin V staining assay was used to confirm if the title compound induced apoptosis. To identify the synthetic compounds, Nuclear Magnetic Resonance (NMR) spectroscopy and high-resolution mass spectrometry (HR-MS) were conducted. As molecular symmetry was observed in the NMR spectroscopic data, the three dimensional structures were determined from ab initio calculations and X-ray crystallography. RESULTS: The results obtained from NMR spectroscopy, ab initio calculations, and X-ray crystallography revealed that the diethyl 2,6-dimethyl-1,4-dihydropyridine-3,5-dicarboxylate derivatives synthesized in this research have symmetric structures. The cytotoxicities of the 21 derivatives were tested in the HCT116 human colon cancer cell lines, and their half-maximal cell growth inhibitory concentrations ranged between 16.29 and 68.88 µM. Structure-cytotoxicity relationships demonstrated that bulky substitutions were preferred, para-positioned substituents tended to have better cytotoxic values, and the polarity may have a function as well. The cytotoxicity of the title compound in HCT116 colon cancer cells was mediated through apoptotic cell death. CONCLUSION: To obtain chemotherapeutic agents that induce apoptosis, 21 diethyl 2,6-dimethyl- 1,4-dihydropyridine-3,5-dicarboxylates were designed and synthesized. NMR spectroscopy, ab initio calculations, and X-ray crystallography demonstrated that the diethyl 2,6-dimethyl-1,4- dihydropyridine-3,5-dicarboxylate derivatives synthesized in this research had symmetric structures. Even if the half-maximal cell growth inhibitory concentrations of the 21 derivatives did not show dramatic inhibitory activity against HCT116 human colon cancer cells, small changes in the structure affected the anticancer activities. Treatment with diethyl 4-(4-chlorophenyl)-2,6- dimethyl-1,4-dihydropyridine-3,5-dicarboxylate substantially reduced the cell viability and the cytotoxicity against HCT116 colon cancer cells was mediated through apoptotic cell death. As the ability of diethyl 2,6-dimethyl-1,4-dihydropyridine-3,5-dicarboxylates to induce apoptosis has not been previously reported, we have now reported their design, synthesis, cytotoxicity, and structureactivity relationships.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Di-Hidropiridinas/farmacologia , Ésteres/farmacologia , Niacina/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Caspase 7/metabolismo , Caspase 9/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Di-Hidropiridinas/síntese química , Di-Hidropiridinas/química , Ésteres/síntese química , Ésteres/química , Células HCT116 , Humanos , Modelos Químicos , Niacina/análogos & derivados , Niacina/síntese química , Teoria Quântica , Relação Estrutura-Atividade
9.
Menopause ; 25(1): 77-81, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28796699

RESUMO

OBJECTIVE: Serum uric acid levels increase in postmenopausal women, but decrease when hormone therapy (HT) is administered. No study has, however, evaluated the effects of different types of HT on serum uric acid levels. We therefore examined whether estrogen therapy (ET), estrogen plus progestogen therapy (EPT), and tibolone use affected serum uric acid levels in this population. METHODS: We performed a retrospective cohort study of postmenopausal women. From 2005 to 2015, postmenopausal women who had undergone blood uric acid-level testing at least twice were enrolled. Participants were grouped according to HT regimen: ET, EPT, or tibolone. The nonhormone therapy group did not receive HT. Differences in serum uric acid levels were examined in each group. Our analysis was adjusted to accommodate different follow-up intervals for individual participants. Multiple variables were adjusted using the Tukey-Kramer method. Age, body mass index, hypertension, diabetes mellitus, dyslipidemia, estimated glomerular filtration rate, alcohol consumption, smoking status, and comedications were also adjusted. RESULTS: After adjusting for multiple variables, the serum uric acid level increased to 0.87 ±â€Š0.27 mg/dL (least squares mean ±â€Šstandard error) in the nonhormone therapy group, and serum uric levels in the EPT group were found to be significantly lower (-0.38 ±â€Š0.29 mg/dL, P < 0.001). The serum uric acid levels in the ET and tibolone groups did not, however, differ significantly from the nonhormone therapy group level. CONCLUSIONS: We attribute our findings to the effects of progestogen, rather than estrogen.


Assuntos
Terapia de Reposição de Estrogênios , Pós-Menopausa , Ácido Úrico/sangue , Estudos de Coortes , Estrogênios/administração & dosagem , Feminino , Humanos , Pessoa de Meia-Idade , Progestinas/administração & dosagem , Estudos Retrospectivos
10.
Biochem Soc Trans ; 45(4): 855-870, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28673936

RESUMO

Hepatocyte growth factor (HGF)-induced activation of its cell surface receptor, the Met tyrosine kinase, drives mitogenesis, motogenesis and morphogenesis in a wide spectrum of target cell types and embryologic, developmental and homeostatic contexts. Typical paracrine HGF/Met signaling is regulated by HGF activation at target cell surfaces, HGF binding-induced receptor activation, internalization and degradation. Despite these controls, HGF/Met signaling contributes to oncogenesis, tumor angiogenesis and invasiveness, and tumor metastasis in many types of cancer, leading to the rapid growth of pathway-targeted anticancer drug development programs. We review here HGF and Met structure and function, basic properties of HGF/Met pathway antagonists now in clinical development, and recent clinical trial results. Presently, the main challenges facing the effective use of HGF/Met-targeted antagonists for cancer treatment include optimal patient selection, diagnostic and pharmacodynamic biomarker development, and the identification and testing of effective therapy combinations. The wealth of basic information, analytical reagents and model systems available regarding normal and oncogenic HGF/Met signaling will continue to be invaluable in meeting these challenges and moving expeditiously toward more effective cancer treatment.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Modelos Biológicos , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Animais , Antineoplásicos/uso terapêutico , Fator de Crescimento de Hepatócito/química , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Terapia de Alvo Molecular/tendências , Mutação , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Conformação Proteica , Proteínas Proto-Oncogênicas c-met/química , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo
11.
Med Chem ; 13(2): 168-175, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27220561

RESUMO

BACKGROUND: Since flavonoids fused by benzene have been known for their potent chemopreventive effects, in this study, we examined the relationship between the structures and activities of benzoflavones, benzoflavanones, benzochalcones, and benzochalcone derivatives bearing the pyrazole moiety against human colon cancer cells. METHODS: We investigated the effect of 34 benzoflavonoids on the inhibition of colon cancer cells based on the clonogenicity. The biological activity values used for the quantitative structure-activity relationship (QSAR) calculations were obtained from the cell growth inhibition on the basis of clonogenicity. 3D-QSAR calculations were performed using comparative molecular field analyses (CoMFA) and comparative molecular similarity index analyses (CoMSIA). RESULTS: Of several CoMFA and CoMSIA models, the best models showing the highest cross validated correlation coefficient were selected and validated. The cell growth inhibition values were calculated using the above models. The structural conditions to show good cell growth inhibitory effects on human colon cancer cells were analyzed by CoMFA and CoMSIA contour maps. The contribution of steric fields remarkably decreased without any change in the contribution of the electrostatic field, which means that electrostatic contribution is more crucial than the steric contribution in the modification of benzoflavonoids. Furthermore, the increase in the hydrogen bond donor contribution was approximately proportional to the decrease in steric field contribution. CONCLUSION: This study demonstrated that benzoflavonoids structure hinders colon cancer clonogenicity. Most of the benzoflavonoids structures comprised a C-3 linkage between the naphthalene and phenyl moieties, which contained diverse functional moieties such as oxygen-fused rings, double bonds, pyrazole rings, and sulfur constituents, and were able to exhibit great potential in diverse anticancer effects. Also, the positions of the hydroxyl group close to the naphthalene and phenyl rings were crucial for activity against colon cancer. The structural conditions obtained here may help us design potent benzoflavonoids against colon cancer cells and predict their activities.


Assuntos
Neoplasias do Colo/patologia , Flavonoides/química , Flavonoides/farmacologia , Linhagem Celular Tumoral , Humanos , Modelos Moleculares , Conformação Molecular , Pirazóis/química , Relação Quantitativa Estrutura-Atividade
12.
J Cancer ; 7(10): 1205-14, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27390595

RESUMO

BACKGROUND: Clear cell renal cell carcinoma (ccRCC) is the most lethal form of kidney cancer. Small molecule VEGFR inhibitors are widely used but are not curative and various resistance mechanisms such as activation of the MET pathway have been described. Dual MET/VEGFR2 inhibitors have recently shown clinical benefit but limited preclinical data evaluates their effects in ccRCC. METHODS: An interrogation of the Cancer Genome Atlas (TCGA) dataset was performed to evaluate oncogenic alterations in the MET/VEGFR2 pathway. We evaluated the in vitro effects of Cabozantinib, a dual MET/VEGFR2 inhibitor, using a panel of ccRCC cell lines. Drug effects of cell viability and proliferation, migration, cell scatter, anchorage independent growth, and downstream MET/VEGFR2 signaling pathways were assessed. RESULTS: Twelve percent of TCGA cases had possible MET/HGF oncogenic alterations with co-occurrence noted (p<0.001). MET/HGF altered cases had worse overall survival (p=0.044). Cabozantinib was a potent inhibitor of MET and VEGFR2 in vitro in our cell line panel. PI3K, MAPK and mTOR pathways were also suppressed by cabozantinib, however the effects on cell viability in vitro were modest. At nanomolar concentrations of cabozantinib, HGF-stimulated migration, invasion, cellular scattering and soft agar colony formation were inhibited. CONCLUSIONS: We provide further preclinical rationale for dual MET/VEGFR2 inhibition in ccRCC. While the MET pathway is implicated in VEGFR resistance, dual inhibitors may have direct anti-tumor effects in a patient subset with evidence of MET pathway involvement. Cabozantinib is a potent dual MET/VEGFR2 inhibitor, significantly inhibits cell migration and invasion in vitro and likely has anti-angiogenic effects similar to other VEGFR tyrosine kinase inhibitors. Future work involving in vivo models will be useful to better define mechanisms of potential anti-tumor activity.

13.
Cell Signal ; 28(8): 1114-23, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27224506

RESUMO

Hepatocyte growth factor (HGF) is a pleiotrophic factor involved in cellular proliferation, migration and morphogenesis. HGF is required for normal tissue and organ development during embryogenesis, but in the adult HGF has been demonstrated to drive normal tissue repair and inhibit fibrotic remodeling. HGF has two naturally occurring human isoforms as a result of alternative splicing, NK1 and NK2. While NK1 has been defined as an agonist for HGF receptor, Met, NK2 is defined as a partial Met antagonist. Furthermore, under conditions of fibrotic remodeling, NK2 is still expressed while full length HGF is suppressed. Furthermore, the mechanism by which NK2 partially signals through Met is not completely understood. Here, we investigated the mitogenic, motogenic, and anti-apoptotic activities of NK2 compared with full length HGF in primary human bronchial epithelial cells (BEpC) and bovine pulmonary artery endothelial cells (PAEC). In human BEpC, NK2 partial activated Met, inducing Met phosphorylation at Y1234/1235 in the tyrosine-kinase domain but not at Y1349 site in the multifunctional docking domain. Partial phosphorylation of Met by NK2 resulted in activation of MAPK and STAT3, but not AKT. This correlated with motogenesis and survival in a MAPK-dependent manner, but not cell proliferation. Overexpression of a constitutively active AKT complemented NK2 signaling, allowing NK2 to induce cell proliferation. These data indicate that NK2 and HGF drive motogenic and anti-apoptotic signaling but only HGF drives cell proliferation by activating AKT-pathway signaling. These results have implications for the biological consequences of differential regulation of the two isoforms under pro-fibrotic conditions.


Assuntos
Movimento Celular/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Angiotensina II/farmacologia , Animais , Apoptose/efeitos dos fármacos , Bovinos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Isoformas de Proteínas/farmacologia , Proteólise/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína bcl-X/metabolismo
14.
Diabetologia ; 59(7): 1372-1375, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27115412

RESUMO

Glucagon plays important roles in normal glucose homeostasis and in metabolic abnormalities, particularly diabetes. Glucagon excess, rather than insulin deficiency, is essential for the development of diabetes for several reasons. Glucagon increases hepatic glucose and ketone production, the catabolic features of insulin deficiency. Hyperglucagonaemia is present in every form of diabetes. Beta cell destruction in glucagon receptor null mice does not cause diabetes unless mice are administered adenovirus encoding the glucagon receptor. In rodent studies the glucagon suppressors leptin and glucagon receptor antibody suppressed all catabolic manifestations of diabetes during insulin deficiency. Insulin prevents hyperglycaemia; however, insulin monotherapy cannot cure diabetes such that non-diabetic glucose homeostasis is achieved. Glucose-responsive beta cells normally regulate alpha cells, and diminished insulin action on alpha cells will favour hypersecretion of glucagon by the alpha cells, thus altering the insulin:glucagon ratio. Treating diabetes by suppression of glucagon, with leptin or antibody against the glucagon receptor, normalised glucose level (without glycaemic volatility) and HbA1c. Glucagon suppression also improved insulin sensitivity and glucose tolerance. If these results can be translated to humans, suppression of glucagon action will represent a step forward in the treatment of diabetes. This review summarises a presentation given at the 'Novel data on glucagon' symposium at the 2015 annual meeting of the EASD. It is accompanied by two other reviews on topics from this symposium (by Mona Abraham and Tony Lam, DOI: 10.1007/s00125-016-3950-3 , and by Russell Miller and Morris Birnbaum, DOI: 10.1007/s00125-016-3955-y ) and an overview by the Session Chair, Isabel Valverde (DOI: 10.1007/s00125-016-3946-z ).


Assuntos
Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Glucagon/metabolismo , Animais , Hemoglobinas Glicadas/metabolismo , Humanos , Insulina/metabolismo , Camundongos
15.
Clin Exp Metastasis ; 32(7): 659-76, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26231668

RESUMO

Signaling by human hepatocyte growth factor (hHGF) via its cell surface receptor (MET) drives mitogenesis, motogenesis and morphogenesis in a wide spectrum of target cell types and embryologic, developmental and homeostatic contexts. Oncogenic pathway activation also contributes to tumorigenesis and cancer progression, including tumor angiogenesis and metastasis, in several prevalent malignancies. The HGF gene encodes full-length hHGF and two truncated isoforms known as NK1 and NK2. NK1 induces all three HGF activities at modestly reduced potency, whereas NK2 stimulates only motogenesis and enhances HGF-driven tumor metastasis in transgenic mice. Prior studies have shown that mouse HGF (mHGF) also binds with high affinity to human MET. Here we show that, like NK2, mHGF stimulates cell motility, invasion and spontaneous metastasis of PC3M human prostate adenocarcinoma cells in mice through human MET. To identify target genes and signaling pathways associated with motogenic and metastatic HGF signaling, i.e., the HGF invasive program, gene expression profiling was performed using PC3M cells treated with hHGF, NK2 or mHGF. Results obtained using Ingenuity Pathway Analysis software showed significant overlap with networks and pathways involved in cell movement and metastasis. Interrogating The Cancer Genome Atlas project also identified a subset of 23 gene expression changes in PC3M with a strong tendency for co-occurrence in prostate cancer patients that were associated with significantly decreased disease-free survival.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Invasividade Neoplásica/patologia , Neoplasias da Próstata/patologia , Transdução de Sinais/fisiologia , Animais , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Fator de Crescimento de Hepatócito/genética , Xenoenxertos , Humanos , Masculino , Camundongos , Invasividade Neoplásica/genética , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias da Próstata/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo
16.
Cancers (Basel) ; 6(4): 2313-29, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25534569

RESUMO

There is mounting evidence of oncogenic hepatocyte growth factor (HGF)/Met signaling in urothelial carcinoma (UC) of the bladder. The effects of three kinase inhibitors, cabozantinib, crizotinib and EMD1214063, on HGF-driven signaling and cell growth, invasion and tumorigenicity were analyzed in cultured UC cell lines. SW780 xenograft growth in SCID and human HGF knock-in SCID (hHGF/SCID) mice treated with cabozantinib or vehicle, as well as tumor levels of Met and pMet, were also determined. Met content was robust in most UC-derived cell lines. Basal pMet content and effector activation state in quiescent cells were low, but significantly enhanced by added HGF, as were cell invasion, proliferation and anchorage independent growth. These HGF-driven effects were reversed by Met inhibitor treatment. Tumor xenograft growth was significantly higher in hHGF/SCID mice vs. SCID mice and significantly inhibited by cabozantinib, as was tumor phospho-Met content. These studies indicate the prevalence and functionality of the HGF/Met signaling pathway in UC cells, suggest that paracrine HGF may contribute to UC tumor growth and progression, and that support further preclinical investigation of Met inhibitors for the treatment of UC is warranted.

18.
J Biol Chem ; 289(30): 20448-61, 2014 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-24914205

RESUMO

Hepatocyte growth factor (HGF) signaling promotes tumor invasiveness in renal cell carcinoma (RCC) and other cancers. In clear cell RCC, VHL loss generates pseudohypoxia that exacerbates HGF-driven invasion through ß-catenin deregulation. Hypoxia also enhances HGF-driven invasiveness by papillary RCC cells, but in the absence of VHL, loss signaling integration involves three parallel routes: 1) hypoxia-induced reactive oxygen species production and decreased DUSP2 expression, leading to enhanced mitogen-activated protein kinase (MAPK) cascade activation; 2) reactive oxygen species-induced diacylglycerol production by phospholipase Cγ, leading to protein kinase C activation and increased protein phosphatase- 2A activity, thereby suppressing HGF-induced Akt activation; and 3) a profound shift from HGF-enhanced, proliferation- oriented metabolism to autophagy-dependent invasion and suppression of proliferation. This tripartite signaling integration was not unique to RCC or HGF; in RCC cells, invasive synergy induced by the combination of hypoxia and epidermal growth factor occurred through the same mechanism, and in estrogen receptor-positive breast cancer cells, this mechanism was suppressed in the absence of estrogen. These results define the molecular basis of growth factor and hypoxia invasive synergy in VHL-competent papillary RCC cells, illustrate the plasticity of invasive and proliferative tumor cell states, and provide signaling profiles by which they may be predicted.


Assuntos
Neoplasias da Mama/metabolismo , Carcinoma de Células Renais/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Neoplasias Renais/metabolismo , Sistema de Sinalização das MAP Quinases , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Hipóxia Celular/genética , Linhagem Celular Tumoral , Fosfatase 2 de Especificidade Dupla/genética , Fosfatase 2 de Especificidade Dupla/metabolismo , Feminino , Regulação Enzimológica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Fator de Crescimento de Hepatócito/genética , Humanos , Neoplasias Renais/genética , Neoplasias Renais/patologia , Invasividade Neoplásica , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
19.
Cancer Cell ; 23(2): 228-37, 2013 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-23352416

RESUMO

The natural product englerin A (EA) binds to and activates protein kinase C-θ (PKCθ). EA-dependent activation of PKCθ induces an insulin-resistant phenotype, limiting the access of tumor cells to glucose. At the same time, EA causes PKCθ-mediated phosphorylation and activation of the transcription factor heat shock factor 1, an inducer of glucose dependence. By promoting glucose addiction, while simultaneously starving cells of glucose, EA proves to be synthetically lethal to highly glycolytic tumors.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Glucose/metabolismo , Resistência à Insulina , Insulina/metabolismo , Isoenzimas/metabolismo , Neoplasias Renais/tratamento farmacológico , Proteína Quinase C/metabolismo , Sesquiterpenos de Guaiano/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Animais , Apoptose , Western Blotting , Proliferação de Células , Fatores de Transcrição de Choque Térmico , Humanos , Imunoprecipitação , Insulina/química , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Camundongos , Fosforilação/efeitos dos fármacos , Proteína Quinase C-theta , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Cell Biochem ; 113(4): 1217-23, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22095683

RESUMO

Abnormal accumulation and activation of the recepteur d'origine nantais (RON) has been implicated in carcinogenesis of epithelial tumors. RON expression was induced by the tumor promoter, phorbol 12-myristate 13-acetate (PMA), in gastric adenocarcinoma AGS cells. Studies with deleted and site-directed mutagenesis of Egr-1 promoter and with expression vectors encoding Egr-1 confirmed that Egr-1 is essential for RON expression. In addition, AGS cells pretreated with PMA showed remarkably enhanced invasiveness, which was partially abrogated by siRNA-targeted RON and Egr-1. These results suggest that tumor promoter induces RON expression via Egr-1, which, in turn, stimulates cell invasiveness in AGS cells.


Assuntos
Adenocarcinoma/patologia , Proteína 1 de Resposta de Crescimento Precoce/fisiologia , Invasividade Neoplásica , Receptores Proteína Tirosina Quinases/metabolismo , Neoplasias Gástricas/patologia , Regulação para Cima , Adenocarcinoma/enzimologia , Adenocarcinoma/metabolismo , Sequência de Bases , Western Blotting , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Primers do DNA , Proteína 1 de Resposta de Crescimento Precoce/genética , Humanos , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Gástricas/enzimologia , Neoplasias Gástricas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA