Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 166(4): 1041-1054, 2016 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-27499020

RESUMEN

We used clinical tissue from lethal metastatic castration-resistant prostate cancer (CRPC) patients obtained at rapid autopsy to evaluate diverse genomic, transcriptomic, and phosphoproteomic datasets for pathway analysis. Using Tied Diffusion through Interacting Events (TieDIE), we integrated differentially expressed master transcriptional regulators, functionally mutated genes, and differentially activated kinases in CRPC tissues to synthesize a robust signaling network consisting of druggable kinase pathways. Using MSigDB hallmark gene sets, six major signaling pathways with phosphorylation of several key residues were significantly enriched in CRPC tumors after incorporation of phosphoproteomic data. Individual autopsy profiles developed using these hallmarks revealed clinically relevant pathway information potentially suitable for patient stratification and targeted therapies in late stage prostate cancer. Here, we describe phosphorylation-based cancer hallmarks using integrated personalized signatures (pCHIPS) that shed light on the diversity of activated signaling pathways in metastatic CRPC while providing an integrative, pathway-based reference for drug prioritization in individual patients.


Asunto(s)
Fosfoproteínas/análisis , Neoplasias de la Próstata Resistentes a la Castración/química , Proteoma/análisis , Algoritmos , Humanos , Masculino , Medicina de Precisión , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Transducción de Señal , Transcriptoma
2.
Proc Natl Acad Sci U S A ; 117(45): 28092-28101, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33106406

RESUMEN

Prostate cancer (CaP) driven by androgen receptor (AR) is treated with androgen deprivation; however, therapy failure results in lethal castration-resistant prostate cancer (CRPC). AR-low/negative (ARL/-) CRPC subtypes have recently been characterized and cannot be targeted by hormonal therapies, resulting in poor prognosis. RNA-binding protein (RBP)/helicase DDX3 (DEAD-box helicase 3 X-linked) is a key component of stress granules (SG) and is postulated to affect protein translation. Here, we investigated DDX3-mediated posttranscriptional regulation of AR mRNA (messenger RNA) in CRPC. Using patient samples and preclinical models, we objectively quantified DDX3 and AR expression in ARL/- CRPC. We utilized CRPC models to identify DDX3:AR mRNA complexes by RNA immunoprecipitation, assess the effects of DDX3 gain/loss-of-function on AR expression and signaling, and address clinical implications of targeting DDX3 by assessing sensitivity to AR-signaling inhibitors (ARSI) in CRPC xenografts in vivo. ARL/- CRPC expressed abundant AR mRNA despite diminished levels of AR protein. DDX3 protein was highly expressed in ARL/- CRPC, where it bound to AR mRNA. Consistent with a repressive regulatory role, DDX3 localized to cytoplasmic puncta with SG marker PABP1 in CRPC. While induction of DDX3-nucleated SGs resulted in decreased AR protein expression, inhibiting DDX3 was sufficient to restore 1) AR protein expression, 2) AR signaling, and 3) sensitivity to ARSI in vitro and in vivo. Our findings implicate the RBP protein DDX3 as a mechanism of posttranscriptional regulation for AR in CRPC. Clinically, DDX3 may be targetable for sensitizing ARL/- CRPC to AR-directed therapies.


Asunto(s)
ARN Helicasas DEAD-box/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/genética , Animales , Línea Celular Tumoral , ARN Helicasas DEAD-box/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Próstata/química , Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/química , Neoplasias de la Próstata Resistentes a la Castración/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Androgénicos/metabolismo
3.
Prostate ; 81(3): 147-156, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33368414

RESUMEN

BACKGROUND: The treatment of castration-resistant prostate cancer (CRPC) is a urological issue. Recent studies have revealed cancer promotion via the C5a-C5a receptor (C5aR) system. To establish a new therapeutic target for CRPC, we investigated an association of the system with CRPC progression and evasion from the antitumor immune responses. METHODS: C5aR and PD-L1 were immunostained in the prostate cancer (PC) tissues. The relationship of PC C5aR expression to clinicopathological parameters was analyzed. CRPC cell lines were examined for C5aR expression by real-time reverse transcription polymerase chain reaction, immunoblotting, and flow cytometry. C5a effects were examined on CRPC cell glutamine consumption, proliferation, invasion, and PD-L1 expression. RESULTS: PC cells expressed C5aR in 83 of the 161 patients (52%) and in three of the six CRPC patients. Basal cells, but not luminal cells, of noncancerous prostate glands expressed C5aR. Three CRPC cell lines expressed C5aR. C5a increased CRPC cell glutamine consumption 2.1-fold, proliferation 1.3-1.6-fold, and invasion 2-3-fold in a C5a-concentration and a C5aR-dependent manner. High expression of C5aR did not relate to the PC patients' clinical parameters but the PD-L1-positive rate was higher in the C5aR high-expression patients (37.5%) compared to low- or no expression patients (17.8%), and double-positive PC cells were present. C5a increased CRPC cell PD-L1 production 1.4-fold and cell-surface expression 2.6-fold. CONCLUSIONS: C5aR expression of PC cells in patients' tissues and C5a augmentation of C5aR-dependent CRPC proliferation, invasion, and PD-L1 expression suggested participation of the C5a-C5aR system in CRPC promotion and evasion from antitumor immune responses. Targeting this signaling pathway may provide a useful therapeutic option for CRPC.


Asunto(s)
Antígeno B7-H1/análisis , Proliferación Celular , Invasividad Neoplásica/patología , Neoplasias de la Próstata Resistentes a la Castración/química , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptor de Anafilatoxina C5a/análisis , Anciano , Antígeno B7-H1/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Complemento C5a/farmacología , Expresión Génica/efectos de los fármacos , Glutamina/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Células PC-3 , Neoplasias de la Próstata Resistentes a la Castración/genética , ARN Mensajero/análisis , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/fisiología
4.
Prostate ; 78(8): 576-582, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29508425

RESUMEN

OBJECTIVE: We examined whether androgen receptor splice variant 7 (AR-V7) in circulating tumor cell(CTC)clusters can be used to predict survival in patients with bone metastatic castration resistant-prostate cancer (mCRPC) treated with abiraterone or enzalutamide. METHODS: We retrospectively enrolled 98 patients with CRPC on abiraterone or enzalutamide, and investigated the prognostic value of CTC cluster detection (+ v -) and AR-V7 detection (+ v -) using a CTC cluster detection - based AR-V7 mRNA assay. We examined ≤50% prostate-specific antigen (PSA) responses, PSA progression-free survival (PSA-PFS), clinical and radiological progression-free survival (radiologic PSF), and overall survival (OS). We then assessed whether AR-V7 expression in CTC clusters identified after On-chip multi-imaging flow cytometry was related to disease progression and survival after first-line systemic therapy. RESULTS: All abiraterone-treated or enzalutamide-treated patients received prior docetaxel. The median follow-up was 20.7 (range: 3.0-37.0) months in the abiraterone and enzalutamide cohorts, respectively. Forty-nine of the 98 men (50.0%) were CTC cluster (-), 23 of the 98 men (23.5%) were CTC cluster(+)/AR-V7(-), and 26 of the 98 men (26.5%) were CTC cluster(+)/AR-V7(+). CTC cluster(+)/AR-V7(+) patients were more likely to have EOD ≥3 at diagnosis (P = 0.003), pain (P = 0.023), higher alkaline phosphatase levels (P < 0.001), and visceral metastases (P < 0.001). On multivariable analysis, pretherapy CTC cluster(+), CTC cluster(+)/AR-V7(-), and ALP >UNL were independently associated with a poor PSA-PFS, radiographic PFS, and OS in abiraterone-treated patients and enzalutamide-treated patients. CONCLUSION: The CTC clusters and AR-V7-positive CTC clusters detected were important for assessing the response to abiraterone or enzalutamide therapy and for predicting disease outcome.


Asunto(s)
Acetato de Abiraterona/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias Óseas/química , Feniltiohidantoína/análogos & derivados , Neoplasias de la Próstata Resistentes a la Castración/química , Receptores Androgénicos/análisis , Anciano , Benzamidas , Biomarcadores de Tumor/análisis , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/mortalidad , Neoplasias Óseas/secundario , Resistencia a Antineoplásicos , Humanos , Masculino , Células Neoplásicas Circulantes/química , Nitrilos , Feniltiohidantoína/uso terapéutico , Valor Predictivo de las Pruebas , Pronóstico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/mortalidad , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/genética
5.
Hinyokika Kiyo ; 63(2): 57-62, 2017 Feb.
Artículo en Japonés | MEDLINE | ID: mdl-28264534

RESUMEN

We retrospectively evaluated the efficacy and toxicity of low-dose estramustine phosphate (EMP) monotherapy in patients with castration-resistant prostate cancer (CRPC). We administered EMP at 140 or 280 mg/day to 89 patients between January 2003 and December 2012. None of the patients were receiving concomitant dexamethasone and none had ever been treated with docetaxel. Fifty-three patients (59.6%) experienced a decline in prostate-specific antigen (PSA) levels, including 20 (22.5%) with a decline of more than 50%. The median time to PSA progression was 90 days. PSA-progression-free survival was significantly longer in patients treated with EMP 140 mg compared with patients treated with EMP 280 mg, and there was no significant difference in the incidence of adverse events between the two groups. The most frequent toxicities were nausea and anorexia. Two patients had grade 3 adverse events of pulmonary embolism and liver dysfunction. EMP treatment was discontinued in nine patients (10.1%) because of side effects (nausea and anorexia in 7, liver dysfunction and lacunar infarction in 1). Low-dose EMP monotherapy is well tolerated and can effectively reduce PSA levels.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Estramustina/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Antineoplásicos Hormonales/administración & dosificación , Progresión de la Enfermedad , Estramustina/administración & dosificación , Humanos , Masculino , Persona de Mediana Edad , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata Resistentes a la Castración/química , Neoplasias de la Próstata Resistentes a la Castración/diagnóstico , Estudios Retrospectivos , Resultado del Tratamiento
6.
Prostate ; 76(10): 927-36, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26997363

RESUMEN

BACKGROUND: Resistance to docetaxel is common in metastatic castration-resistant prostate cancer (mCRPC) and may be caused by sub-therapeutic intratumoral drug concentrations. Cabazitaxel demonstrated survival benefit in docetaxel-pretreated and docetaxel-refractory patients. In this study, we investigated whether the superior antitumor activity of cabazitaxel in mCRPC is explained by higher intratumoral cabazitaxel levels. Since recent studies suggest a reduced efficacy of docetaxel following treatment with novel androgen receptor (AR)-targeted agents, we also investigated taxane efficacy in an enzalutamide-resistant tumor model. METHODS: Intratumoral concentrations of docetaxel and cabazitaxel were correlated with antitumor activity in docetaxel-naïve, docetaxel-resistant, and enzalutamide-resistant patient-derived xenografts (PDXs) of prostate cancer. RESULTS: Intratumoral drug levels were negatively related to intrinsic and acquired resistance to docetaxel. Also, the observed stronger antitumor activity of cabazitaxel was associated with increased cumulative exposure and higher intratumoral of cabazitaxel concentrations in all PDXs. CONCLUSIONS: The superior antitumor activity of cabazitaxel in docetaxel- and enzalutamide-resistant tumors can be partly attributed to higher intratumoral drug concentrations. Especially for patients who are intrinsically resistant to docetaxel resulting from suboptimal intratumoral docetaxel concentrations, cabazitaxel may be the preferred chemotherapeutic agent. Prostate 76:927-936, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración/dietoterapia , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Taxoides/farmacocinética , Taxoides/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Benzamidas , Docetaxel , Resistencia a Antineoplásicos , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Desnudos , Nitrilos , Feniltiohidantoína/análogos & derivados , Feniltiohidantoína/análisis , Feniltiohidantoína/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/química , Receptores Androgénicos/efectos de los fármacos , Taxoides/análisis , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Int Braz J Urol ; 41(5): 1002-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26689527

RESUMEN

Meclofenamic acid is a nonsteroidal anti-inflammatory drug that has shown therapeutic potential for different types of cancers, including androgen-independent prostate neoplasms. The antitumor effect of diverse nonsteroidal anti-inflammatory drugs has been shown to be accompanied by histological and molecular changes that are responsible for this beneficial effect. The objective of the present work was to analyze the histological changes caused by meclofenamic acid in androgen-independent prostate cancer. Tumors were created in a nude mouse model using PC3 cancerous human cells. Meclofenamic acid (10 mg/kg/day; experimental group, n=5) or saline solution (control group, n=5) was administered intraperitoneally for twenty days. Histological analysis was then carried out on the tumors, describing changes in the cellular architecture, fibrosis, and quantification of cellular proliferation and tumor vasculature. Meclofenamic acid causes histological changes that indicate less tumor aggression (less hypercellularity, fewer atypical mitoses, and fewer nuclear polymorphisms), an increase in fibrosis, and reduced cellular proliferation and tumor vascularity. Further studies are needed to evaluate the molecular changes that cause the beneficial and therapeutic effects of meclofenamic acid in androgen-independent prostate cancer.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores de la Ciclooxigenasa/farmacología , Ácido Meclofenámico/farmacología , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/patología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Fibrosis , Humanos , Inmunohistoquímica , Masculino , Ratones Desnudos , Invasividad Neoplásica , Neovascularización Patológica/tratamiento farmacológico , Próstata/efectos de los fármacos , Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/química , Reproducibilidad de los Resultados
8.
Prostate ; 74(3): 306-13, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24249383

RESUMEN

BACKGROUND: Castration-resistant prostate cancer (CRPC) represents a challenge to treat with no effective treatment options available. We recently identified serum response factor (SRF) as a key transcription factor in an in vitro model of castration resistance where we showed that SRF inhibition resulted in reduced cellular proliferation. We also demonstrated an association between SRF protein expression and CRPC in a cohort of castrate-resistant transurethral resections of the prostate (TURPS). The mechanisms regulating the growth of CRPC bone and visceral metastases have not been explored in depth due to the paucity of patient-related material available for analysis. In this study, we aim to evaluate SRF protein expression in prostate cancer (PCa) metastases, which has not previously been reported. METHODS AND RESULTS: We evaluated the nuclear tissue expression profile of SRF by immunohistochemistry in 151 metastatic sites from 42 patients who died of advanced PCa. No relationship between SRF nuclear expression and the site of metastasis was observed (P = 0.824). However, a negative association between SRF nuclear expression in bone metastases and survival from (a) diagnosis with PCa (P = 0.005) and (b) diagnosis with CRPC (P = 0.029) was seen. These results demonstrate that SRF nuclear expression in bone metastases is associated with survival, with patients with the shortest survival showing high SRF nuclear expression and patients with the longest survival having low SRF nuclear expression. CONCLUSION: Our study indicates that SRF is a key factor determining patients' survival in metastatic CRPC and therefore may represent a promising target for future therapies.


Asunto(s)
Neoplasias Óseas/química , Neoplasias Óseas/secundario , Metástasis de la Neoplasia , Neoplasias de la Próstata Resistentes a la Castración/química , Neoplasias de la Próstata/química , Factor de Respuesta Sérica/análisis , Núcleo Celular/química , Humanos , Inmunohistoquímica , Masculino , Análisis Multivariante , Próstata/química , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata Resistentes a la Castración/mortalidad , Neoplasias de la Próstata Resistentes a la Castración/patología , Tasa de Supervivencia
12.
Clin Cancer Res ; 27(21): 6001-6011, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34407973

RESUMEN

PURPOSE: In metastatic castration-resistant prostate cancer (mCRPC) low serum androgens prior to starting abiraterone acetate (AA) is associated with more rapid progression. We evaluated the effect of AA on androgens in castration-resistant prostate cancer (CRPC) metastases and associations of intratumoral androgens with response. EXPERIMENTAL DESIGN: We performed a phase II study of AA plus prednisone in mCRPC. The primary outcome was tissue testosterone at 4 weeks. Exploratory outcomes were association of steroid levels and genomic alterations with response, and escalating AA to 2,000 mg at progression. RESULTS: Twenty-nine of 30 men were evaluable. Testosterone in metastatic biopsies became undetectable at 4 weeks (P < 0.001). Serum and tissue dehydroepiandrosterone sulfate (DHEAS) remained detectable in many patients and was not increased at progression. Serum and tissue DHEAS in the lowest quartile (pretreatment), serum DHEAS in the lowest quartile (4 weeks), and undetectable tissue DHEAS (on-therapy) associated with rapid progression (20 vs. 48 weeks, P = 0.0018; 20 vs. 52 weeks, P = 0.0003; 14 vs. 40 weeks, P = 0.0001; 20 vs. 56 weeks, P = 0.02, respectively). One of 16 men escalating to 2,000 mg had a 30% PSA decline; 13 developed radiographic progression by 12 weeks. Among patients with high serum DHEAS at baseline, wild-type (WT) PTEN status associated with longer response (61 vs. 33 weeks, P = 0.02). CONCLUSIONS: Low-circulating adrenal androgen levels are strongly associated with an androgen-poor tumor microenvironment and with poor response to AA. Patients with CRPC with higher serum DHEAS levels may benefit from dual androgen receptor (AR)-pathway inhibition, while those in the lowest quartile may require combinations with non-AR-directed therapy.


Asunto(s)
Andrógenos/análisis , Andrógenos/sangre , Androstenos/administración & dosificación , Antineoplásicos Hormonales/administración & dosificación , Prednisona/administración & dosificación , Neoplasias de la Próstata Resistentes a la Castración/sangre , Neoplasias de la Próstata Resistentes a la Castración/química , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Corteza Suprarrenal/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Andrógenos/metabolismo , Correlación de Datos , Combinación de Medicamentos , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Neoplasias de la Próstata Resistentes a la Castración/patología , Resultado del Tratamiento
13.
Eur Urol ; 76(4): 469-478, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31345636

RESUMEN

BACKGROUND: Prostate-specific membrane antigen (PSMA; folate hydrolase) prostate cancer (PC) expression has theranostic utility. OBJECTIVE: To elucidate PC PSMA expression and associate this with defective DNA damage repair (DDR). DESIGN, SETTING, AND PARTICIPANTS: Membranous PSMA (mPSMA) expression was scored immunohistochemically from metastatic castration-resistant PC (mCRPC) and matching, same-patient, diagnostic biopsies, and correlated with next-generation sequencing (NGS) and clinical outcome data. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Expression of mPSMA was quantitated by modified H-score. Patient DNA was tested by NGS. Gene expression and activity scores were determined from mCRPC transcriptomes. Statistical correlations utilised Wilcoxon signed rank tests, survival was estimated by Kaplan-Meier test, and sample heterogeneity was quantified by Shannon's diversity index. RESULTS AND LIMITATIONS: Expression of mPSMA at diagnosis was associated with higher Gleason grade (p=0.04) and worse overall survival (p=0.006). Overall, mPSMA expression levels increased at mCRPC (median H-score [interquartile range]: castration-sensitive prostate cancer [CSPC] 17.5 [0.0-60.0] vs mCRPC 55.0 [2.8-117.5]). Surprisingly, 42% (n=16) of CSPC and 27% (n=16) of mCRPC tissues sampled had no detectable mPSMA (H-score <10). Marked intratumour heterogeneity of mPSMA expression, with foci containing no detectable PSMA, was observed in all mPSMA expressing CSPC (100%) and 37 (84%) mCRPC biopsies. Heterogeneous intrapatient mPSMA expression between metastases was also observed, with the lowest expression in liver metastases. Tumours with DDR had higher mPSMA expression (p=0.016; 87.5 [25.0-247.5] vs 20 [0.3-98.8]; difference in medians 60 [5.0-95.0]); validation cohort studies confirmed higher mPSMA expression in patients with deleterious aberrations in BRCA2 (p<0.001; median H-score: 300 [165-300]; difference in medians 195.0 [100.0-270.0]) and ATM (p=0.005; 212.5 [136.3-300]; difference in medians 140.0 [55.0-200]) than in molecularly unselected mCRPC biopsies (55.0 [2.75-117.5]). Validation studies using mCRPC transcriptomes corroborated these findings, also indicating that SOX2 high tumours have low PSMA expression. CONCLUSIONS: Membranous PSMA expression is upregulated in some but not all PCs, with mPSMA expression demonstrating marked inter- and intrapatient heterogeneity. DDR aberrations are associated with higher mPSMA expression and merit further evaluation as predictive biomarkers of response for PSMA-targeted therapies in larger, prospective cohorts. PATIENT SUMMARY: Through analysis of prostate cancer samples, we report that the presence of prostate-specific membrane antigen (PSMA) is extremely variable both within one patient and between different patients. This may limit the usefulness of PSMA scans and PSMA-targeted therapies. We show for the first time that prostate cancers with defective DNA repair produce more PSMA and so may respond better to PSMA-targeting treatments.


Asunto(s)
Antígenos de Superficie/biosíntesis , Reparación del ADN , Glutamato Carboxipeptidasa II/biosíntesis , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Antígenos de Superficie/análisis , Glutamato Carboxipeptidasa II/análisis , Humanos , Masculino , Neoplasias de la Próstata Resistentes a la Castración/química , Estudios Retrospectivos
14.
Oncol Rep ; 41(2): 1275-1283, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30483800

RESUMEN

Prostate cancer (PCa) is the second most common type of male malignancy worldwide. The transcription factor zinc finger E­box binding homeobox 1 (ZEB1) is associated with epithelial­mesenchymal transition and is also involved in regulation of androgen receptor (AR) expression, the main ligands of which are testosterone and dihydrotestosterone (DHT). These androgens are synthesized through the steroidogenic pathway within the prostate, and their synthesis is altered in PCa. The present study aimed to determine the ZEB1­induced alterations in androgen synthesis and AR expression in the DU145 PCa cell line. Reverse transcription­quantitative polymerase chain reaction, western blotting and immunocytochemistry were used to determine the mRNA and protein expression levels, and cellular localization of steroidogenic pathway enzymes in the DU145 cell line in response to ZEB1 silencing. Furthermore, the concentrations of testosterone and DHT were detected in cell culture medium using ELISA. ZEB1­silenced cells exhibited an increase in testosterone and DHT production, an increase in AR expression and an alteration in the steroidogenic pathway. In particular, steroidogenic acute regulatory protein and 5α­reductase 2 expression levels were decreased, whereas cytochrome P450 family 17 subfamily A member 1, 5α­reductase 1, aldo­keto reductase family 1 member D1 and aldo­keto reductase family 1 member C2 expression levels were increased. In conclusion, the present study provided novel information regarding the regulation of intratumoral androgen production in PCa, which is relevant for the progression of the disease to a castration­resistant form.


Asunto(s)
Dihidrotestosterona/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Testosterona/biosíntesis , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/fisiología , Línea Celular Tumoral , Dihidrotestosterona/análisis , Silenciador del Gen , Humanos , Masculino , Próstata/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/química , Receptores Androgénicos/metabolismo , Testosterona/análisis , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
15.
Endocrinology ; 158(10): 3152-3161, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28977599

RESUMEN

The androgen receptor (AR) plays a critical role in the development of castration-resistant prostate cancer (CRPC) as well as in the resistance to the second-generation AR antagonist enzalutamide and the selective inhibitor of cytochrome P450 17A1 (CYP17A1) abiraterone. Novel agents targeting AR may inhibit the growth of prostate cancer cells resistant to enzalutamide and/or abiraterone. Through a high-throughput/high-content screening of a 220,000-member small molecule library, we have previously identified 2-[(isoxazol-4-ylmethyl)thio]-1-(4-phenylpiperazin-1-yl)ethanone (IMTPPE) (SID 3712502) as a novel small molecule capable of inhibiting AR transcriptional activity and protein level in C4-2 prostate cancer cells. In this study, we show that IMTPPE inhibits AR-target gene expression using real-time polymerase chain reaction, Western blot, and luciferase assays. IMTPPE inhibited proliferation of AR-positive, but not AR-negative, prostate cancer cells in culture. IMTPPE inhibited the transcriptional activity of a mutant AR lacking the ligand-binding domain (LBD), indicating that IMTPPE inhibition of AR is independent of the LBD. Furthermore, animal studies showed that IMTPPE inhibited the growth of 22Rv1 xenograft tumor, a model for enzalutamide-resistant prostate cancer. These findings suggest that IMTPPE is a potential lead compound for developing clinical candidates for the treatment of CRPC, including those resistant to enzalutamide.


Asunto(s)
Antagonistas de Receptores Androgénicos/farmacología , Isoxazoles/farmacología , Piperazinas/farmacología , Neoplasias de la Próstata Resistentes a la Castración/química , Receptores Androgénicos/fisiología , Animales , Benzamidas , Bexaroteno , Sitios de Unión/genética , Sitios de Unión/fisiología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Nitrilos , Feniltiohidantoína/análogos & derivados , Regiones Promotoras Genéticas/efectos de los fármacos , Isoformas de Proteínas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Androgénicos/análisis , Receptores Androgénicos/genética , Tetrahidronaftalenos , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Int Urol Nephrol ; 49(3): 369-380, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27987128

RESUMEN

Glucocorticoids are a common class of adjuvant drugs for the treatment of castration-resistant prostate cancer (CRPC) combined with antitumour or antiandrogen agents. Glucocorticoids are administered clinically because they ameliorate toxic side effects and have inhibitory effects on adrenal androgen production, acting as a pituitary suppressant. However, their effects on prostate cancer cells especially the castration resistance prostate cancer cells are poorly defined. Glucocorticoids exert effects depend to a great extent on glucocorticoid receptor. In addition to a number of glucocorticoid receptor isoforms determined, it is found that the actions of glucocorticoids through GRα are influenced by other isoforms, such as GRß and GRγ. Recently, studies found GR confers resistance to androgen deprivation therapy, and various glucocorticoids exert distinct efficacy in CRPC. In this review, we summarized the mechanisms of glucocorticoids and its clinical appliances on the basis of present evidence.


Asunto(s)
Progresión de la Enfermedad , Glucocorticoides/farmacología , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores de Glucocorticoides/metabolismo , Antagonistas de Andrógenos/uso terapéutico , Expresión Génica/efectos de los fármacos , Glucocorticoides/uso terapéutico , Humanos , Masculino , Neoplasias de la Próstata Resistentes a la Castración/química , Neoplasias de la Próstata Resistentes a la Castración/patología , Isoformas de Proteínas/metabolismo , Receptores de Glucocorticoides/análisis , Receptores de Glucocorticoides/genética
17.
Oncotarget ; 7(50): 83735-83743, 2016 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-27863438

RESUMEN

We aimed to evaluate ERG and SOX9 as potential biomarkers of docetaxel response in metastatic castration-resistant prostate cancer (mCRPC) patients. Seventy-one mCRPC patients were evaluated. Tissue microarrays were constructed and immunohistochemistry was performed. Treatment response was assessed by prostate specific antigen (PSA) response rate, PSA progression-free survival (PSA-PFS), clinical/radiologic PFS (C/R-PFS) and overall survival (OS). ERG and SOX9 were found in 13 (18.3%) and 62 (87.3%) patients, respectively. ERG-positive had lower PSA response rates than negative (15.4% vs 62.1%, p = 0.004), and SOX9 showed a same trend (46.8% vs 100.0%, p = 0.003). ERG positivity correlated with a lower PSA-PFS (3.2 mos vs 7.4 mos, p < 0.001), C/R-PFS (3.8 mos vs 9.0 mos, p < 0.001) and OS (10.8 mos vs 21.4 mos, p < 0.001). SOX9 positivity also showed a lower PSA-PFS, C/R-PFS and OS (p =0.006, p =0.012 and p =0.023, respectively). On multivariate analysis, ERG positivity was a significant risk factor for a lower PSA-PFS, C/R-PFS and OS (p < 0.001, p < 0.001 and p =0.001, respectively). SOX9 expression was also a risk factor for a lower PSA-PFS, C/R-PFS and OS (p = 0.018, p = 0.025 and p =0.047, respectively). These findings indicate that ERG and SOX9 is potential biomarkers for prediction to docetaxel treatment in mCRPC patients.


Asunto(s)
Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/análisis , Inmunohistoquímica , Neoplasias de la Próstata Resistentes a la Castración/química , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Factor de Transcripción SOX9/análisis , Taxoides/uso terapéutico , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Biopsia , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Docetaxel , Humanos , Calicreínas/sangre , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Valor Predictivo de las Pruebas , Modelos de Riesgos Proporcionales , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata Resistentes a la Castración/mortalidad , Neoplasias de la Próstata Resistentes a la Castración/patología , Estudios Retrospectivos , Factores de Riesgo , Taxoides/efectos adversos , Factores de Tiempo , Análisis de Matrices Tisulares , Regulador Transcripcional ERG/análisis , Resultado del Tratamiento
18.
Hum Pathol ; 49: 77-82, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26826413

RESUMEN

In vitro studies in prostate cancer (PCa) cell lines have suggested a key and complex role of the store-operated channels (SOCs) in major cancer hallmarks, including proliferation, apoptosis, and migration. In the present study, we investigated in vivo the expression of the SOC components transient receptor potential canonical (TRPC) 1, TRPC4, Orai1, and stromal interaction molecule 1 (STIM1), during all stages of PCa progression, and evaluated their prognostic impact in clinically localized cancer (CLC). The expressions of TRPC1, TRPC4, Orai1, STIM1, and the androgen receptor and the proliferation marker Ki-67 were evaluated by immunohistochemistry on tissue microarrays containing samples of normal prostate tissues (n=91), prostatic intraepithelial neoplasia (n=61), CLC surgically treated (n=238), and castration-resistant prostate cancer (CRPC; n=45). All markers significantly increased in CLC compared with normal tissues and (for Orai1 and STIM1) in advanced pT3 tumors compared with pT2. In contrast, their expression decreased in CRPC, particularly for Orai1. In CLC, staining for TRPC1, Orai1 and STIM1 correlated with androgen receptor expression, and TRPC1 status was associated with lower proliferation and longer recurrence-free survival, after adjusting for classical prognostic markers. Although increased SOC expression during PCa progression supports a role in cancer cell migration, the inverse association between TRPC1 and biochemical relapse suggests a protective effect in CLC. Moreover, the dramatic down-regulation of Orai1 in CRPC supports its role in apoptosis at this stage of the disease. These results call for caution when considering SOCs as potential therapeutic targets for PCa.


Asunto(s)
Biomarcadores de Tumor/análisis , Canales de Calcio/análisis , Proteínas de la Membrana/análisis , Proteínas de Neoplasias/análisis , Neoplasia Intraepitelial Prostática/química , Neoplasias de la Próstata Resistentes a la Castración/química , Neoplasias de la Próstata/química , Canales Catiónicos TRPC/análisis , Anciano , Estudios de Casos y Controles , Supervivencia sin Enfermedad , Humanos , Inmunohistoquímica , Antígeno Ki-67/análisis , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Recurrencia Local de Neoplasia , Estadificación de Neoplasias , Proteína ORAI1 , Prostatectomía , Neoplasia Intraepitelial Prostática/mortalidad , Neoplasia Intraepitelial Prostática/patología , Neoplasia Intraepitelial Prostática/cirugía , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/cirugía , Neoplasias de la Próstata Resistentes a la Castración/mortalidad , Neoplasias de la Próstata Resistentes a la Castración/patología , Neoplasias de la Próstata Resistentes a la Castración/cirugía , Factores Protectores , Receptores Androgénicos/análisis , Factores de Riesgo , Molécula de Interacción Estromal 1 , Factores de Tiempo , Análisis de Matrices Tisulares , Resultado del Tratamiento
19.
Bull Cancer ; 102(6): 497-500, 2015 Jun.
Artículo en Francés | MEDLINE | ID: mdl-26028493

RESUMEN

Castration-resistant prostate cancer was subjected to a paradigm switch from hormone resistance to androgen deprivation therapy resistance during the last decade. Indeed, new therapeutics targeting the androgen receptor showed clinical efficacy in patients with progressive disease under castration. Thus, it is a proof that the AR remains a dominant driver of oncogenesis in earlier-called hormone resistant prostate cancer. This review summarizes the molecular mechanisms involved in castration-resistant prostate cancer.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/fisiología , Testosterona/metabolismo , Dihidrotestosterona/metabolismo , Epigénesis Genética , Humanos , Masculino , Mutación , Próstata/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/química , Isoformas de Proteínas/metabolismo , Receptores Androgénicos/química , Receptores Androgénicos/genética
20.
Urology ; 86(6): 1206-11, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26375845

RESUMEN

OBJECTIVE: To conduct a phase II trial of the combination of carboplatin, prednisone, and everolimus in metastatic castrate-resistant prostate cancer (mCRPC) as mTOR inhibition can overcome resistance to chemotherapy in prostate cancer. METHODS: Patients with progressive mCRPC pretreated with docetaxel-based regimen were eligible. Performance status of 0-1 and adequate bone marrow, renal, and liver function were required. Primary end point was time to progression. Treatment consisted of carboplatin (starting dose equal to area under the curve (AUC of 5) intravenously every 21 days along with oral everolimus 5 mg once daily and prednisone 5 mg twice daily. RESULTS: Twenty-six patients were enrolled with median age of 69 years with 8 patients of African American origin. Grade 3 or 4 thrombocytopenia or neutropenia in 4 of 6 initial patients required dose adjustment of carboplatin to AUC of 4 for subsequent patients. There were no pharmacokinetic interactions between carboplatin and everolimus. The median time to progression was 2.5 months (90% confidence interval [CI], 1.8-4.3 months), and median overall survival was 12.5 months (90% CI, 7.7-18.7 months). Of 10 patients, 8 that demonstrated positive nuclear phosphorylated AKT (pAKT) staining on immunohistochemistry progressed within 9 weeks, whereas 2 patients with negative staining continued without progression for prolonged durations of 30 and 48 weeks. TSC1 gene mutations did not correlate with clinical outcome. CONCLUSION: The addition of the mTOR inhibitor everolimus to carboplatin demonstrated minimal clinical efficacy in metastatic prostate cancer. pAKT testing warrants further evaluation as a predictive marker of response to everolimus therapy.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/secundario , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/patología , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carboplatino/administración & dosificación , Carboplatino/farmacocinética , Progresión de la Enfermedad , Docetaxel , Everolimus/administración & dosificación , Everolimus/farmacocinética , Humanos , Masculino , Células Neoplásicas Circulantes , Fosforilación , Prednisona/administración & dosificación , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata Resistentes a la Castración/química , Proteínas Proto-Oncogénicas c-akt/análisis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tasa de Supervivencia , Serina-Treonina Quinasas TOR/análisis , Taxoides/administración & dosificación , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA