Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Mol Cancer Ther ; 23(7): 973-994, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38507737

RESUMEN

Metastatic castration-resistant prostate cancer remains incurable regardless of recent therapeutic advances. Prostate cancer tumors display highly glycolytic phenotypes as the cancer progresses. Nonspecific inhibitors of glycolysis have not been utilized successfully for chemotherapy, because of their penchant to cause systemic toxicity. This study reports the preclinical activity, safety, and pharmacokinetics of a novel small-molecule preclinical candidate, BKIDC-1553, with antiglycolytic activity. We tested a large battery of prostate cancer cell lines for inhibition of cell proliferation, in vitro. Cell-cycle, metabolic, and enzymatic assays were used to demonstrate their mechanism of action. A human patient-derived xenograft model implanted in mice and a human organoid were studied for sensitivity to our BKIDC preclinical candidate. A battery of pharmacokinetic experiments, absorption, distribution, metabolism, and excretion experiments, and in vitro and in vivo toxicology experiments were carried out to assess readiness for clinical trials. We demonstrate a new class of small-molecule inhibitors where antiglycolytic activity in prostate cancer cell lines is mediated through inhibition of hexokinase 2. These compounds display selective growth inhibition across multiple prostate cancer models. We describe a lead BKIDC-1553 that demonstrates promising activity in a preclinical xenograft model of advanced prostate cancer, equivalent to that of enzalutamide. BKIDC-1553 demonstrates safety and pharmacologic properties consistent with a compound that can be taken into human studies with expectations of a good safety margin and predicted dosing for efficacy. This work supports testing BKIDC-1553 and its derivatives in clinical trials for patients with advanced prostate cancer.


Asunto(s)
Proliferación Celular , Glucólisis , Ensayos Antitumor por Modelo de Xenoinjerto , Masculino , Humanos , Animales , Ratones , Glucólisis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
2.
bioRxiv ; 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-37461469

RESUMEN

Purpose: Metastatic castration-resistant prostate cancer remains incurable regardless of recent therapeutic advances. Prostate cancer tumors display highly glycolytic phenotypes as the cancer progresses. Non-specific inhibitors of glycolysis have not been utilized successfully for chemotherapy, because of their penchant to cause systemic toxicity. This study reports the preclinical activity, safety, and pharmacokinetics of a novel small molecule preclinical candidate, BKIDC-1553, with antiglycolytic activity. Experimental design: We tested a large battery of prostate cancer cell lines for inhibition of cell proliferation, in vitro. Cell cycle, metabolic and enzymatic assays were used to demonstrate their mechanism of action. A human PDX model implanted in mice and a human organoid were studied for sensitivity to our BKIDC preclinical candidate. A battery of pharmacokinetic experiments, absorption, distribution, metabolism, and excretion experiments, and in vitro and in vivo toxicology experiments were carried out to assess readiness for clinical trials. Results: We demonstrate a new class of small molecule inhibitors where antiglycolytic activity in prostate cancer cell lines is mediated through inhibition of hexokinase 2. These compounds display selective growth inhibition across multiple prostate cancer models. We describe a lead BKIDC-1553 that demonstrates promising activity in a preclinical xenograft model of advanced prostate cancer, equivalent to that of enzalutamide. BKIDC-1553 demonstrates safety and pharmacologic properties consistent with a compound that can be taken into human studies with expectations of a good safety margin and predicted dosing for efficacy. Conclusion: This work supports testing BKIDC-1553 and its derivatives in clinical trials for patients with advanced prostate cancer.

3.
Front Oncol ; 10: 580617, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33163409

RESUMEN

Metabolic reprogramming is associated with re/activation and antagonism of androgen receptor (AR) signaling that drives prostate cancer (PCa) progression to castration resistance, respectively. In particular, AR signaling influences the fates of citrate that uniquely characterizes normal and malignant prostatic metabolism (i.e., mitochondrial export and extracellular secretion in normal prostate, mitochondrial retention and oxidation to support oxidative phenotype of primary PCa, and extra-mitochondrial interconversion into acetyl-CoA for fatty acid synthesis and epigenetics in the advanced PCa). The emergence of castration-resistant PCa (CRPC) involves reactivation of AR signaling, which is then further targeted by androgen synthesis inhibitors (abiraterone) and AR-ligand inhibitors (enzalutamide, apalutamide, and daroglutamide). However, based on AR dependency, two distinct metabolic and cellular adaptations contribute to development of resistance to these agents and progression to aggressive and lethal disease, with the tumor ultimately becoming highly glycolytic and with imaging by a tracer of tumor energetics, 18F-fluorodoxyglucose (18F-FDG). Another major resistance mechanism involves a lineage alteration into AR-indifferent carcinoma such a neuroendocrine which is diagnostically characterized by robust 18F-FDG uptake and loss of AR signaling. PCa is also characterized by metabolic alterations such as fatty acid and polyamine metabolism depending on AR signaling. In some cases, AR targeting induces rather than suppresses these alterations in cellular metabolism and energetics, which can be explored as therapeutic targets in lethal CRPC.

4.
Mol Cancer Ther ; 19(4): 1059-1069, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32054790

RESUMEN

Androgen deprivation therapy and second-generation androgen receptor signaling inhibitors such as enzalutamide are standard treatments for advanced/metastatic prostate cancer. Unfortunately, most men develop resistance and relapse; signaling via insulin-like growth factor (IGF) has been implicated in castration-resistant prostate cancer. We evaluated the antitumor activity of xentuzumab (IGF ligand-neutralizing antibody), alone and in combination with enzalutamide, in prostate cancer cell lines (VCaP, DuCaP, MDA PCa 2b, LNCaP, and PC-3) using established in vitro assays, and in vivo, using LuCaP 96CR, a prostate cancer patient-derived xenograft (PDX) model. Xentuzumab + enzalutamide reduced the viability of phosphatase and tensin homolog (PTEN)-expressing VCaP, DuCaP, and MDA PCa 2b cells more than either single agent, and increased antiproliferative activity and apoptosis induction in VCaP. Xentuzumab or xentuzumab + enzalutamide inhibited IGF type 1 receptor and AKT serine/threonine kinase (AKT) phosphorylation in VCaP, DuCaP, and MDA PCa 2b cells; xentuzumab had no effect on AKT phosphorylation and proliferation in PTEN-null LNCaP or PC-3 cells. Knockdown of PTEN led to loss of antiproliferative activity of xentuzumab and reduced activity of xentuzumab + enzalutamide in VCaP cells. Xentuzumab + enzalutamide inhibited the growth of castration-resistant LuCaP 96CR PDX with acquired resistance to enzalutamide, and improved survival in vivo The data suggest that xentuzumab + enzalutamide combination therapy may overcome castration resistance and could be effective in patients who are resistant to enzalutamide alone. PTEN status as a biomarker of responsiveness to combination therapy needs further investigation.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Neutralizantes/farmacología , Factor II del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Feniltiohidantoína/análogos & derivados , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Animales , Apoptosis , Benzamidas , Ciclo Celular , Proliferación Celular , Quimioterapia Combinada , Humanos , Masculino , Ratones , Ratones SCID , Nitrilos , Feniltiohidantoína/farmacología , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Expert Opin Ther Targets ; 22(3): 201-216, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29417861

RESUMEN

INTRODUCTION: The androgen receptor variant AR-V7 is gaining attention as a potential predictive marker for as well as one of the resistance mechanisms to the most current anti-androgen receptor (AR) therapies in castration-resistant prostate cancer (CRPC). Accordingly, development of next-generation drugs that directly or indirectly target AR-V7 signaling is urgently needed. Areas covered: We review proposed mechanisms of drug resistance in relation to AR-V7 status, the mechanisms of generation of AR-V7, and its transcriptome, cistrome, and interactome. Pharmacological agents that interfere with these processes are being developed to counteract pan AR and AR-V7-specific signaling. Also, we address the current status of the preclinical and clinical studies targeting AR-V7 signaling. Expert opinion: AR-V7 is considered a true therapeutic target, however, it remains to be determined if AR-V7 is a principal driver or merely a bystander requiring heterodimerization with co-expressed full-length AR or other variants to drive CRPC progression. While untangling AR-V7 biology, multiple strategies are being developed to counteract drug resistance, including selective blockade of AR-V7 signaling as well as inhibition of pan-AR signaling. Ideally anti-AR therapies will be combined with agents preventing activation and enrichment of AR negative tumor cells that are otherwise depressed by AR activity axis.


Asunto(s)
Antagonistas de Andrógenos/farmacología , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Receptores Androgénicos/efectos de los fármacos , Biomarcadores de Tumor/metabolismo , Diseño de Fármacos , Resistencia a Antineoplásicos , Humanos , Masculino , Terapia Molecular Dirigida , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/metabolismo , Transducción de Señal/efectos de los fármacos
6.
Cancer Res ; 78(8): 2065-2080, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29358171

RESUMEN

Primary prostate cancer is generally treatable by androgen deprivation therapy, however, later recurrences of castrate-resistant prostate cancer (CRPC) that are more difficult to treat nearly always occur due to aberrant reactivation of the androgen receptor (AR). In this study, we report that CRPC cells are particularly sensitive to the growth-inhibitory effects of reengineered tricyclic sulfonamides, a class of molecules that activate the protein phosphatase PP2A, which inhibits multiple oncogenic signaling pathways. Treatment of CRPC cells with small-molecule activators of PP2A (SMAP) in vitro decreased cellular viability and clonogenicity and induced apoptosis. SMAP treatment also induced an array of significant changes in the phosphoproteome, including most notably dephosphorylation of full-length and truncated isoforms of the AR and downregulation of its regulatory kinases in a dose-dependent and time-dependent manner. In murine xenograft models of human CRPC, the potent compound SMAP-2 exhibited efficacy comparable with enzalutamide in inhibiting tumor formation. Overall, our results provide a preclinical proof of concept for the efficacy of SMAP in AR degradation and CRPC treatment.Significance: A novel class of small-molecule activators of the tumor suppressor PP2A, a serine/threonine phosphatase that inhibits many oncogenic signaling pathways, is shown to deregulate the phosphoproteome and to destabilize the androgen receptor in advanced prostate cancer. Cancer Res; 78(8); 2065-80. ©2018 AACR.


Asunto(s)
Activadores de Enzimas/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/enzimología , Proteína Fosfatasa 2C/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Animales , Línea Celular Tumoral , Activadores de Enzimas/farmacología , Xenoinjertos , Humanos , Masculino , Ratones , Ratones SCID , Fosfoproteínas/metabolismo , Proteína Fosfatasa 2C/metabolismo , Proteómica , ARN Mensajero/genética , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología
7.
Endocr Relat Cancer ; 24(9): R335-R348, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28808043

RESUMEN

Organisms have evolved to generate biological complexity in their proteome and transcriptome from a limited number of genes. This concept holds true for the androgen receptor, which displays a diversity of inclusion/exclusion events in its structural motifs as a mechanism of resistance to the most forefront anti-androgen therapies. More than 20 androgen receptor variants that lack various portions of ligand-binding domain have been identified in human prostate cancer (PCa) samples. Most of the variants are inactive on their own, with a few exceptions displaying constitutive activity. The full-length receptor and one or more variants can be co-expressed in the same cell under many circumstances, which raises the question of how these variants physically and functionally interact with the full-length receptor or one another in the course of PCa progression. To address this issue, in this review, we will characterize and discuss androgen receptor variants, including the novel variants discovered in the last couple of years (i) individually, (ii) with respect to their physical and functional interaction with one another and (iii) in clinical relevance. Here, we also introduce the very recent understanding of AR-Vs obtained through successful development of some AR-V-specific antibodies as well as identification of novel AR-Vs by data mining approaches.


Asunto(s)
Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Animales , Humanos , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Multimerización de Proteína , Receptores Androgénicos/química , Receptores Androgénicos/genética , Transcriptoma
8.
Biomaterials ; 35(28): 8164-74, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24942815

RESUMEN

Synthetic biomaterial scaffolds show promise for in vitro and in vivo 3D cancer models. Tumors engineered in biomaterial scaffolds have shown evidence of being more physiologically relevant than some traditional preclinical model systems, and synthetic biomaterials provide the added benefit of defined and consistent microenvironmental control. Here, we examine sphere-templated poly(2-hydroxyethyl methacrylate) (pHEMA) scaffolds as the basis for engineering xenografts from multiple human prostate cancer cell lines. pHEMA scaffolds seeded and pre-cultured with tumorigenic M12 cells prior to implantation generated tumors in athymic nude mice, demonstrating the ability of the scaffolds to be used as a synthetic vehicle for xenograft generation. pHEMA scaffolds seeded with LNCaP C4-2 cells, which require Matrigel or stromal cell support for tumor formation, were poorly tumorigenic up to 12 weeks after implantation even when Matrigel was infused into the scaffold, demonstrating a lack of necessary pro-tumorigenic signaling within the scaffolds. Finally, M12mac25 cells, which are ordinarily rendered non-tumorigenic through the expression of the tumor suppressor insulin-like growth factor binding protein 7 (IGFBP7), displayed a tumorigenic response when implanted within porous pHEMA scaffolds. These M12mac25 tumors showed significant macrophage infiltration within the scaffolds driven by the foreign body response to the materials. These findings show the potential for this biomaterials-based model system to be used in the study of prostate cancer tumorigenesis and dormancy escape.


Asunto(s)
Materiales Biocompatibles/química , Polihidroxietil Metacrilato/química , Neoplasias de la Próstata/terapia , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Colágeno/química , Combinación de Medicamentos , Humanos , Imagenología Tridimensional , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Laminina/química , Masculino , Ratones , Ratones Desnudos , Microscopía Electrónica de Rastreo , Trasplante de Neoplasias , Proteoglicanos/química , Propiedades de Superficie
9.
Horm Cancer ; 5(4): 207-17, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24798453

RESUMEN

Resistance to the latest advanced prostate cancer therapies, including abiraterone and enzalutamide, is associated with increased expression of constitutively active androgen receptor splice variants (AR-Vs). The exact mechanism by which these therapies result in AR-Vs is unknown, but may include genomic rearrangement of the androgen receptor gene as well as alternative splicing of the AR pre-messenger RNA (mRNA). An additional complication that hinders further development of effective AR strategies is that the mechanisms by which the directed therapies are bypassed may vary. Finally, the question must be addressed as to whether the androgen receptor remains to be the driver of most castration resistant disease or whether truly AR-independent tumors arise after successful androgen ablation therapy. In this review, we will examine androgen receptor splice variants as an alternative mechanism by which prostate cancer becomes resistant to androgen receptor-directed therapy.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/metabolismo , Empalme Alternativo , Animales , Humanos , Masculino , Neoplasias de la Próstata Resistentes a la Castración/genética , Isoformas de Proteínas , Receptores Androgénicos/genética
10.
J Clin Endocrinol Metab ; 98(5): E820-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23533230

RESUMEN

CONTEXT: Prostate cancer patients at increased risk for relapse after prostatectomy were treated in a neoadjuvant study with androgen deprivation therapy (ADT) in combination with cixutumumab, an inhibitory fully human monoclonal antibody against IGF receptor 1 (IGF-IR). OBJECTIVE: A clinical trial with prospective collection of serum and tissue was designed to test the potential clinical efficacy of neoadjuvant IGF-IR blockade combined with ADT in these patients. The effect of body mass index (BMI) on response of IGF-IR/insulin components to IGF-IR blockade was also examined. DESIGN: Eligibility for the trial required the presence of high-risk prostate adenocarcinoma. Treatment consisted of bicalutamide, goserelin, and cixutumumab for 13 weeks before prostatectomy. Here we report on an analysis of serum samples from 29 enrolled patients. Changes in IGF and glucose homeostasis pathways were compared to control samples from patients in a concurrent clinical trial of neoadjuvant ADT alone. RESULTS: Significant increases were seen in GH (P = .001), IGF-I (P < .0001), IGF-II (P = .003), IGF binding protein (IGFBP)-3 (P < .0001), C-peptide (P = .0038), and insulin (P = .05) compared to patients treated with ADT alone. IGFBP-1 levels were significantly lower in the cixutumumab plus ADT cohort (P = .001). No significant changes in blood glucose were evident. Patients with BMIs in the normal range had significantly higher GH (P < .05) and IGFBP-1 (P < 0.5) levels compared to overweight and obese patients. CONCLUSIONS: Patients with IGF-IR blockade in combination with ADT demonstrated significant changes in IGF and glucose homeostasis pathway factors compared to patients receiving ADT alone. In the patients receiving combination therapy, patients with normal BMI had serum levels of glucose homeostasis components similar to individuals in the ADT-alone cohort, whereas patients with overweight and obese BMIs had serum levels that differed from the ADT cohort.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antagonistas de Andrógenos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Terapia Neoadyuvante , Obesidad/complicaciones , Neoplasias de la Próstata/tratamiento farmacológico , Receptor IGF Tipo 1/antagonistas & inhibidores , Adenocarcinoma/complicaciones , Adenocarcinoma/prevención & control , Adenocarcinoma/cirugía , Antagonistas de Andrógenos/administración & dosificación , Anilidas/administración & dosificación , Anilidas/uso terapéutico , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Índice de Masa Corporal , Estudios de Cohortes , Goserelina/administración & dosificación , Goserelina/uso terapéutico , Humanos , Insulina/sangre , Resistencia a la Insulina , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Masculino , Nitrilos/administración & dosificación , Nitrilos/uso terapéutico , Prostatectomía , Neoplasias de la Próstata/complicaciones , Neoplasias de la Próstata/prevención & control , Neoplasias de la Próstata/cirugía , Receptor IGF Tipo 1/metabolismo , Riesgo , Prevención Secundaria , Somatomedinas/análisis , Compuestos de Tosilo/administración & dosificación , Compuestos de Tosilo/uso terapéutico , Estados Unidos/epidemiología
11.
Tissue Eng Part C Methods ; 19(9): 738-44, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23373788

RESUMEN

Biomaterial-based tissue-engineered tumor models are now widely used in cancer biology studies. However, specific methods for efficient and reliable cell seeding into these and tissue-engineering constructs used for regenerative medicine often remain poorly defined. Here, we describe a capillary force-based method for seeding the human prostate cancer cell lines M12 and LNCaP C4-2 into sphere-templated poly(2-hydroxyethyl methacrylate) hydrogels. The capillary force seeding method improved the cell number and distribution within the porous scaffolds compared to well-established protocols such as static and centrifugation seeding. Seeding efficiency was found to be strongly dependent on the rounded cell diameter relative to the pore diameter and pore interconnect size, parameters that can be controllably modulated during scaffold fabrication. Cell seeding efficiency was evaluated quantitatively using a PicoGreen DNA assay, which demonstrated some variation in cell retention using the capillary force method. When cultured within the porous hydrogels, both cell lines attached and proliferated within the network, but histology showed the formation of a necrotic zone by 7 days likely due to oxygen and nutrient diffusional limitations. The necrotic zone thickness was decreased by dynamically culturing cells in an orbital shaker. Proliferation analysis showed that despite a variable seeding efficiency, by 7 days in culture, scaffolds contained a roughly consistent number of cells as they proliferated to fill the pores of the scaffold. These studies demonstrate that sphere-templated polymeric scaffolds have the potential to serve as an adaptable cell culture substrate for engineering a three-dimensional prostate cancer model.


Asunto(s)
Hidrogeles/farmacología , Fenómenos Mecánicos , Neoplasias de la Próstata/patología , Ingeniería de Tejidos/métodos , Ensayos Antitumor por Modelo de Xenoinjerto , Línea Celular Tumoral , ADN de Neoplasias/metabolismo , Humanos , Masculino , Polihidroxietil Metacrilato/farmacología , Coloración y Etiquetado , Andamios del Tejido
12.
J Clin Invest ; 120(8): 2715-30, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20644256

RESUMEN

Progression of prostate cancer following castration is associated with increased androgen receptor (AR) expression and signaling despite AR blockade. Recent studies suggest that these activities are due to the generation of constitutively active AR splice variants, but the mechanisms by which these splice variants could mediate such effects are not fully understood. Here we have identified what we believe to be a novel human AR splice variant in which exons 5, 6, and 7 are deleted (ARv567es) and demonstrated that this variant can contribute to cancer progression in human prostate cancer xenograft models in mice following castration. We determined that, in human prostate cancer cell lines, ARv567es functioned as a constitutively active receptor, increased expression of full-length AR (ARfl), and enhanced the transcriptional activity of AR. In human xenografts, human prostate cancer cells transfected with ARv567es cDNA formed tumors that were resistant to castration. Furthermore, the ratio of ARv567es to ARfl expression within the xenografts positively correlated with resistance to castration. Importantly, we also detected ARv567es frequently in human prostate cancer metastases. In summary, these data indicate that constitutively active AR splice variants can contribute to the development of castration-resistant prostate cancers and may serve as biomarkers for patients who are likely to suffer from early recurrence and are candidates for therapies directly targeting the AR rather than ligand.


Asunto(s)
Orquiectomía , Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Antagonistas de Andrógenos/uso terapéutico , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Ácidos Hidroxámicos/farmacología , Masculino , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia , Empalme del ARN , Receptores Androgénicos/fisiología , Trasplante Heterólogo , Vorinostat
13.
Int J Cancer ; 127(12): 2739-48, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21351253

RESUMEN

Age is the greatest risk factor for the development of epithelial cancers. In this minireview, we will examine key extracellular matrix and matricellular components, their changes with aging, and discuss how these alterations might influence the subsequent progression of cancer in the aged host. Because of the tight correlation between advanced age and the prevalence of prostate cancer, we will use prostate cancer as the model throughout this minireview.


Asunto(s)
Envejecimiento/fisiología , Matriz Extracelular/metabolismo , Espacio Extracelular/fisiología , Neoplasias/fisiopatología , Microambiente Tumoral/fisiología , Progresión de la Enfermedad , Humanos , Neoplasias/metabolismo
14.
Clin Cancer Res ; 15(24): 7634-7641, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19996218

RESUMEN

PURPOSE: Inhibition of insulin-like growth factor (IGF) signaling using the human IGF-I receptor monoclonal antibody A12 is most effective at inducing apoptosis in prostate cancer xenografts in the presence of androgen. We undertook this study to determine mechanisms for increased apoptosis by A12 in the presence of androgens. Experimental Methods: The castrate-resistant human xenograft LuCaP 35 V was implanted into intact or castrate severe combined immunodeficient mice and treated with A12 weekly. After 6 weeks of tumor growth, animals were sacrificed and tumors were removed and analyzed for cell cycle distribution/apoptosis and cDNA arrays were done. RESULTS: In castrate mice, the tumors were delayed in G(2) with no apoptosis; in contrast, tumors from intact mice underwent apoptosis with either G(1) or G(2) delay. Transforming growth factor-beta-stimulated clone-22 (TSC-22) was significantly elevated in tumors from the intact mice compared with castrate mice, especially in those tumors with the highest levels of apoptosis. To further determine the function of TSC-22, we transfected various human prostate cancer cell lines with a plasmid expressing TSC-22. Cell lines overexpressing TSC-22 showed an increase in apoptosis and a delay in G(1). When these cell lines were placed subcutaneously in athymic nude mice, a decreased number of animals formed tumors and the rate of tumor growth was decreased compared with control tumors. CONCLUSIONS: These data indicate that IGF-I receptor inhibition in the presence of androgen has an enhanced effect on decreasing tumor growth, in part, through increased expression of the tumor suppressor gene TSC-22. (Clin Cancer Res 2009;15(24):7634-41).

15.
Neoplasia ; 10(12): 1350-61, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19048114

RESUMEN

Prostate cancer is an age-associated epithelial cancer, and as such, it contributes significantly to the mortality of the elderly. Senescence is one possible mechanism by which the body defends itself against various epithelial cancers. Senescent cells alter the microenvironment, in part, through changes to the extracellular matrix. Laminins (LMs) are extracellular proteins important to both the structure and function of the microenvironment. Overexpression of the senescence-associated gene mac25 in human prostate cancer cells resulted in increased mRNA levels of the LM alpha4 and beta2 chains compared to empty vector control cells. The purpose of this study was to examine the effects of these senescence-induced LM chains on tumorigenicity of prostate cancer cells. We created stable M12 human prostate cancer lines overexpressing either the LM alpha4 or beta2 chain or both chains. Increased expression of either the LM alpha4 or beta2 chain resulted in increased in vitro migration and in vivo tumorigenicity of those cells, whereas high expression of both chains led to decreased in vitro proliferation and in vivo tumorigenicity compared to M12 control cells. This study demonstrates that senescent prostate epithelial cells can alter the microenvironment and that these changes modulate progression of prostate cancer.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Laminina/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Senescencia Celular , Humanos , Laminina/biosíntesis , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cicatrización de Heridas
16.
Oncogene ; 23(26): 4584-93, 2004 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-15077158

RESUMEN

Overexpression of mac25 in the prostate cancer cell line M12 effects a dramatic reversal of the transformed phenotype. cDNA array analysis of RNA from cells overproducing the mac25 protein (M12/mac25) indicated upregulation of the sex determining transcription factor SOX9. In this study, we have confirmed increased expression of SOX9 in M12/mac25 cells and have further investigated the physiological effects of increased SOX9 production. Greatly increased levels of SOX9 RNA and mature protein were demonstrated in cells transfected with a SOX9 cDNA (M12/SOX9), and gel mobility shift assays confirmed binding of nuclear protein from these cells to an oligonucleotide containing the SOX9 consensus binding sequence. M12/SOX9 cells assumed the spindle-shaped morphology characteristic of M12/mac25 cells, suggesting that SOX9 mediates some effects of mac25. Elevated expression of SOX9 resulted in a decreased rate of cellular proliferation, cell cycle arrest in G0/G1, and increased sensitivity to apoptosis. Tumor development in athymic nude mice was inhibited by 80%. Finally, prostate-specific antigen and the androgen receptor, two genes whose expression is characteristic of differentiated cells, were both upregulated in M12/SOX9 cells. These data indicate that SOX9 contributes to growth regulation by mac25 via inhibition of cell growth and promotion of differentiation.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Factores de Transcripción/genética , Animales , Antígenos de Neoplasias , Apoptosis/genética , Biomarcadores , Pruebas de Carcinogenicidad , Ciclo Celular/genética , Diferenciación Celular/genética , División Celular/genética , Proteínas Ligadas a GPI , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Desnudos , Proteínas de Neoplasias/genética , Antígeno Prostático Específico/genética , Receptores Androgénicos/genética , Factor de Transcripción SOX9 , Factores de Transcripción/metabolismo
17.
Prostate ; 56(2): 115-22, 2003 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12746836

RESUMEN

BACKGROUND: In the prostate, insulin-like growth factor binding protein-related protein-1 (IGFBP-rP1/mac 25) appears to be decreased in cancer. Likewise, mice injected with prostate cells over-expressing IGFBP-rP1/mac 25 showed decreased tumor formation and tumor volume. METHODS: Immunohistochemistry was used to determine the presence of IGFBP-rP1/mac 25 in human prostate tissue and in LuCaP 23.12 prostate cancer xenografts. RESULTS: Immunoreactivity for IGFBP-rP1/mac 25 was detected in the cytoplasm and nuclei of benign prostate epithelium. Cancer cells also stained but the level of intensity was less than that observed in benign epithelium (P < 0.05). Within the stroma, peripheral nerves, and endothelial vessels reacted with IGFBP-rP1/mac 25 antibodies. IGFBP-rP1/mac 25 was also observed in the nuclei of LuCaP 23.12 cells. LuCaP xenografts with smaller tumor volumes demonstrated more staining, and larger tumor volumes had less staining (P = 0.0033, R = 0.610). There was also a significant inverse correlation between IGFBP-rP1/mac 25 levels in LuCaP 23.12 cells and the proliferative index, measured by percent of cells staining for 5-bromo-2'-deoxyuridine (Br-dU) (P = 0.0045, R = 0.594). CONCLUSIONS: IGFBP-rP1/mac 25 is present in normal prostate epithelium and is decreased in human prostate malignancy and higher IGFBP-rP1/mac 25 levels are associated with reduced tumor growth.


Asunto(s)
Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Animales , División Celular , Línea Celular , Células Epiteliales/metabolismo , Humanos , Inmunohistoquímica , Masculino , Ratones , Trasplante de Neoplasias , Próstata/citología , Próstata/metabolismo , Trasplante Heterólogo
18.
Oncogene ; 22(7): 1024-34, 2003 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-12592389

RESUMEN

Increased expression of mac25/insulin-like growth factor binding-protein related protein-1 (IGFBP-rP1) in human breast and prostate epithelial cell lines results in the suppression of tumor growth. CDNA expression array analysis revealed increased manganese superoxide dismutase (SOD-2) expression in the mac25/IGFBP-rP1-transfected M12 human prostate cancer cell line compared to M12 control cells. SOD-2 has been postulated to be a tumor suppressor. SOD-2 was also increased in LNCaP cells stably transfected with mac25/IGFBP-rP1, but not in mac25/IGFBP-rP1-transfected PC-3 cells. Mac25 LNCaP cells had a marked decrease in tumor growth in nude mice compared to controls, but there was no difference in tumor growth in mac25 PC-3 cells compared to control. Phosphorylated Erk and Akt were increased in the M12 and LNCaP transfected mac25/IGFBP-rP1 cells but not PC-3 mac25. Inhibition of PI-3 kinase results in a marked decrease in viability of the M12-mac25 cells compared to M12 controls. Cells treated with H(2)O(2) result in an increase in phospho-ERK. Transfection of SOD-2 in M12 cells markedly decreased tumor growth, apoptosis, G1 delay in the cell cycle, and expression of senescence associated beta-galactosidase. These results suggest that one of the downstream mediators of the senescence-associated tumor suppression effect of mac25/IGFBP-rP1 is SOD-2.


Asunto(s)
Adenocarcinoma/patología , Proteínas Portadoras/fisiología , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina , Proteínas de Neoplasias/fisiología , Neoplasias de la Próstata/patología , Transducción de Señal/fisiología , Superóxido Dismutasa/fisiología , Adenocarcinoma/enzimología , Animales , Apoptosis/fisiología , Línea Celular Transformada/enzimología , Línea Celular Transformada/trasplante , Senescencia Celular , Cromonas/farmacología , Inducción Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Fase G1/fisiología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Morfolinas/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Fosfatidilinositol 3-Quinasas/fisiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Regiones Promotoras Genéticas , Neoplasias de la Próstata/enzimología , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes de Fusión/fisiología , Superóxido Dismutasa/biosíntesis , Superóxido Dismutasa/genética , Transfección , Células Tumorales Cultivadas/enzimología , Células Tumorales Cultivadas/trasplante , beta-Galactosidasa/biosíntesis , beta-Galactosidasa/genética
19.
Prostate ; 51(2): 141-52, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11948969

RESUMEN

BACKGROUND: Insulin-like growth factor binding protein (IGFBP-3) levels are significantly reduced in malignant prostate epithelial cells. In this study, we evaluated the role of endogenous IGFBP-3 on prostate cancer cell growth and tumorigenesis. METHODS: IGFBP-3 was re-expressed by stable transfection of human IGFBP-3 cDNA in a model of human prostate cancer, M12, a malignant subline in which IGFBP-3 levels are undetectable in comparison to the parent epithelial cell, P69. Effect of IGFBP-3 re-expression (M12-BP-3) on growth kinetics, morphology, propensity to apoptosis, and in vivo tumor formation were studied. RESULTS: M12-BP-3 cells secreted IGFBP-3 and growth arrested at a cell density that was threefold lower than control cells and this was associated with marked alteration in cell morphology. Control cells when grown in conditioned media secreted by M12-BP-3 also showed altered morphology compared to when cultured in IGFBP-3-immunodepleted conditioned media. The M12-BP-3 clones showed altered mitochondrial membrane potential, increased PARP cleavage, increase in sub-G1 peak, decreased levels of neuron specific enolase, and decreased tumor formation in athymic, nude mice. CONCLUSIONS: These data suggest that IGFBP-3 induces early apoptosis and has potential tumor suppressive effect in prostate cancer. Prostate 51: 141-152, 2002.


Asunto(s)
Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Transformación Celular Neoplásica , Regulación Neoplásica de la Expresión Génica , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/biosíntesis , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/farmacología , Neoplasias de la Próstata/patología , Western Blotting , Medios de Cultivo , ADN Complementario/genética , ADN de Neoplasias/genética , Electroforesis en Gel de Poliacrilamida , Células Epiteliales/fisiología , Humanos , Inmunohistoquímica , Masculino , Células Tumorales Cultivadas
20.
Oncogene ; 21(1): 140-7, 2002 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-11791184

RESUMEN

In the present study, we examined the effects of over-expression of the potential tumor suppressor gene IGFBP-rP1/mac25 on cell-cycle kinetics in prostate cancer cells. The majority of the high expressing IGFBP-rP1/mac25 cell population was located in the G1 and sub-G0/G1 peaks; synchronizing cells in G2/M with nocodazole demonstrated the high expressing IGFBP-rP1/mac25 clones were delayed in the G1 phase of the cell cycle. Unscheduled expression of cyclin A in the sub-G0/G1 peak occurred in the IGFBP-rP1/mac25 clones. Immunoblots showed decreased cyclin D1 and p21 and increased cyclin E, p16, and p27 in the high expressing IGFBP-rP1/mac25 clones compared to the control cells. Cyclin D1/cdk-4,6 and cyclin E/cdk-2 kinase activities decreased but cyclin A/cdk-2 kinase activity increased for the high expressing IGFBP-rP1/mac25 clones compared to control cells. A pRb immunoprecipitation demonstrated more binding of E2F-1 to pRb in the high expressing IGFBP-rP1/mac25 clones than in control cells. Finally, cell senescence, as assessed by senescence-associated beta-galactosidase, demonstrated significantly more staining in the IGFBP-rP1/mac25 cells than control cells. These results suggest that IGFBP-rP1/mac25 alters the cell cycle kinetics of the M12 prostate cell line by delaying the cells in the G1 phase of the cell cycle. In addition, the appearance of cyclin A in the sub-G0/G1 phase of the cell cycle and the increased kinase activity of cyclin A/cdk-2 in the IGFBP-rP1/mac25 clones suggests that cyclin A is associated with the apoptotic cells.


Asunto(s)
Adenocarcinoma/patología , Apoptosis/genética , Proteínas Portadoras/fisiología , Ciclina A/fisiología , Proteínas de Unión al ADN , Fase G1/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina , Proteínas de Neoplasias/fisiología , Neoplasias de la Próstata/patología , Adenocarcinoma/genética , Western Blotting , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , División Celular , Senescencia Celular , Citoesqueleto/efectos de los fármacos , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Humanos , Sustancias Macromoleculares , Masculino , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Nocodazol/farmacología , Neoplasias de la Próstata/genética , Proteínas Recombinantes de Fusión/fisiología , Proteína de Retinoblastoma/metabolismo , Factores de Transcripción/metabolismo , Transfección , Células Tumorales Cultivadas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA