Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Med Chem ; 67(8): 6081-6098, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38401050

RESUMO

In this work, we investigated the anticancer activity of several novel silver(I) 2,2'-bipyridine complexes containing either triphenylphosphane (PPh3) or 1,2-bis(diphenylphosphino)ethane (dppe) ligands. All compounds were characterized by diverse analytical methods including ESI-MS spectrometry; NMR, UV-vis, and FTIR spectroscopies; and elemental analysis. Moreover, several compounds were also studied by X-ray single-crystal diffraction. Subsequently, the compounds were investigated for their anticancer activity against drug-resistant and -sensitive cancer cells. Noteworthily, neither carboplatin and oxaliplatin resistance nor p53 deletion impacted on their anticancer efficacy. MES-OV cells displayed exceptional hypersensitivity to the dppe-containing drugs. This effect was not based on thioredoxin reductase inhibition, enhanced drug uptake, or apoptosis induction. In contrast, dppe silver drugs induced paraptosis, a novel recently described form of programmed cell death. Together with the good tumor specificity of this compound's class, this work suggests that dppe-containing silver complexes could be interesting drug candidates for the treatment of resistant ovarian cancer.


Assuntos
2,2'-Dipiridil , Antineoplásicos , Fosfinas , Prata , Humanos , Fosfinas/química , Fosfinas/farmacologia , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/síntese química , Prata/química , Prata/farmacologia , 2,2'-Dipiridil/química , 2,2'-Dipiridil/farmacologia , Linhagem Celular Tumoral , Complexos de Coordenação/farmacologia , Complexos de Coordenação/química , Complexos de Coordenação/síntese química , Apoptose/efeitos dos fármacos , Cristalografia por Raios X , Ligantes , Morte Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Relação Estrutura-Atividade , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos
2.
Molecules ; 25(19)2020 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-32998226

RESUMO

The tree fern Metaxya rostrata (Kunth) C. Presl is common in the rainforests of Central and South America, where suspensions of the dried rhizome are traditionally used to treat intestinal diseases. Two compounds from this plant, 2-deprenyl-rheediaxanthone B (XB) and 2-deprenyl-7-hydroxy-rheediaxanthone B (OH-XB), have been shown to be biologically highly active against colorectal cancer (CRC) cells in previous studies. The current investigation resulted in the isolation of the previously undescribed methylated xanthones 2-deprenyl-6-O-methyl-7-hydroxy-rheediaxanthone B, 2-deprenyl-5-O-methyl-7-methoxy-rheediaxanthone B, 2-deprenyl-5-O-methyl- 7-hydroxy-rheediaxanthone B and 2-deprenyl-7-methoxy-rheediaxanthone B. All compounds were isolated by column chromatography, structures were elucidated by one- and two-dimensional NMR-experiments and the identities of the compounds were confirmed by LC-HRMS. In logarithmically growing SW480 CRC cell cultures, cytotoxicity by neutral red uptake and MTT assays as well as caspase activation was analyzed. Cellular targets were examined by Western blot, and topoisomerase I (topo I) inhibition potential was tested. Comparing the structure-activity relationship with XB and OH-XB, the monomethylated derivatives showed qualitatively similar effects/mechanisms to their nonmethylated analogues, while dimethylation almost abolished the activity. Inhibition of topo I was dependent on the presence of an unmethylated 7-OH group.


Assuntos
Neoplasias Colorretais/patologia , Gleiquênias/química , Raízes de Plantas/química , Xantonas/farmacologia , Espectroscopia de Ressonância Magnética Nuclear de Carbono-13 , Caspases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , DNA Topoisomerases Tipo I/metabolismo , Humanos , Metilação , Espectroscopia de Prótons por Ressonância Magnética , Inibidores da Topoisomerase/farmacologia , Xantonas/química
3.
Cells ; 8(9)2019 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-31527546

RESUMO

Even though distinctive advances in the field of esophageal cancer therapy have occurred over the last few years, patients' survival rates remain poor. FGF8, FGF18, and FGFR4 have been identified as promising biomarkers in a number of cancers; however no data exist on expression of FGF8, FGF18, and FGFR4 in adenocarcinomas of the esophago-gastric junction (AEG). A preliminary analysis of the Cancer Genome Atlas (TCGA) database on FGF8, FGF18, and FGFR4 mRNA expression data of patients with AEG was performed. Furthermore, protein levels of FGF8, FGF18, and FGFR4 in diagnostic biopsies and post-operative specimens in neoadjuvantly treated and primarily resected patients using immunohistochemistry were investigated. A total of 242 patients was analyzed in this study: 87 patients were investigated in the TCGA data set analysis and 155 patients in the analysis of protein expression using immunohistochemistry. High protein levels of FGF8, FGF18, and FGFR4 were detected in 94 (60.7%), 49 (31.6%) and 84 (54.2%) patients, respectively. Multivariable Cox proportional hazard regression models revealed that high expression of FGF8 was an independent prognostic factor for diminished overall survival for all patients and for neoadjuvantly treated patients. By contrast, FGF18 overexpression was significantly associated with longer survival rates in neoadjuvantly treated patients. In addition, FGF8 protein level correlated with Mandard regression due to neoadjuvant therapy, indicating potential as a predictive marker. In summary, FGF8 and FGF18 are promising candidates for prognostic factors in adenocarcinomas of the esophago-gastric junction and new potential targets for new anti-cancer therapies.


Assuntos
Adenocarcinoma/metabolismo , Neoplasias Esofágicas/metabolismo , Fator 8 de Crescimento de Fibroblasto/biossíntese , Fatores de Crescimento de Fibroblastos/biossíntese , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/biossíntese , Adenocarcinoma/diagnóstico , Adenocarcinoma/patologia , Idoso , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/patologia , Feminino , Fator 8 de Crescimento de Fibroblasto/genética , Fatores de Crescimento de Fibroblastos/genética , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/genética , Taxa de Sobrevida
4.
J Cancer Res Clin Oncol ; 145(1): 77-86, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30276721

RESUMO

PURPOSE: Therapy response to neoadjuvant radiochemotherapy (nRCT) of locally advanced rectal cancer varies widely so that markers predicting response are urgently needed. Fibroblast growth factor (FGF) and FGF receptor (FGFR) signaling is involved in pro-survival signaling and thereby may result in radiation resistance. METHODS: In a cohort of 43 rectal cancer patients, who received nRCT, we analyzed protein levels of FGF 8 and its downstream target Survivin by immunohistochemistry to assess their impact on nRCT response. In vitro resistance models were created by exposing colorectal cancer cell lines to fractionated irradiation and selecting long-term survivors. RESULTS: Our findings revealed significantly higher FGF8 and Survivin staining scores in pre-treatment biopsies as well as in surgical specimens of non-responsive compared to responsive patients. Functional studies demonstrated dose-dependent induction of FGF8 mRNA expression in mismatch-incompetent DLD1 cells already after one dose of irradiation. Surviving clones after one or two series of radiation were more resistant to an additional radiation fraction than non-irradiated controls and showed a significant increase in expression of the FGF8 receptor FGFR3 and of Survivin on both the RNA and the protein levels. CONCLUSION: The results of this study suggest that FGF8 and Survivin contribute to radiation resistance in rectal cancer and may serve as markers to select patients who may not benefit from neoadjuvant radiotherapy.


Assuntos
Quimiorradioterapia Adjuvante , Fator 8 de Crescimento de Fibroblasto/fisiologia , Tolerância a Radiação/fisiologia , Neoplasias Retais/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Quimiorradioterapia , Resistencia a Medicamentos Antineoplásicos , Feminino , Fator 8 de Crescimento de Fibroblasto/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Neoplasias Retais/patologia , Neoplasias Retais/cirurgia , Estudos Retrospectivos , Transdução de Sinais/fisiologia , Survivina/metabolismo
5.
Transl Oncol ; 10(3): 332-339, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28340475

RESUMO

BACKGROUND: Irinotecan (IRI) is an integral part of colorectal cancer (CRC) therapy, but response rates are unsatisfactory and resistance mechanisms are still insufficiently understood. As fibroblast growth factor receptor 3 (FGFR3) mediates essential survival signals in CRC, it is a candidate gene for causing intrinsic resistance to IRI. METHODS: We have used cell line models overexpressing FGFR3 to study the receptor's impact on IRI response. For pathway blockade, a dominant-negative receptor mutant and a small molecule kinase inhibitor were employed. RESULTS: IRI exposure induced expression of FGFR3 as well as its ligands FGF8 and FGF18 both in cell cultures and in xenograft tumors. As overexpression of FGFR3 mitigated IRI-induced apoptosis in CRC cell models, this suggests that the drug itself activated a survival response. On the cellular level, the antiapoptotic protein bcl-xl was upregulated and caspase 3 activation was inhibited. Targeting FGFR3 signaling using a dominant-negative receptor mutant sensitized cells for IRI. In addition, the FGFR inhibitor PD173074 acted synergistically with the chemotherapeutic drug and significantly enhanced IRI-induced caspase 3 activity in vitro. In vivo, PD173074 strongly inhibited growth of IRI-treated tumors. CONCLUSION: Together, our results indicate that targeting FGFR3 can be a promising strategy to enhance IRI response in CRC patients.

6.
Mol Carcinog ; 54(9): 789-799, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24619956

RESUMO

Expansion of a stem-like subpopulation with increased growth and survival potential is thought to drive colorectal tumor growth and progression. We investigated a CD44-positive (CD44((+))) subpopulation with extended growth and survival capacity in the human colon adenoma cell line LT97. This subpopulation expressed elevated levels of fibroblast growth factor 18 (FGF18) and fibroblast growth factor receptor FGFR3-IIIc. Expression levels of the FGFR3-IIIb, which does not bind FGF18, were similar in CD44((+)) and CD44((-)). Addition of FGF18 to the medium or its overexpression from an adenoviral vector increased the colony formation capacity of CD44((+)) threefold, and stimulated phosphorylation of ERK and GSK3ß in both total LT97 populations and CD44((+)) cells. FGFR3 signaling blockade by expression of a dominant-negative FGFR3-IIIc mutant led to inhibition of both colony formation and down-stream signaling in the CD44((+)) cells. CD44((-)) cells did not respond. Blockade of the wnt-pathway by a dominant-negative Tcf4-mutant inhibited FGFR3 activation in LT97 cells as well as in HT29 colorectal cancer cells. The chemical wnt-inhibitor sulindac sulfide amide inhibited expression of FGF18 and FGFR3-IIIc and led to inhibition of receptor activation to less than 30% of control treated cells, both in LT97 and HT29 cultures. Our results demonstrate that an FGF18/FGFR3-IIIc autocrine growth and survival loop is up-regulated in a wnt-dependent manner and drives tumor cell growth in a subpopulation of colon adenoma cells. This subpopulation can be regarded as a precursor of colon cancer development and can be targeted for CRC-prevention by blocking either wnt- or FGFR3-signaling.


Assuntos
Adenoma/patologia , Neoplasias Colorretais/patologia , Fatores de Crescimento de Fibroblastos/metabolismo , Receptores de Hialuronatos/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , Adenoma/tratamento farmacológico , Adenoma/metabolismo , Antineoplásicos/farmacologia , Colo/efeitos dos fármacos , Colo/metabolismo , Colo/patologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Humanos , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Reto/efeitos dos fármacos , Reto/metabolismo , Reto/patologia , Transdução de Sinais/efeitos dos fármacos , Sulindaco/análogos & derivados , Sulindaco/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos
7.
J Cancer Res Clin Oncol ; 137(1): 29-39, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20221637

RESUMO

BACKGROUND: Peroxisome-proliferator-activated receptors (PPARs) are nuclear receptors for fatty acids and their derivatives. PPAR subtypes PPARγ and PPARß/δ are suspected to modulate cancer development in the colon, but their exact role is still discussed controversially. METHODS: The present study investigated the impact of PPARγ and PPARß/δ on vascular endothelial growth factor (VEGF) and cyclooxygenase 2 (COX-2) expressions induced by synthetic and physiological agonists in the colorectal tumor cell lines SW480 and HT29 using reporter gene assays, qRT-PCR and ELISA. RESULTS: Activation of both PPARγ and PPARß/δ induced expression of VEGF mRNA and protein in a PPAR-dependent way. The PPARγ agonists ciglitazone and PGJ(2) were the most effective inducers with up to ninefold and threefold increases in VEGF mRNA in SW480 and HT29 cultures, respectively. VEGF secretion was doubled in both cell lines. The PPARß/δ agonists GW501516 and PGI(2) caused stimulations of only 1.5-fold in both cell lines. In addition, all PPAR agonists induced COX-2 mRNA and secretion of the COX-2 product PGE(2) in HT29 cells. However, this effect was not blocked by knock-down of PPAR expression nor was it essential for VEGF expression as shown by the lack of effect of the COX-2 inhibitor SC236. CONCLUSION: In summary, our results identify both PPARγ and PPARß/δ as an alternative COX-independent mechanism of VEGF induction in colorectal tumor cells.


Assuntos
Neoplasias Colorretais/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Ciclo-Oxigenase 2/genética , Humanos , PPAR delta/agonistas , PPAR delta/metabolismo , PPAR gama/agonistas , PPAR gama/metabolismo , PPAR beta/agonistas , PPAR beta/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/agonistas , RNA Mensageiro/metabolismo , Tiazóis/farmacologia , Tiazolidinedionas/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA