Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Br J Cancer ; 113(5): 817-26, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26151456

RESUMEN

BACKGROUND: Observational studies have reported a modest association between obesity and risk of ovarian cancer; however, whether it is also associated with survival and whether this association varies for the different histologic subtypes are not clear. We undertook an international collaborative analysis to assess the association between body mass index (BMI), assessed shortly before diagnosis, progression-free survival (PFS), ovarian cancer-specific survival and overall survival (OS) among women with invasive ovarian cancer. METHODS: We used original data from 21 studies, which included 12 390 women with ovarian carcinoma. We combined study-specific adjusted hazard ratios (HRs) using random-effects models to estimate pooled HRs (pHR). We further explored associations by histologic subtype. RESULTS: Overall, 6715 (54%) deaths occurred during follow-up. A significant OS disadvantage was observed for women who were obese (BMI: 30-34.9, pHR: 1.10 (95% confidence intervals (CIs): 0.99-1.23); BMI: ⩾35, pHR: 1.12 (95% CI: 1.01-1.25)). Results were similar for PFS and ovarian cancer-specific survival. In analyses stratified by histologic subtype, associations were strongest for women with low-grade serous (pHR: 1.12 per 5 kg m(-2)) and endometrioid subtypes (pHR: 1.08 per 5 kg m(-2)), and more modest for the high-grade serous (pHR: 1.04 per 5 kg m(-2)) subtype, but only the association with high-grade serous cancers was significant. CONCLUSIONS: Higher BMI is associated with adverse survival among the majority of women with ovarian cancer.


Asunto(s)
Neoplasias Glandulares y Epiteliales/patología , Obesidad/patología , Neoplasias Ováricas/patología , Índice de Masa Corporal , Carcinoma Epitelial de Ovario , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Neoplasias Glandulares y Epiteliales/mortalidad , Obesidad/mortalidad , Neoplasias Ováricas/mortalidad
2.
Gynecol Oncol ; 132(3): 566-72, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24368279

RESUMEN

OBJECTIVE: Folate is essential for DNA synthesis and methylation and is implicated in tumour progression. Few studies have examined its role in ovarian cancer survival. Our objective was to determine relationships between intake of folate, related one-carbon nutrients, single nucleotide polymorphisms (SNPs) in folate-metabolising genes and survival following ovarian cancer diagnosis. METHODS: This analysis included 1270 women with invasive epithelial ovarian cancer diagnosed in 2002-2006. Pre-diagnostic and some post-diagnostic lifestyle, dietary, and sociodemographic information was collected via self-administered questionnaires. DNA samples were genotyped for SNPs in methylenetetrahydrofolate reductase (MTHFR), methionine synthase (MTR) and methionine synthase reductase (MTRR) genes. Adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated using Cox regression. RESULTS: Multivariate analyses did not identify associations between higher pre-diagnostic intake of folate, folic acid, vitamins B2, B6, and B12, methionine, betaine or choline and survival overall. In stratified analyses, higher folic acid and folate intake was associated with significantly worse survival among women with mucinous tumours (HRs per 100 µg 1.30 and 1.43, respectively) and smokers (HRs per 100 µg 1.23 and 1.16 respectively). There was also a suggestion that higher supplemental folic acid use post-diagnosis was associated with worse survival (HR per 100 µg 1.03, 95%CI 1.00-1.05). MTHFR SNP rs2066470 was significantly associated with survival (per allele HR 0.81, 95%CI 0.67-0.98). CONCLUSIONS: Our data provide little evidence that folate intake affects ovarian cancer survival. However, combined effects with smoking, and findings within the mucinous subtype and for post-diagnosis folic acid, warrant further investigation.


Asunto(s)
Dieta/estadística & datos numéricos , Ácido Fólico/administración & dosificación , Micronutrientes/administración & dosificación , Neoplasias Glandulares y Epiteliales/mortalidad , Neoplasias Ováricas/mortalidad , Anciano , Consumo de Bebidas Alcohólicas/epidemiología , Australia/epidemiología , Carcinoma Epitelial de Ovario , Estudios de Casos y Controles , Estudios de Cohortes , Neoplasias de las Trompas Uterinas/genética , Neoplasias de las Trompas Uterinas/metabolismo , Neoplasias de las Trompas Uterinas/mortalidad , Neoplasias de las Trompas Uterinas/patología , Femenino , Ácido Fólico/metabolismo , Humanos , Persona de Mediana Edad , Análisis Multivariante , Estadificación de Neoplasias , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/metabolismo , Neoplasias Peritoneales/mortalidad , Neoplasias Peritoneales/patología , Polimorfismo de Nucleótido Simple , Fumar/epidemiología , Encuestas y Cuestionarios
3.
Int J Oncol ; 18(4): 697-704, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11251163

RESUMEN

7-Hydroxystaurosporine (UCN-01), a non-selective inhibitor of protein kinase C (PKC), and phorbol ester (PMA), a PKC activator, are undergoing clinical evaluations. We investigated the effects of UCN-01 and PMA on a panel of prostate cancer cell lines. While PMA induced p21WAF1/CIP1 and arrest growth of LNCaP cancer cells (IC50 = 0.5-1 nM), aggressive cancer cell lines (DU145, PC3, and PC3M) were resistant to PMA (IC50 >5000 nM). Low concentrations (25-50 nM) of UCN-01 abrogated PMA-induced p21 and growth arrest in LNCaP cells. These low doses of UCN-01 however did not inhibit proliferation of any prostate cancer cell line. PMA-sensitive LNCaP cells were resistant to clinically relevant concentrations of UCN-01 (IC50 = 1.2 microM), but UCN-01 inhibited growth of DU145 and PC3/3M with an IC50 of 200-400 nM. For comparison, PMA-sensitive HL60 leukemia cells were sensitive to UCN-01 due to rapid apoptosis caused by UCN-01. In PMA-resistant prostate cancer cells, UCN-01 downregulated cyclin D1, induced p21, caused morphological differentiation, and G1-phase arrest leading to slow cell death without caspase activation. Importantly, normal prostate epithelial cells (PrEC) were very sensitive to both PMA (IC50 = 0.2 nM) and UCN-01. In PrEC, UCN-01 downregulated cyclin D1 and arrest growth with an IC50 less than 100 nM. We conclude that loss of sensitivity to either UCN-01 or PMA accompanies progression of prostate cancer.


Asunto(s)
Alcaloides/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclinas/metabolismo , Neoplasias de la Próstata/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , ADN de Neoplasias/metabolismo , Regulación hacia Abajo , Resistencia a Antineoplásicos , Formazáns/metabolismo , Humanos , Immunoblotting , Masculino , Neoplasias de la Próstata/patología , Transducción de Señal , Estaurosporina/análogos & derivados , Sales de Tetrazolio/metabolismo , Transfección , Células Tumorales Cultivadas/citología , Células Tumorales Cultivadas/efectos de los fármacos
4.
Pharmacol Rev ; 53(1): 73-91, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11171939

RESUMEN

Prostate-specific antigen is a serine protease that is a member of the kallikrein family. It is widely used as an indicator of tumor burden and as a surrogate marker for disease progression in men with androgen-independent prostate cancer. It has been shown that the expression and/or secretion of this glycoprotein can be regulated by pharmacological agents. The effects of these agents on PSA may be independent of their effects on cell growth. For example, a pharmacological agent may down-regulate PSA expression/secretion but have no effect on tumor cell growth. In this case, a patient receiving this therapeutic agent might be falsely considered as having a clinical response. Alternatively, an agent might up-regulate PSA expression/secretion and have an inhibitory effect on cell growth. A patient receiving this therapeutic agent might be diagnosed with progressive disease unless an alternative method for assessing tumor burden is used. Thus, when an agent is to be evaluated in a clinical trial utilizing PSA as a marker for disease progression, it is important to prospectively test whether the agent has an effect on PSA expression and/or secretion. In addition, it is equally important to understand how these regulatory effects relate to cell growth. The purpose of this review is to describe several agents that have been tested for their regulatory effects on PSA and to discuss potential mechanisms of by which this regulation may occur. The implications of these findings in the evaluation of new agents in androgen-independent prostate cancer will be considered.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Antígeno Prostático Específico/biosíntesis , Antígeno Prostático Específico/genética , Animales , Regulación hacia Abajo/efectos de los fármacos , Humanos , Masculino , Receptores Androgénicos/efectos de los fármacos , Receptores Androgénicos/metabolismo , Regulación hacia Arriba/efectos de los fármacos
5.
J Urol ; 163(3): 1022-6, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10688042

RESUMEN

PURPOSE: Once a relapse occurs following primary endocrine treatment, metastatic prostate cancer is one of the most therapy-resistant human neoplasms. Ketoconazole is used for complete androgen deprivation, and recent data suggest it has direct activity against prostate cancer cells. MATERIALS AND METHODS: LNCaP, DU145, and PC3 cells, human prostate cancer cell lines, and HL60, a human leukemia cell line, were lysed and soluble proteins were harvested. Cells were plated in 96-well flat bottom plates and then exposed to the pharmacological agents, ketoconazole, vinblastine and paclitaxel. DNA synthesis was monitored by 3H-thymidine incorporation. RESULTS: We demonstrate that ketoconazole exerts a cytostatic effect on a panel of human prostate cancer cell lines, with IC50 of 4 to 5 microg./ml., 12 microg./ml., and 25 microg./ml. for LNCaP, PC3/PC3M, and DU145 cells, respectively. On the other hand, using microtubule-active drugs, vinblastine and paclitaxel, we found that PC3M and PC3 cells were more resistant than either DU145 or LNCaP cells. This resistance was associated with a lesser degree of Raf-1 and Bcl-2 phosphorylation following exposure to microtubule-active drugs. Combinations of microtubule-active drugs with ketoconazole were a beneficial treatment in DU145 cancer cells. Furthermore, ketoconazole blocked recovery of all the prostate cancer cell lines following 24 hours-pulse treatment with vinblastine. CONCLUSION: Pulse-administration of vinblastine followed by continuous administration of ketoconazole warrants investigation in the treatment of hormone-independent metastatic prostate cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Cetoconazol/uso terapéutico , Paclitaxel/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/secundario , Vinblastina/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Masculino , Microtúbulos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Células Tumorales Cultivadas
6.
Biochem Biophys Res Commun ; 268(1): 183-91, 2000 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-10652234

RESUMEN

Endostatin has demonstrated potent antiangiogenic and antitumor activity in mouse models. We have investigated the ex vivo rat aortic ring assay and a human vein model to assess the biological activity of murine and human endostatin. Rat aortic rings were exposed to recombinant murine endostatin (Spodoptera frugipera; Calbiochem, San Diego, CA) or recombinant human endostatin (Pichia pastoris; EntreMed, Rockville, MD). After 5 days, murine endostatin (500 microgram/ml) demonstrated inhibition of microvessel outgrowth with dose-dependent effects (down to 16 microgram/ml). No significant inhibition was observed with human endostatin in the rat assay. Human endostatin at 250 and 500 microgram/ml inhibited outgrowths from human saphenous vein rings after a 14-day incubation. Electron microscopy assessed the formation of basal lamina, confirming that the microvessels were progenitors of patent vessels. Immunostaining for Factor VIII or CD34 demonstrated that the microvessel cells were endothelial. BrdU incorporation assays supported the presence of proliferating endothelial cells, correlating with neovascularization from the aortic wall. We conclude that the rat aortic ring assay confirms the antiangiogenic activity of murine but not human endostatin, suggesting that the model may have species specificity. However, the human form shows biological activity against human vascular tissue.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Aorta Torácica/efectos de los fármacos , Colágeno/farmacología , Fragmentos de Péptidos/farmacología , Inhibidores de la Angiogénesis/genética , Animales , Antígenos CD34/metabolismo , Colágeno/genética , Endostatinas , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Humanos , Técnicas In Vitro , Masculino , Ratones , Microscopía Electrónica , Neovascularización Fisiológica/efectos de los fármacos , Fragmentos de Péptidos/genética , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Vena Safena/efectos de los fármacos , Especificidad de la Especie
7.
J Pharmacol Exp Ther ; 292(1): 31-7, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10604929

RESUMEN

The induction of angiogenesis is known to play a critical role in the successful growth, invasion, and metastasis of a tumor. A tumor will not grow beyond a few cubic millimeters without the formation of its own capillary network. Several antiangiogenic agents are under investigation in the clinic setting for the treatment of cancer. Carboxyamido-triazole (CAI), an inhibitor of Ca(2+)-mediated signal transduction, has been previously shown to inhibit angiogenesis in vitro and in vivo and to down-regulate matrix metalloproteinase-2 in vitro. Diminished levels of intracellular Ca(2+) result in decreased nitric-oxide synthase (NOS) activity and thereby inhibit the production and release of NO. The antiangiogenic activity of CAI was investigated by assessing microvessel growth from rat aortic segments and in cell culture using human aortic endothelial cells (HAECs). With these models, vascular endothelial growth factor (VEGF) and NOS production and secretion were evaluated. CAI concentrations ranging from 0.25 to 12.0 microg/ml inhibited new microvessel formation in rat aortic cultures and HAEC proliferation in a dose-dependent manner. Additionally, HAECs treated with CAI showed a dose-dependent decrease of NOS expression and a decrease in both VEGF expression and secretion. Rat aortic segments demonstrated decreased VEGF expression in situ on immunostaining. These data suggest that modulation of the NOS-NO-VEGF pathway through Ca(2+)-mediated signaling by CAI inhibits angiogenesis in vitro.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Factores de Crecimiento Endotelial/antagonistas & inhibidores , Linfocinas/antagonistas & inhibidores , Neovascularización Patológica/prevención & control , Óxido Nítrico Sintasa/antagonistas & inhibidores , Triazoles/farmacología , Animales , Aorta Torácica/fisiología , División Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Factores de Crecimiento Endotelial/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Humanos , Técnicas In Vitro , Linfocinas/metabolismo , Masculino , Microcirculación/metabolismo , Microcirculación/patología , Óxido Nítrico Sintasa/metabolismo , Ratas , Ratas Sprague-Dawley , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
8.
Neoplasma ; 46(2): 117-23, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10466436

RESUMEN

Following hormonal therapy, few treatment regimens have activity in metastatic prostate cancer. Cytotoxic agents have minimal activity in this disease. However, combinations of cytotoxic agents may be beneficial. The activity of estramustine, vinblastine, etoposide, and suramin on cell growth was evaluated. Prostate specific antigen (PSA) is routinely used as a surrogate marker for disease progression. Many pharmacological agents alter PSA levels independently of their effect on tumor growth, the effect of these agents on PSA secretion was determined. Each agent was evaluated alone and in combination with the other drugs in two prostate cancer cell lines. In LNCaP cells, estramustine and suramin were cytostatic, while vinblastine and etoposide were cytotoxic. Estramustine down-regulated etoposide PSA secretion, while suramin had no effect. The effects of etoposide and vinblastine on PSA secretion were not evaluable. In PC-3 cells, only etoposide was cytotoxic. Tandem combinations were more cytotoxic than single agents in both cell lines. The addition of a third agent to the tandem combination produced less cytotoxicity. In our hands, the best combinations were estramustine/vinblastine, suramin/vinblastine, and suramin/etoposide. These combinations yielded 20-60% higher cytotoxicity than any of the drugs alone.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , División Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Estramustina/administración & dosificación , Etopósido/administración & dosificación , Humanos , Masculino , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Suramina/administración & dosificación , Células Tumorales Cultivadas , Vinblastina/administración & dosificación
9.
Clin Cancer Res ; 5(7): 1738-44, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10430077

RESUMEN

Granulocyte macrophage colony-stimulating factor is a pleiotropic cytokine capable of inducing systemic immune responses against experimental and human tumors. To evaluate the efficacy of GM-CSF treatment in patients with hormone-refractory prostate cancer, we conducted sequential Phase II studies in 36 men with progressive disease after androgen deprivation and antiandrogen withdrawal. In a first cohort of patients (n = 23), GM-CSF was administered s.c. at a dose of 250 microg/m2 daily for 14 days of a 28-day treatment period. After we observed oscillating prostate-specific antigen (PSA) responses in several patients in this first cohort, a second trial was performed in which patients (n = 13) received maintenance GM-CSF (250 microg/m2 three times weekly) after the first 14 days of daily GM-CSF. All patients were treated until disease progression. Response was assessed by evaluation of serial changes in serum PSA and sequential imaging studies. In cohort I, 10 of 22 patients (45%) had a PSA versus time plot with a sawtooth pattern, with PSA declining during GM-CSF therapy and climbing during the off-therapy period; 5 patients had at least two consecutive declines in PSA, with a median response duration of 3.5 months. All but one patient in cohort II experienced a decline in PSA (median decline, 32%), but a PSA decline greater than 50% and sustained for more than 6 weeks was seen in only one patient, who had a >99% decline in PSA and an improvement in bone scan lasting for 14+ months. Changes in PSA levels could not be attributed to direct or indirect effects of GM-CSF on the PSA assay or down-regulation of PSA expression by GM-CSF. Toxicity was very mild, consisting primarily of transient constitutional symptoms and injection site reactions. These data suggest that GM-CSF may have antitumor activity in advanced prostate cancer, and the use of GM-CSF may be a confounding variable when PSA responses are used as an end point in clinical trials evaluating new regimens for the treatment of advanced prostate cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Anciano , Antineoplásicos/efectos adversos , División Celular/efectos de los fármacos , Estudios de Cohortes , Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos adversos , Humanos , Masculino , Estudios Prospectivos , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Resultado del Tratamiento
10.
J Mol Med (Berl) ; 77(5): 419-26, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10426191

RESUMEN

The action of androgens in the development and growth of prostate carcinomas is well documented. The androgen receptor (AR) facilitates androgen-induced regulation of genes involved in cellular proliferation and differentiation. Since the early 1940s androgen ablation has been the cornerstone of treatment for metastatic prostate cancer. Although initially highly effective, hormonal therapy is not curative, and resistant disease will ultimately prevail. Mutations that alter AR conformation, function, and regulation may provide a selective growth advantage for subpopulations of cells within the tumor that are then able to proliferate in an androgen-deprived environment. Clinically, these mutations are important because they may lead to the growth of androgen-independent tumors and progression to a refractory state. Further characterization of AR mutations will lead to a more thorough understanding of their role in the development of prostate carcinomas. This information, in addition to discovering which genes are regulated by the AR, can aid in the future development of more effective pharmacotherapy for prostate cancer.


Asunto(s)
Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Antagonistas de Andrógenos/uso terapéutico , Dihidrotestosterona/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Receptores Androgénicos/fisiología
11.
Cancer Chemother Pharmacol ; 43 Suppl: S78-84, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10357564

RESUMEN

Thalidomide has been shown to have species- and metabolic-dependent antiangiogenic activity in vitro and in vivo, suggesting its potential in treating human angiogenesis-dependent pathologies such as solid tumors. Based on promising preclinical studies, thalidomide has entered phase II clinical trials for prostate, brain, breast cancer, and Kaposi's sarcoma. However, the antiangiogenic mechanism of action is largely unresolved, as are its effects on tumor-associated gene expression, cytokine secretion, etc. We have investigated the effects of thalidomide on: 1) the secretion of prostate-specific antigen (PSA) in a human androgen-dependent prostate cell line; 2) growth and viability of human prostate cells; and 3) differential gene expression profiles of thalidomide-treated vs untreated human prostate cells. A human androgen-dependent prostate carcinoma cell line (LNCaP) and a human androgen-independent prostate carcinoma cell line (PC-3) were incubated with thalidomide 0.6, 6, or 60 microg/mL for 5-6 days. Secreted PSA from LNCaP cells was measured using a commercial enzyme-linked immunosorbant assay. Cell viability studies were conducted in both LNCaP and PC-3 cells using the same thalidomide concentrations. Furthermore, the differential gene expression of thalidomide-treated LNCaP cells was compared to that of untreated control cells using a commercially available human cancer cDNA expression array system. Thalidomide-treated LNCaP cells demonstrated increased PSA/cell levels at all concentrations tested compared to untreated control cells. Thalidomide demonstrated a cytostatic effect in LNCaP cells but had no appreciable effect on PC-3 cell viability compared to untreated control cells. Comparison of cDNA expression arrays hybridized with thalidomide-treated LNCaP cDNA probes suggests that thalidomide may up- or downregulate expression of angiogenesis-related genes, i.e., vitronectin, but these differential effects require further verification. Thalidomide over a range of doses has demonstrated nontoxic, cytostatic activity in LNCaP cells and significant upregulation of LNCaP cell PSA secretion in vitro. Furthermore, preliminary data from cDNA nucleic acid arrays of thalidomide-treated LNCaP cells suggest that thalidomide upregulates a potential angiogenic modulatory protein, the vitronectin precursor, which may eventually link thalidomide's antiangiogenic activity with modulation of angiogenic vascular integrin pathways.


Asunto(s)
Antineoplásicos/farmacología , Antígeno Prostático Específico/biosíntesis , Próstata/metabolismo , Talidomida/farmacología , División Celular , Línea Celular , Supervivencia Celular , ADN Complementario/análisis , Humanos , Masculino , Próstata/citología , Próstata/efectos de los fármacos , Antígeno Prostático Específico/análisis , Regulación hacia Arriba
12.
Br J Cancer ; 79(9-10): 1588-93, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10188911

RESUMEN

Prostate-specific antigen, PSA, is regarded as a reliable surrogate marker for androgen-independent prostate cancer (AIPC). Concern has been raised that investigational agents may affect PSA secretion without altering tumour growth or volume. In a phase I trial, several patients with AIPC had elevated serum PSA levels while receiving TNP-470 that reversed upon discontinuation. TNP-470 inhibits capillary growth in several angiogenesis models. These observations prompted us to determine if TNP-470, or its metabolite, AGM-1883, altered PSA secretion. Intracellular protein and transcriptional levels of PSA and androgen receptor were also determined. The highest TNP-470 concentration produced a 40.6% decrease in cell number; AGM-1883 had minimal effects on cell viability. PSA secretion per cell was induced 1.1- to 1.5-fold following TNP-470 exposure. The same trend was observed for AGM-1883. PSA and AR were transcriptionally up-regulated within 30 min after exposure to TNP-470. PSA transcription was increased 1.4-fold, while androgen receptor (AR) transcription was induced 1.2-fold. The increased PSA transcriptional activity accounts for the increased PSA secretion. Increased AR transcription was also reflected at the protein level. In conclusion, TNP-470 and AGM-1883 both up-regulated PSA making clinical utilization of this surrogate marker problematic.


Asunto(s)
Acetamidas/farmacología , Antibióticos Antineoplásicos/farmacología , Neovascularización Patológica/prevención & control , Antígeno Prostático Específico/efectos de los fármacos , Receptores Androgénicos/efectos de los fármacos , Sesquiterpenos/farmacología , Compuestos de Espiro/farmacología , Recuento de Células/efectos de los fármacos , Ciclohexanos , Humanos , Masculino , O-(Cloroacetilcarbamoil) Fumagilol , Antígeno Prostático Específico/metabolismo , Receptores Androgénicos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transcripción Genética/efectos de los fármacos , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo
13.
Biochem Pharmacol ; 55(11): 1827-34, 1998 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-9714301

RESUMEN

Thalidomide has been shown to be an inhibitor of angiogenesis in a rabbit cornea micropocket model; however, it has failed to demonstrate this activity in other models. These results suggest that the anti-angiogenic effects of thalidomide may only be observed following metabolic activation of the compound. This activation process may be species specific, similar to the teratogenic properties associated with thalidomide. Using a rat aorta model and human aortic endothelial cells, we co-incubated thalidomide in the presence of either human, rabbit, or rat liver microsomes. These experiments demonstrated that thalidomide inhibited microvessel formation from rat aortas and slowed human aortic endothelial cell proliferation in the presence of human or rabbit microsomes, but not in the presence of rat microsomes. In the absence of microsomes, thalidomide had no effect on either microvessel formation or cell proliferation, thus demonstrating that a metabolite of thalidomide is responsible for its anti-angiogenic effects and that this metabolite can be formed in both humans and rabbits, but not in rodents.


Asunto(s)
Antineoplásicos/farmacología , Endotelio Vascular/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Neovascularización Patológica/patología , Neovascularización Fisiológica/efectos de los fármacos , Talidomida/farmacología , Animales , Antineoplásicos/metabolismo , Aorta Torácica/citología , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , División Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Conejos , Ratas , Especificidad de la Especie , Talidomida/metabolismo , Células Tumorales Cultivadas
15.
Invasion Metastasis ; 18(4): 209-18, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-10640907

RESUMEN

Angiogenesis is required for tumor formation and growth; inhibition of angiogenesis is a promising new approach in cancer therapy. UCN-01, a protein kinase C (PKC) inhibitor, induces growth arrest and apoptosis in cancer cells and was recently introduced in a phase I clinical trial. We demonstrate that UCN-01, at concentrations lower than those necessary to inhibit cancer cell growth, inhibit proliferation of human endothelial cells in vitro. Moreover, UCN-01, at concentrations as low as 32 nM, prevent microvessel outgrowth from explant cultures of rat aortic rings. Since hypoxia activates hypoxia-inducible factor (HIF-1)-dependent transcription in cancer cells that, in a paracrine fashion, drive tumor angiogenesis, we investigated the effects of UCN-01 on HIF-1-responsive promoter constructs. We report that, in addition to direct inhibitory effects on endothelial cell growth, UCN-01 abrogates hypoxia-mediated transactivation of HIF-1-responsive promoters in a prostate cancer cell line. We conclude that UCN-01, at clinically relevant concentrations, exerts an anti-neovascularization effect by blocking two important steps in vessel formation: (1) the response of cancer cells to hypoxia, and (2) endothelial cell proliferation.


Asunto(s)
Alcaloides/farmacología , Inhibidores de la Angiogénesis/farmacología , Hipoxia de la Célula/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Neovascularización Patológica/tratamiento farmacológico , Proteína Quinasa C/antagonistas & inhibidores , Factores de Transcripción , Animales , Aorta Torácica/citología , Proteínas de Unión al ADN/fisiología , Endotelio Vascular/enzimología , Endotelio Vascular/patología , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Masculino , Proteínas Nucleares/fisiología , Técnicas de Cultivo de Órganos , Neoplasias de la Próstata/patología , Ratas , Ratas Sprague-Dawley , Estaurosporina/análogos & derivados , Transcripción Genética/efectos de los fármacos , Células Tumorales Cultivadas/efectos de los fármacos
16.
Expert Opin Investig Drugs ; 7(6): 889-904, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15992005

RESUMEN

Apoptosis is a genetically programmed cell death mechanism that appears to occur in all multicellular organisms. It is a normal process that serves to maintain cellular homeostasis. However, in many diseases there is a disruption in the equilibrium between cell proliferation and cell death that contributes directly to the disease. In these cases, a possible therapeutic intervention would be to restore the skewed equilibrium by pushing it in the desired direction through the use of pharmacological agents or genetic approaches. These observations have instigated substantial research in the field of apoptosis, resulting in an increasingly detailed analysis of the molecular mechanisms and the sequence of events that occur in this cell death pathway. In addition, by trying to understand this pathway, several potential therapeutic agents have arisen from those used in chemo-, radio-, and cytokine therapy. While these agents have been relatively successful, it is rare that their effect is complete. Thus, the search continues for a strategy to conquer those cells that are resistant to these regimens. Genetic approaches are novel and have been shown to be quite successful in several in vitro and animal models. They also tend to have low toxicity. It is believed that using a more traditional front-line approach of therapy, supplemented by appropriate genetic intervention, will allow substantial increases in the efficacy of treatment, while at the same time introducing little or no additional toxicity.

17.
Cancer Lett ; 113(1-2): 111-6, 1997 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-9065809

RESUMEN

Secondary hormonal manipulations are common following the failure of combined androgen blockade in patients with metastatic prostate cancer. Ketoconazole has been shown to have activity in this disease by inhibiting cytochrome P450 steroid hormone biosynthesis, thus inducing androgen deprivation. Gallium nitrate has been reported to target tumor tissue in vitro and some preliminary data suggests activity in patients with prostate cancer. Thus, we conducted a Phase II study of gallium nitrate in patients with androgen-independent prostate cancer. Two patients with progressive prostate cancer were removed from this study and subsequently placed on ketoconazole, as a palliative agent. Surprisingly, both of these patients had a greater than 50% decline in their prostate specific antigen (PSA) with this secondary endocrine maneuver. Based on this clinical observation, we conducted the following in vitro study to determine if there was a substantial additive effect of gallium nitrate followed by ketoconazole. Gallium nitrate or ketoconazole was added to the androgen-independent prostatic epithelial cell line, PC-3. One hundred and twenty hours (120 h) following the addition of one of the agents, the media was aspirated and the second agent was added to the wells. One plate was assayed every 24 h for cell viability using a non-isotopic cell proliferation assay kit. Cells treated with gallium nitrate followed by ketoconazole were 70-100% of control at the end of the gallium nitrate treatment; ketoconazole was then added and viability either remained constant or dropped steadily. Gallium nitrate by itself had a weak inhibitory effect on cell viability that only became apparent at the highest concentration evaluated. Ketoconazole, on the other hand, showed a substantial growth inhibition that was concentration-dependent. Cells treated with this agent alone showed a pronounced steady decrease in viability. Exposure to ketoconazole for 120 h followed by incubation in culture medium alone for 120 h caused a decrease in cell viability to 26.0% of control. Our in vitro results suggest that the combination of gallium nitrate and ketoconazole has no additive activity in the PC-3 cell line. Furthermore, this study confirms that ketoconazole added to prostate cancer cells has antiproliferative activity. The in vitro activity of ketoconazole has traditionally been thought to result from its inhibition of cytochrome P450-dependent enzymes responsible for steroidogenesis; however, an alternative hypothesis is necessary to explain the cytotoxic effect in the absence of adrenal and testicular androgen production as found in an in vitro system.


Asunto(s)
Antifúngicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Galio/farmacología , Cetoconazol/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Masculino , Factores de Tiempo , Células Tumorales Cultivadas
18.
Ann Pharmacother ; 31(1): 76-82, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8997471

RESUMEN

OBJECTIVE: To review the current literature regarding the role of apoptosis in the development of malignant cells and how the induction of this pathway could be used in cancer therapy. DATA SOURCE: A MEDLINE search of basic science articles pertinent to the understanding of the normal physiologic process of apoptosis was conducted. STUDY SELECTION: Because of the rapidly growing literature regarding apoptosis, only articles describing key processes in the biology of the cell and the genetic control of apoptosis were included. DATA SYNTHESIS: Apoptosis is imperative for host survival since it discards unwanted, damaged, and atypical cells. The process is therefore implicated in the continuous regulation of development, differentiation, and homeostasis. Furthermore, apoptosis is a response to physiologic and pathologic stresses that disrupt the balanced rates of cell generation and elimination. In a disease such as cancer, there is a lack of equilibrium between the rates of cell division and cell death; agents that promote or suppress apoptosis can manipulate these rates, influencing the anomalous accumulation of neoplastic cells. Pharmacologic manipulation of apoptosis can manipulate these rates, influencing the anomalous accumulation represents a novel approach in targeting malignant cells and has far-reaching implications for new directions in cancer therapy. CONCLUSIONS: Apoptosis is a highly organized physiologic mechanism of destroying injured and abnormal cells as well as maintaining homeostasis in multicellular organisms. Both the activation and inhibition of apoptosis are tightly controlled. Pharmacologic manipulation of this pathway is a novel therapeutic target in cancer therapy.


Asunto(s)
Apoptosis/genética , Apoptosis/fisiología , Neoplasias/terapia , Animales , Muerte Celular/genética , Muerte Celular/fisiología , Humanos
19.
Anim Genet ; 23(3): 203-10, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1503256

RESUMEN

Bivariate flow karyotypes of chromosomes from sheep, cattle and pig lymphocytes and from a cattle-mouse somatic cell hybrid line were obtained using a dual laser fluorescence-activated cell sorter (FACS). Pig chromosomes were resolved into 19-20 peaks, indicating that most, if not all, pig chromosomes could be separated by this technique. Sheep chromosomes showed incomplete separation but three clear peaks, presumably representing the three large metacentric chromosomes, plus five other clusters were obtained. Cattle chromosomes showed poor separation but about four peaks could be distinguished, indicating that certain chromosomes could be sorted in this species. The use of cattle-mouse hybrids may enable other individual cattle chromosomes to be obtained. It is concluded that FACS separation will be a useful additional tool for gene mapping.


Asunto(s)
Animales Domésticos/genética , Mapeo Cromosómico/métodos , Animales , Bovinos , Línea Celular , Separación Celular , Células Cultivadas , Estudios de Factibilidad , Citometría de Flujo/métodos , Células Híbridas/fisiología , Cariotipificación/métodos , Linfocitos/citología , Ratones , Ovinos , Porcinos
20.
Cytogenet Cell Genet ; 61(2): 128-31, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1395721

RESUMEN

Chromosomes were prepared from lymphocytes of a male domestic pig and flow-sorted on a dual-laser FACS. Twenty spots were observed, corresponding to the known pig karyotype of 18 pairs of autosomes plus the X and Y. DNA was isolated from 10,000 copies of the presumed chromosome 1 spot, restricted with Sau3A, ligated into the vector pGEM4z, and PCR amplified using universal primers; the products were then re-ligated into pUC18. After transformation into Escherichia coli, 210,000 independent colonies were obtained, 5% of which contained only vector DNA. The average insert size of the library was 405 bp. Southern blotting revealed that 36% of the clones contained single-copy DNA and that the remainder contained moderately or highly repetitive DNA. Screening with a (CA)n probe revealed that roughly 1% of the clones contained microsatellite sequences. A bulk insert of the library was biotinylated by PCR and used as a probe for chromosomal in situ suppression hybridization to pig chromosomes, which confirmed that the library is specific for chromosome 1. However, sequences from the centromeric and telomeric regions seem to be underrepresented in the library.


Asunto(s)
Mapeo Cromosómico , ADN/genética , Porcinos/genética , Animales , Secuencia de Bases , Células Cultivadas , ADN/aislamiento & purificación , Biblioteca de Genes , Hibridación in Situ , Cariotipificación , Linfocitos/fisiología , Masculino , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...