Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Crit Rev Oncol Hematol ; 96(3): 518-26, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26321263

RESUMO

BACKGROUND: Renal cell carcinoma (RCC) is a heterogeneous disease with regards to histology, progression, and response to treatment. Cytotoxic chemotherapy has been extensively studied in metastatic RCC (mRCC). Responses in most studies are modest and the mechanisms of resistance remain poorly understood. Targeted therapies have significantly improved outcomes in mRCC; however, most patients eventually relapse and die of their disease. Early clinical data suggest that combinations of chemotherapy and targeted agents are clinically active and are well tolerated. METHODS: We reviewed the available literature for published clinical trials incorporating traditional chemotherapeutic agents in the treatment of mRCC. These papers were identified through a Medline search and were included if they employed at least one chemotherapeutic agent in the treatment of mRCC. The literature was also reviewed for information regarding mechanisms of chemotherapy resistance. RESULTS: The data regarding the use of cytotoxic chemotherapy in mRCC consist of small, non-randomized phase I and II studies. The major response proportions with single agent chemotherapies are low but combination regimens either with other cytotoxic agents, cytokines, or targeted agents have demonstrated moderate activity. Disparate trial designs and lack of head to head clinical trials make it difficult to compare the efficacy of chemotherapy with that of immunotherapy or targeted agents. Chemotherapy is particularly useful in patients with collecting duct histology and predominantly sarcomatoid differentiation. Chemotherapy resistance may be mediated by overexpression of p-glycoprotein efflux pumps and the dysregulation of the microtubule-hypoxia inducible factor signaling axis. CONCLUSIONS: The role of cytotoxic chemotherapy in the treatment for clear cell RCC remains poorly defined. Cytotoxic chemotherapy is considered a standard of care in patients with mRCC with predominantly sarcomatoid differentiation and collecting duct RCC variants (Motzer et al., 2014). Early trials combining chemotherapy with targeted therapies are generally well tolerated and show clinical activity. A better understanding of the biology of aggressive subsets of RCC and mechanisms of resistance will help elucidate the role of cytotoxic agents in the current treatment paradigm of RCC.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/patologia , Neoplasias Renais/tratamento farmacológico , Terapia de Alvo Molecular , Animais , Humanos
2.
Horm Metab Res ; 40(11): 772-8, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18726827

RESUMO

Treatment with glucocorticoids is one of a limited number of options for androgen independent prostate cancer. Neuroendocrine differentiation has been shown to contribute to androgen-independent prostate cancer progression. To study the potential link between neuroendocrine differentiation and the glucocorticoid action, we investigated the effects of the product of neuroendocrine differentiation--bombesin on glucocorticoid metabolizing enzymes--11beta-hydroxysteroid dehydrogenases in PC-3 cells. Our Western analysis, RT-PCR, and activity assays demonstrate that while 18-hour exposure to bombesin reduces 11beta-hydroxy-steroid dehydrogenases-1 profiles (activities 25% less, protein level 29% lower, mRNA levels 45% lower), contrarily it increases 11beta-hydroxysteroid dehydrogenases-2 profiles (activities 34%, protein levels 100%, mRNA levels 120%). Blockade bombesin action with bombesin receptor antagonists and the enzyme degrading bombesin prevented these changes, suggesting the observed modulations were bombesin receptor-specific. In addition, bombesin increased the amounts of interleukin-8 and mRNA of vascular endothelial growth factor receptor 2, which were lowered in the presence of cortisol, suggesting that neuropeptide blockade may extend the benefits of glucocorticoids in treating androgen-independent prostate cancer.


Assuntos
11-beta-Hidroxiesteroide Desidrogenases/genética , Androgênios , Bombesina/farmacologia , Expressão Gênica/efeitos dos fármacos , Glucocorticoides/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Linhagem Celular Tumoral , Resistência a Medicamentos , Glucocorticoides/antagonistas & inibidores , Glucocorticoides/metabolismo , Humanos , Interleucina-8/metabolismo , Masculino , Neovascularização Patológica/prevenção & controle , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/enzimologia , RNA Mensageiro/análise , Receptores da Bombesina/antagonistas & inibidores , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
3.
Prostate Cancer Prostatic Dis ; 11(1): 79-87, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17563767

RESUMO

Neutral endopeptidase (NEP) is a cell surface peptidase that catalytically inactivates a variety of physiologically active peptides including basic fibroblast growth factor (FGF-2). We investigated the effect of using lentivirus to overexpress NEP in NEP-deficient DU145 prostate cancer cells. Third-generation lentiviral vectors encoding wild-type NEP (L-NEP), catalytically inactive mutant NEP (L-NEPmu), and green fluorescent protein (L-GFP) were stably introduced into DU145 cells. FGF-2 levels in cell culture supernatants decreased by 80% in L-NEP-infected DU145 cells compared to cells infected with L-NEPmu or L-GFP (P<0.05) while levels of other angiogenic factors were not altered. In vitro tubulogenesis of human vascular endothelial cells induced by conditioned media from DU145 cells infected with L-NEP was significantly reduced compared with that from DU145 cells infected with L-GFP (P<0.05). Tumor xenografts from L-NEP-infected DU145 cells were significantly smaller compared to control cell xenografts and vascularity within these tumors was decreased (P<0.05). Our data suggest that stable expression of NEP in DU145 cells inhibits prostate cancer tumorigenicity by inhibiting angiogenesis, with a probable mechanism being proteolytic inactivation of FGF-2.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Neovascularização Patológica/prevenção & controle , Neprilisina/metabolismo , Neoplasias da Próstata/irrigação sanguínea , Neoplasias da Próstata/terapia , Animais , Proliferação de Células , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Vetores Genéticos , Humanos , Immunoblotting , Imunoprecipitação , Lentivirus/genética , Masculino , Camundongos , Camundongos Nus , Invasividade Neoplásica , Neprilisina/genética , Neoplasias da Próstata/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Cancer Gene Ther ; 14(6): 583-9, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17415380

RESUMO

Neprilysin (neutral endopeptidase, NEP) is a cell surface peptidase whose expression is lost in approximately 50% of prostate cancers (PC). NEP normally functions to inactivate peptides such as bombesin and endothelin-1, and potentiates the effects of the PTEN tumor suppressor via a direct protein-protein interaction. NEP loss contributes to PC progression. We investigated the therapeutic efficacy of using a lentiviral vector system to restore NEP expression in PC cells. Third-generation lentiviral vectors encoding wild-type NEP (L-NEP) or green fluorescent protein (L-GFP) were introduced into NEP-deficient 22RV1 PC cells. Cells infected with L-NEP or L-GFP at a multiplicity of infection of 10 demonstrated NEP enzyme activity of 1171.2+/-4.9 and 17.2+/-5.3 pmol/microg/min (P<0.0001), respectively. Cell viability, proliferation and invasion were each significantly inhibited in 22RV1 cells expressing NEP compared with control cells infected with L-GFP (P<0.01). Analysis of known downstream effects of NEP showed NEP-expressing cells exhibiting decreased Akt and focal adhesion kinase phosphorylation and increased PTEN protein expression. Finally, injection of L-NEP into established 22RV1 xenograft tumors significantly inhibited tumor growth (P<0.01). These experiments demonstrate that lentiviral NEP gene transfer is a novel targeted strategy for the treatment of NEP-deficient PC.


Assuntos
Lentivirus/genética , Invasividade Neoplásica/fisiopatologia , Neprilisina/uso terapêutico , Neoplasias da Próstata/terapia , Animais , Linhagem Celular , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Masculino , Camundongos , Camundongos Nus , Neprilisina/genética , Neprilisina/imunologia , Neoplasias da Próstata/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Oncogene ; 25(44): 5942-52, 2006 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-16652149

RESUMO

The neuropeptides bombesin and endothelin-1 stimulate prostate cancer (PC) cell migration and invasion (J Clin Invest, 2000; 106: 1399-1407). The intracellular signaling pathways that direct this cell movement are not well delineated. The monomeric GTPase RhoA is required for migration in several cell types including neutrophils, monocytes and fibroblasts. We demonstrate that bombesin-stimulated PC cell migration occurs via the heterotrimeric G-protein-coupled receptors (G-protein) G alpha 13 subunit leading to activation of RhoA, and Rho-associated coiled-coil forming protein kinase (ROCK). Using siRNA to suppress expression of the three known G-protein alpha-subunit-associated RhoA guanine nucleotide exchange factors (GEFs), we also show that two of these RhoA GEFs, PDZ-RhoGEF and leukemia-associated RhoGEF (LARG), link bombesin receptors to RhoA in a non-redundant manner in PC cells. We next show that focal adhesion kinase, which activates PDZ-RhoGEF and LARG, is required for bombesin-stimulated RhoA activation. Neutral endopeptidase (NEP) is expressed on normal prostate epithelium whereas loss of NEP expression contributes to PC progression. We also demonstrate that NEP inhibits neuropeptide activation of RhoA. Together, these results establish a contiguous signaling pathway from the bombesin receptor to ROCK in PC cells, and they implicate NEP as a major regulator of neuropeptide-stimulated RhoA in these cells. This work also identifies members of this signaling pathway as potential targets for rational pharmacologic manipulation of neuropeptide-stimulated migration of PC cells.


Assuntos
Movimento Celular/fisiologia , Neprilisina/fisiologia , Neuropeptídeos/fisiologia , Neoplasias da Próstata/enzimologia , Transdução de Sinais/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia , Bombesina/fisiologia , Linhagem Celular Tumoral , Citoesqueleto/metabolismo , Endotelina-1/fisiologia , Ativação Enzimática/fisiologia , Humanos , Masculino , Neoplasias da Próstata/patologia
6.
Oncogene ; 25(14): 2011-21, 2006 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-16434977

RESUMO

Androgen receptor signaling in prostate cancer cells is augmented by the androgen receptor (AR) coactivator p300, which transactivates and acetylates the AR in the presence of dihydrotestosterone (DHT). As prostate cancer (PC) cells progress to androgen independence, AR signaling remains intact, indicating that other factors stimulate AR activities in the absence of androgen. We previously reported that neuropeptide growth factors could transactivate the AR in the presence of very low concentrations of DHT. Here, we examine the involvement of p300 in neuropeptide activation of AR signaling. Transfection of increasing concentrations of p300 in the presence of bombesin into PC-3 cells resulted in a linear increase in AR transactivation, suggesting that p300 acts as a coactivator in neuropeptide-mediated AR transactivation. P300 is endowed with histone acetyltransferase (HAT) activity. Therefore, we examine the effect of bombesin on p300 HAT activity. At 4 h after the addition of bombesin, p300 HAT activity increased 2.0-fold (P<0.01). Incubation with neutral endopeptidase, which degrades bombesin, or bombesin receptor antagonists blocked bombesin-induced p300 HAT activity. To explore the potential signaling pathways involved in bombesin-induced p300 HAT activity, we examined Src and PKCdelta pathways that mediate bombesin signaling. Inhibitors of Src kinase activity or Src kinase siRNA blocked bombesin-induced p300 HAT activity, whereas PKCdelta inhibitors or PKCdelta siRNA significantly increased bombesin-induced p300 HAT activity suggesting that Src kinase and PKCdelta kinase are involved in the regulation of p300 HAT activity. As AR is acetylated in the presence of 100 nM DHT, we next examined whether bombesin-induced p300 HAT activity would result in enhanced AR acetylation. Bombesin-induced AR acetylation at the same motif KLKK observed in DHT-induced acetylation. Elimination of p300 using p300 siRNA reduced AR acetylation, demonstrating that AR acetylation was mediated by p300. AR acetylation results in AR transactivation and the expression of the AR-regulated gene prostate-specific antigen (PSA). Therefore, we examined bombesin-induced AR transactivation and PSA expression in the presence and absence of p300 siRNA and found inhibition of p300 expression reduced bombesin-induced AR transactivation and PSA expression. Together these results demonstrate that bombesin, via Src and PKCdelta signaling pathways, activates p300 HAT activity which leads to enhanced acetylation of AR resulting in increased expression of AR-regulated genes.


Assuntos
Bombesina/farmacologia , Proteínas de Ciclo Celular/metabolismo , Histona Acetiltransferases/metabolismo , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Acetilação , Western Blotting , Linhagem Celular Tumoral , Ativação Enzimática , Humanos , Masculino , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Proteína Quinase C-delta/metabolismo , RNA Interferente Pequeno , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Transdução de Sinais , Ativação Transcricional , Fatores de Transcrição de p300-CBP
7.
Br J Cancer ; 91(1): 164-70, 2004 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-15188008

RESUMO

Interferon alpha (IFNalpha) is used to treat patients with advanced renal cell carcinoma (RCC) despite limited clinical benefit. IFNalpha can induce Fas receptor-mediated apoptosis by direct activation of pro-caspase-8 followed by activation of caspase-3. Alternative, indirect activation of caspase-3 via mitochondrial release of cytochrome c can occur and may explain the rescue from Fas-activated cell death by the antiapoptotic members of the Bcl-2 family. In this study, we examined G3139, a novel antisense compound targeting Bcl-2, in combination with IFNalpha. Human RCC lines (SK-RC-44 and SK-RC-07) were treated with IFNalpha, G3139 or a combination of the two. Fas-mediated cytotoxicity was induced by anti-Fas mAb, CH11. An analysis of Bcl-2, Fas and the cleavage of PARP was performed. IFNalpha induced Fas and Bcl-2 in SK-RC-44 and SK-RC-07. IFNalpha sensitised SK-RC-44 to anti-Fas and induced PARP cleavage confirming that IFNalpha has a cytotoxic effect on RCC lines by induction of the Fas antigen. Cytotoxicity was not evident in SK-RC-07 cells treated with IFNalpha. G3139 induced a specific downregulation of Bcl-2 in SK-RC-07 cells, which were then sensitised to anti-Fas after treatment with IFNalpha. Taken together, these results suggest that Fas-dependent pathways as well as alternative pathways, which can be inhibited by Bcl-2, exist in renal cell carcinoma. G3139 in combination with IFNalpha is a potential therapy in patients with metastatic renal cell carcinoma.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma de Células Renais/patologia , Interferon-alfa/farmacologia , Neoplasias Renais/patologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Tionucleotídeos/farmacologia , Receptor fas/farmacologia , Carcinoma de Células Renais/metabolismo , Regulação para Baixo , Humanos , Neoplasias Renais/metabolismo , Oligonucleotídeos Antissenso , Proteínas Proto-Oncogênicas c-bcl-2/genética , Células Tumorais Cultivadas
9.
Clin Cancer Res ; 7(5): 1370-7, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11350908

RESUMO

Expression of neutral endopeptidase (NEP) 24.11 is diminished in metastatic, androgen-independent prostate cancers (PCs; C. N. Papandreou et al., NAT: MED:, 4: 50--57, 1998). To determine the effects on androgen-independent PC cells of overexpressing cell-surface NEP, an inducible tetracycline-regulatory gene expression system was used to stably introduce and express the NEP gene in androgen-independent TSU-Pr1 cells generating WT-5 cells, which expressed high levels of enzymatically active NEP protein when cultured in the absence of tetracycline. TN12 cells, which contain the identical vectors without the NEP gene and do not express NEP, were used as control. Expression of NEP in WT-5 cells after removal of tetracycline from the media resulted in a >80% inhibition in cell proliferation over a 1-week period (P < 0.005) compared with control cells. Tumor formation occurred in the prostate glands of orthotopically injected athymic mice killed at 30 days in 4 of 5 mice that were given injections of 2 x 10(6) WT-5 cells and were fed doxycycline (NEP suppressed), and in all mice that were given injections of TN12 cells and were fed with or without doxycycline. In contrast, only 1 of 5 mouse prostates developed a tumor in mice that were given injections of WT-5 cells and that did not receive doxycycline. Analysis of the mechanisms of NEP-induced growth suppression revealed that NEP expression in WT-5 cells induced a 4-fold increase in the number of PC cells undergoing apoptosis, and increased the expression of p21 tumor suppressor gene protein and the level of unphosphorylated retinoblastoma protein as determined by Western blot. Flow cytometric analysis show that induced NEP expression in WT-5 cells resulted in a G(1) cell cycle arrest. These data show that NEP can inhibit PC cell growth and tumorigenicity and suggest that NEP has potential as therapy for androgen-independent PC.


Assuntos
Apoptose , Genes Supressores de Tumor/fisiologia , Neprilisina/metabolismo , Neoplasias da Próstata/enzimologia , Androgênios/metabolismo , Animais , Apoptose/fisiologia , Testes de Carcinogenicidade , Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Nus , Neprilisina/genética , Transfecção , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancer Res ; 61(8): 3294-8, 2001 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11309283

RESUMO

G-protein coupled receptor (GPCR) agonists such as neuropeptides activate the insulin-like growth factor-1 receptor (IGF-IR) or the serine-threonine protein kinase Akt, suggesting that neuropeptides-GPCR signaling can cross-communicate with IGF-IR-Akt signaling pathways. Neutral endopeptidase 24.11 (NEP) is a cell-surface peptidase that cleaves and inactivates the neuropeptides endothelin-1 (ET-1) and bombesin, which are implicated in progression to androgen-independent prostate cancer (PC). We investigated the mechanisms of NEP regulation of neuropeptide-mediated cell survival in PC cells, including whether neuropeptide substrates of NEP induce phosphorylations of IGF-IR and Akt in PC cells. Western analyses revealed ET-1 and bombesin treatment induced phosphorylation of IGF-IRbeta and Akt independent of IGF-I in TSU-Pr1, DU145, and PC-3 PC cells, which lack NEP expression, but not in NEP-expressing LNCaP cells. Recombinant NEP and induced NEP expression in TSU-Pr1 cells using a tetracycline-repressive expression system inhibited ET-1-mediated phosphorylation of IGF-IRbeta and Akt, and blocked the protective effects of ET-1 against apoptosis induced by serum starvation. Incubation of TSU-Pr1 cells with specific kinase inhibitors together with ET-1 or bombesin showed that IGF-IR activation is required for neuropeptide-induced Akt phosphorylation, and that neuropeptide-induced Akt activation is predominantly mediated by Src and phosphatidylinositol 3-kinase but not by mitogen-activated protein kinase or protein kinase C. These data show that the neuropeptides ET-1 and bombesin stimulate ligand-independent activation of the IGF-IR, which results in Akt activation, and that this cross-communication between GPCR and IGF-IR signaling is inhibited by NEP.


Assuntos
Bombesina/antagonistas & inibidores , Endotelina-1/antagonistas & inibidores , Neprilisina/fisiologia , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/fisiologia , Receptor IGF Tipo 1/fisiologia , Bombesina/farmacologia , Sobrevivência Celular/fisiologia , Endotelina-1/farmacologia , Ativação Enzimática , Humanos , Masculino , Fosforilação , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais/fisiologia , Ativação Transcricional , Células Tumorais Cultivadas , Quinases da Família src/metabolismo
11.
Cancer Res ; 61(6): 2774-81, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11289161

RESUMO

Clinical and preclinical studies suggest that retinoids can inhibit the growth of a small percentage of human renal cancers (RCs), although the majority of RCs both in vitro and in vivo are retinoid resistant. Our recent studies indicate that the metabolism of retinol to retinyl esters is greatly reduced in human carcinoma cell lines of the oral cavity, skin, and breast as compared with their normal epithelial counterparts, suggesting that human carcinoma cells are retinoid deficient relative to normal epithelial cells. We considered whether retinoid resistance in RCs was related to an abnormality in retinoid metabolism. The metabolism of [3H]retinol and of [3H]retinoic acid (RA) was examined in RC cell lines and normal human kidney (NK) epithelial cells cultured in media, in RA, or in RA plus IFN-alpha. The expression of LRAT (lecithin:retinol acyltransferase) was assessed by Northern and Western analysis. Retinol and retinyl ester levels were determined in tissue samples of normal human kidney and renal cell carcinoma. NK cells esterified all of the 50 nM [3H]retinol in which they were cultured. In contrast, six of the seven RC cell lines metabolized only trace amounts of [3H]retinol to [3H]retinyl esters. Consistent with this relative lack of [3H]retinol esterification by the tumor cells, the tumor cells exhibited LRAT transcripts of aberrantly low sizes relative to those in normal epithelial cells. Moreover, the NK cells expressed abundant levels of LRAT protein by Western analysis, whereas the RC cells did not express LRAT protein. When samples of human kidney tumor tissue were compared with samples of normal kidney tissue from patients who had undergone surgery for primary RC, the normal kidney tissues contained much higher levels of retinol and retinyl esters (approximately 0.5-2 microg/gram wet weight) than the tumor tissues in all seven patients examined. Culture of the RC lines in IFN-alpha plus all-trans-RA, a combination therapy used clinically, resulted in higher intracellular levels of [3H]retinol and [3H]retinyl esters. The metabolism of [3H]RA was also examined in these RC lines versus NK cells. Although the NK epithelial cells metabolized [3H]RA, the majority of the RC lines metabolized [3H]RA at a much slower rate. Most of the RC lines metabolized only 10-30% of the 50 nM [3H]RA over 6 h of culture. These data indicate that RCs both in vitro and in vivo are retinol and retinyl ester deficient relative to the normal human kidney, and they suggest that the aberrant differentiation of the neoplastic renal cells results in part from a defect in retinoid metabolism.


Assuntos
Carcinoma de Células Renais/metabolismo , Neoplasias Renais/metabolismo , Tretinoína/análogos & derivados , Vitamina A/metabolismo , Aciltransferases/biossíntese , Aciltransferases/genética , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carcinoma de Células Renais/patologia , Divisão Celular/efeitos dos fármacos , Sistema Enzimático do Citocromo P-450/biossíntese , Sistema Enzimático do Citocromo P-450/genética , Ésteres/metabolismo , Feminino , Expressão Gênica , Humanos , Interferon alfa-2 , Interferon-alfa/farmacologia , Neoplasias Renais/patologia , Túbulos Renais Proximais/metabolismo , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes , Ácido Retinoico 4 Hidroxilase , Tretinoína/metabolismo , Tretinoína/farmacologia , Células Tumorais Cultivadas
12.
Curr Treat Options Oncol ; 2(5): 437-45, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12057107

RESUMO

Advanced renal cell carcinoma (RCC) is a disease that is highly resistant to systemic therapy and is difficult to treat. Nephrectomy should be seriously considered in patients who present with metastatic disease prior to systemic therapy, and surgery remains a reasonable option in patients who present with resectable metastases. Numerous studies with many different treatment modalities, including chemotherapy, immunotherapy, and radiation therapy, have failed to consistently benefit patients, with no single agent or combination therapy showing a reproducible response proportion of 20% or higher. Interleukin-2 (IL-2) and interferon-alfa (IFN alfa)-based therapies remain the most commonly used agents to treat patients with advanced disease, demonstrating low but reproducible response proportions in the 10% to 20% range, with durable responses of 5% or less. Recent randomized studies demonstrate a survival advantage for patients receiving systemic IFN-based therapy, but this advantage is marginal. Novel treatment strategies are being investigated, with some encouraging early results using vaccines and allogeneic bone marrow transplant. The identification of new agents with more effective antitumor activity is a high priority in the treatment of advanced RCC.


Assuntos
Carcinoma de Células Renais/patologia , Neoplasias Renais/patologia , Idoso , Anticorpos Monoclonais/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Transplante de Medula Óssea , Carcinoma de Células Renais/mortalidade , Carcinoma de Células Renais/terapia , Terapia Combinada , Métodos Epidemiológicos , Feminino , Humanos , Fatores Imunológicos/uso terapêutico , Imunoterapia , Imunoterapia Ativa , Interferon-alfa/uso terapêutico , Interleucina-2/uso terapêutico , Neoplasias Renais/mortalidade , Neoplasias Renais/terapia , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Nefrectomia , Radioterapia Adjuvante , Fatores de Risco , Resultado do Tratamento
13.
Methods Mol Med ; 53: 245-53, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-21318799

RESUMO

A human renal cancer was first established in continuous culture in 1962. Currently, there are well over 100 different characterized renal cancer cell lines derived from both primary and metastatic renal cell carcinomas (RCCs) (1-3). The biological phenotype of cultured renal cancer cells typically includes a sustained and essentially unlimited growth capacity, a lack of contact inhibition and anchorage dependence, a capacity to form tumors in athymic mice, and an aneuploid karyotype including nonrandom chromosomal abnormalities (1,2). The antigenic phenotype of RCCs as determined by monoclonal antibodies (mAbs) generated against cell-surface glycoproteins, glycolipids, and blood-group antigens of renal cancers provide a series of phenotypic markers which characterize these tumors (4-6). Many of these mAbs also react with the proximal tubule portion of the human nephron, confirming earlier studies indicating that >90% of renal cancers derive from epithelial cells of the proximal tubule (7,8). While established RCC cell lines have frequently been analyzed for molecular defects, their greatest utility has been to screen combinations of chemotherapeutic and biologic agents for antiproliferative activity (9-12). Short-term cultures of renal cancer cells derived from fresh tumor specimens have similarly been used to screen drugs (13), but inhibitory effects in vitro have not been shown to predict a response in vivo (i.e., in human patients).

14.
J Clin Invest ; 106(11): 1399-407, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11104793

RESUMO

Neutral endopeptidase 24.11 (NEP, CD10) is a cell-surface enzyme expressed by prostatic epithelial cells that cleaves and inactivates neuropeptides implicated in the growth of androgen-independent prostate cancer (PC). NEP substrates such as bombesin and endothelin-1 induce cell migration. We investigated the mechanisms of NEP regulation of cell migration in PC cells, including regulation of phosphorylation on tyrosine of focal adhesion kinase (FAK). Western analyses and cell migration assays revealed an inverse correlation between NEP expression and the levels of FAK phosphorylation and cell migration in PC cell lines. Constitutively expressed NEP, recombinant NEP, and induced NEP expression using a tetracycline-repressive expression system inhibited bombesin- and endothelin-1-stimulated FAK phosphorylation and cell migration. This results from NEP-induced inhibition of neuropeptide-stimulated association of FAK with cSrc protein. Expression of a mutated catalytically inactive NEP protein also resulted in partial inhibition of FAK phosphorylation and cell migration. Coimmunoprecipitation experiments show that NEP associates with tyrosine-phosphorylated Lyn kinase, which then binds the p85 subunit of phosphatidylinositol 3-kinase (PI3-K) resulting in an NEP-Lyn-PI3-K protein complex. This complex competitively blocks FAK-PI3-K interaction, suggesting that NEP protein inhibits cell migration via a protein-protein interaction independent of its catalytic function. These experiments demonstrate that NEP can inhibit FAK phosphorylation on tyrosine and PC cell migration through multiple pathways and suggest that cell migration which contributes to invasion and metastases in PC cells can be regulated by NEP.


Assuntos
Movimento Celular , Neprilisina/metabolismo , Fenilalanina/análogos & derivados , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Animais , Bombesina/farmacologia , Células COS , Movimento Celular/efeitos dos fármacos , Meios de Cultura Livres de Soro/farmacologia , DNA Recombinante/genética , DNA Recombinante/metabolismo , Endotelina-1/farmacologia , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Neprilisina/genética , Organofosfonatos/farmacologia , Fenilalanina/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Células Tumorais Cultivadas , Quinases da Família src/metabolismo
15.
Curr Oncol Rep ; 2(5): 417-22, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11122873

RESUMO

Metastatic renal cell carcinoma remains a disease that is highly resistant to systemic therapy and difficult to treat. Numerous studies with many different treatment modalities have resulted in only minor advances. No single agent or combination therapy has consistently shown a response proportion of 20% or higher. Interleukin-2 and interferon-alpha-based therapies are most commonly used to treat advanced disease, demonstrating low but reproducible response proportions in the 10% to 20% range, with durable responses of 5% or less. In the last few years, randomized studies have for the first time demonstrated a survival advantage for patients receiving systemic therapy, although this advantage is marginal. A number of novel treatment strategies are being investigated with some encouraging early results. The identification of new agents with better antitumor activity remains a high priority in the treatment of advanced renal cell carcinoma.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Interferon-alfa/uso terapêutico , Interleucina-2/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Anticorpos Monoclonais/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Transplante de Medula Óssea , Vacinas Anticâncer , Carcinoma de Células Renais/patologia , Quimioterapia Adjuvante , Ensaios Clínicos como Assunto , Intervalo Livre de Doença , Humanos , Neoplasias Renais/patologia , Prognóstico , Linfócitos T/imunologia
16.
Cancer Res ; 60(23): 6590-6, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11118039

RESUMO

Phorbol esters induce apoptosis in androgen-sensitive LNCaP cells, which express neutral endopeptidase (NEP), but not in androgen-independent prostate cancer (PC) cells, which lack NEP expression. We investigated the role of NEP in PC cell susceptibility to 12-O-tetradecanoylphorbol-13-acetate (TPA). Western analysis showed that expression of NEP and protein kinase Cdelta (PKCdelta) correlated with PC cell sensitivity to TPA-induced growth arrest and apoptosis in LNCaP cells and in TSU-Prl cells expressing an inducible wild-type NEP protein. Inhibition of NEP enzyme activity using the specific NEP inhibitor CGS24592, or inhibition of PKCdelta using Rottlerin at concentrations that inhibit PKCdelta but not PKCalpha, significantly inhibited TPA-induced growth inhibition and cell death. Furthermore, pulse-chase experiments showed PKCdelta is stabilized in LNCaP cells and in TSU-Pr1 cells overexpressing wild-type NEP compared with PC cells lacking NEP expression. This results from NEP inactivation of its neuropeptide substrates (bombesin and endothelin-1), which in the absence of NEP stimulate cSrc kinase activity and induce rapid degradation of PKCdelta protein. These results indicate that expression of enzymatically active NEP by PC cells is necessary for TPA-induced apoptosis, and that NEP inhibits neuropeptide-induced, cSrc-mediated PKCdelta degradation.


Assuntos
Apoptose/efeitos dos fármacos , Bombesina/antagonistas & inibidores , Endotelina-1/antagonistas & inibidores , Isoenzimas/metabolismo , Neprilisina/fisiologia , Neoplasias da Próstata/enzimologia , Proteína Quinase C/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Sequência de Aminoácidos , Apoptose/fisiologia , Bombesina/farmacologia , Proteína Tirosina Quinase CSK , Divisão Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Endotelina-1/farmacologia , Ativação Enzimática/efeitos dos fármacos , Inibidores do Crescimento/farmacologia , Humanos , Isoenzimas/biossíntese , Masculino , Dados de Sequência Molecular , Neprilisina/metabolismo , Neoplasias da Próstata/patologia , Proteína Quinase C/biossíntese , Proteína Quinase C-delta , Proteínas Tirosina Quinases/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos , Quinases da Família src
17.
J Interferon Cytokine Res ; 20(9): 787-94, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11032398

RESUMO

Retinoic acid (RA) can potentiate the antitumor effect of interferons (IFN) in a variety of tumor types, including renal cell carcinoma (RCC). The mechanisms by which RA and IFN increase the antitumor effects in RCC are unknown. We used growth assays and mobility shift assays to examine the effects of combining 13-cis-retinoic acid (CRA) and IFN-alpha (plus IFN-gamma) on proliferation and on the expression of the IFN-specific transcription factor IFN-stimulated gene factor 3 (ISGF3) in RCC cell lines. Combining CRA and IFN-alpha resulted in a significant increase in growth inhibition in four cell lines compared with IFN-alpha or CRA alone. Binding of nuclear extracts from RCC cells to an IFN-stimulated response element (ISRE) oligonucleotide probe following incubation with IFN-alpha was not increased by CRA but was significantly increased by pretreatment by IFN-gamma in a time-dependent fashion. Proliferation assays showed that sequential addition of IFN-gamma and IFN-alpha significantly increased growth inhibition. IFN-alpha but not IFN-gamma or CRA increased the cellular levels Stat2 and p48 but not Statl. IFN-gamma pretreatment enhanced the upregulation of p48 levels by IFN-alpha. Combining RA and IFN results in additive growth inhibition on RCC cell lines. This increase in growth inhibition is not mediated by increased ISGF3 expression.


Assuntos
Antineoplásicos/farmacologia , Interferon-alfa/farmacologia , Tretinoína/farmacologia , Carcinoma de Células Renais/patologia , Divisão Celular/efeitos dos fármacos , Interações Medicamentosas , Sinergismo Farmacológico , Humanos , Interferon gama/farmacologia , Isotretinoína/farmacologia , Neoplasias Renais/patologia , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
18.
J Clin Oncol ; 18(16): 2972-80, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10944130

RESUMO

PURPOSE: A randomized phase III trial was conducted to determine whether combination therapy with 13-cis-retinoic acid (13-CRA) plus interferon alfa-2a (IFNalpha2a) is superior to IFNalpha2a alone in patients with advanced renal cell carcinoma (RCC). PATIENTS AND METHODS: Two hundred eighty-four patients were randomized to treatment with IFNalpha2a plus 13-CRA or treatment with IFNalpha2a alone. IFNalpha2a was given daily subcutaneously, starting at a dose of 3 million units (MU). The dose was escalated every 7 days from 3 to 9 MU (by increments of 3 MU), unless >/= grade 2 toxicity occurred, in which case dose escalation was stopped. Patients randomized to combination therapy were given oral 13-CRA 1 mg/kg/d plus IFNalpha2a. Quality of life (QOL) was assessed. RESULTS: Complete or partial responses were achieved by 12% of patients treated with IFNalpha2a plus 13-CRA and 6% of patients treated with IFNalpha2a (P =.14). Median duration of response (complete and partial combined) in the group treated with the combination was 33 months (range, 9 to 50 months), versus 22 months (range, 5 to 38 months) for the second group (P =.03). Nineteen percent of patients treated with IFNalpha2a plus 13-CRA were progression-free at 24 months, compared with 10% of patients treated with IFNalpha2a alone (P =.05). Median survival time for all patients was 15 months, with no difference in survival between the two treatment arms (P =.26). QOL decreased during the first 8 weeks of treatment, and a partial recovery followed. Lower scores were associated with the combination therapy. CONCLUSION: Response proportion and survival did not improve significantly with the addition of 13-CRA to IFNalpha2a therapy in patients with advanced RCC. 13-CRA may lengthen response to IFNalpha2a therapy in patients with IFNalpha2a-sensitive tumors. Treatment, particularly the combination therapy, was associated with a decrease in QOL.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Interferon-alfa/uso terapêutico , Isotretinoína/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Administração Oral , Adulto , Idoso , Idoso de 80 Anos ou mais , Intervalo Livre de Doença , Feminino , Humanos , Injeções Subcutâneas , Interferon alfa-2 , Interferon-alfa/administração & dosagem , Isotretinoína/administração & dosagem , Masculino , Pessoa de Meia-Idade , Modelos Estatísticos , Análise Multivariada , Qualidade de Vida , Proteínas Recombinantes , Estatísticas não Paramétricas , Inquéritos e Questionários , Análise de Sobrevida , Resultado do Tratamento
19.
Endocrinology ; 141(5): 1699-704, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10803579

RESUMO

Androgen-mediated growth repression of androgen-independent prostate cancer (AIPC) cells has been reported in androgen-independent PC-3 cells overexpressing the androgen receptor, and in androgen-independent derivatives of LNCaP cells that develop following prolonged culture in androgen-free media. Using two models of AIPC, PC3/AR cells and LNCaP-OM1 cells, a subclone of LNCaP cells derived by prolonged culturing in charcoal-stripped media, we investigated whether expression of neutral endopeptidase 24.11 (NEP), a cell-surface peptidase that cleaves and inactivates neuropeptides implicated in the growth of AIPC, is induced by androgen, and whether NEP contributes to the observed androgen-mediated growth repression. These cell lines each express high levels of androgen receptor. Culturing in dihyrotestosterone (DHT) resulted in a 30-56% (PC3) and 35-43% (LNCaP-OM1) decrease in cell number over 7 days concomitant with a significant increase in NEP enzyme specific activity. Northern analysis detected an increase in NEP transcripts following DHT treatment in PC3/AR cells. The addition of the NEP enzyme inhibitor phosphoramidon to PC3 and LNCaP-OM1 or the NEP competitive inhibitor CGS 24592 to LNCaP-OM1 blocked the increase in NEP enzyme activity and reversed the DHT-induced growth inhibition. Neither phosphoramidon or CGS 24592 alone inhibited cell growth. Furthermore, the reversal of growth inhibition in LNCaPOM1 cells was dose dependent on the concentration of CGS 24592. These data indicate that androgen-induced growth repression of AIPC cells PC3 and LNCaP-OM1 results in part from androgen-induced expression of NEP in these cells.


Assuntos
Androgênios/fisiologia , Neprilisina/metabolismo , Neoplasias da Próstata/patologia , Receptores Androgênicos/biossíntese , Animais , Western Blotting , Divisão Celular/efeitos dos fármacos , Di-Hidrotestosterona/farmacologia , Masculino , Ratos , Receptores Androgênicos/genética , Transfecção , Células Tumorais Cultivadas
20.
Clin Cancer Res ; 6(5): 1664-70, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10815884

RESUMO

Neutral endopeptidase 24.11 (NEP) is a cell surface peptidase expressed by prostatic epithelial cells that cleaves and inactivates neuropeptide growth factors implicated in the growth of androgen-independent prostate cancer (PC). Decreased NEP expression in hormone-refractory metastatic PCs can result from hormonal therapies because NEP transcription is induced by androgens and down-regulated by androgen withdrawal. NEP is encoded by a gene that contains a 5' CpG island spanning a transcriptional regulatory region. In this study, we investigate whether DNA hypermethylation of the NEP promoter accompanies decreased NEP expression in PC cell lines and whether it occurs in human PC tissues in vivo. DNA isolated from PC cell lines and from normal and neoplastic human prostate tissues was restriction-digested with a methylation-sensitive restriction endonuclease and analyzed by Southern blot using a 5' sequence-specific NEP probe. Methylation-specific PCR was performed using PCR primers designed to discriminate between methylated and unmethylated alleles, and reverse transcription-PCR using NEP-specific primers was performed on cDNA extracted from PC cells treated with 5-aza-2'-deoxycytidine. Methylation of the NEP promoter was present in androgen-independent PC cell lines but not in androgen-dependent or small-cell derived PC cell lines and in 3 of 21 (14%) primary PCs from patients with androgen-dependent disease. Exposure of PC cells to the demethylating agent 5-aza-2'-deoxycytidine led to an increase in NEP transcripts in DU-145 and PC-3 cells. These data show that hypermethylation of the 5' CpG NEP island is associated with a loss of NEP expression in PC. Loss of NEP expression via hypermethylation of the NEP promoter may contribute to the development of neuropeptide-stimulated PCs.


Assuntos
Metilação de DNA , Neprilisina/metabolismo , Regiões Promotoras Genéticas , Neoplasias da Próstata/metabolismo , Azacitidina/análogos & derivados , Azacitidina/farmacologia , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Decitabina , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Neprilisina/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...