Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nucleic Acids Res ; 47(22): 11623-11636, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31647098

ABSTRACT

Alternative splicing is emerging as an oncogenic mechanism. In prostate cancer, generation of constitutively active forms of androgen receptor (AR) variants including AR-V7 plays an important role in progression of castration-resistant prostate cancer (CRPC). AR-V7 is generated by alternative splicing that results in inclusion of cryptic exon CE3 and translation of truncated AR protein that lacks the ligand binding domain. Whether AR-V7 can be a driver for CRPC remains controversial as the oncogenic mechanism of AR-V7 activation remains elusive. Here, we found that KDM4B promotes AR-V7 and identified a novel regulatory mechanism. KDM4B is phosphorylated by protein kinase A under conditions that promote castration-resistance, eliciting its binding to the splicing factor SF3B3. KDM4B binds RNA specifically near the 5'-CE3, upregulates the chromatin accessibility, and couples the spliceosome to the chromatin. Our data suggest that KDM4B can function as a signal responsive trans-acting splicing factor and scaffold that recruits and stabilizes the spliceosome near the alternative exon, thus promoting its inclusion. Genome-wide profiling of KDM4B-regulated genes also identified additional alternative splicing events implicated in tumorigenesis. Our study defines KDM4B-regulated alternative splicing as a pivotal mechanism for generating AR-V7 and a contributing factor for CRPC, providing insight for mechanistic targeting of CRPC.


Subject(s)
Alternative Splicing/genetics , Gene Expression Regulation, Neoplastic/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Androgen/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Chromatin/metabolism , HEK293 Cells , Humans , Male , Protein Isoforms/genetics , Receptors, Androgen/metabolism , Spliceosomes/genetics
2.
Am J Pathol ; 185(9): 2505-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26362718

ABSTRACT

Active Stat5a/b predicts early recurrence and disease-specific death in prostate cancer (PC), which both typically are caused by development of metastatic disease. Herein, we demonstrate that Stat5a/b induces epithelial-to-mesenchymal transition (EMT) of PC cells, as shown by Stat5a/b regulation of EMT marker expression (Twist1, E-cadherin, N-cadherin, vimentin, and fibronectin) in PC cell lines, xenograft tumors in vivo, and patient-derived PCs ex vivo using organ explant cultures. Jak2-Stat5a/b signaling induced functional end points of EMT as well, indicated by disruption of epithelial cell monolayers and increased migration and adhesion of PC cells to fibronectin. Knockdown of Twist1 suppressed Jak2-Stat5a/b-induced EMT properties of PC cells, which were rescued by re-introduction of Twist1, indicating that Twist1 mediates Stat5a/b-induced EMT in PC cells. While promoting EMT, Jak2-Stat5a/b signaling induced stem-like properties in PC cells, such as sphere formation and expression of cancer stem cell markers, including BMI1. Mechanistically, both Twist1 and BMI1 were critical for Stat5a/b induction of stem-like features, because genetic knockdown of Twist1 suppressed Stat5a/b-induced BMI1 expression and sphere formation in stem cell culture conditions, which were rescued by re-introduction of BMI1. By using human prolactin knock-in mice, we demonstrate that prolactin-Stat5a/b signaling promoted metastases formation of PC cells in vivo. In conclusion, our data support the concept that Jak2-Stat5a/b signaling promotes metastatic progression of PC by inducing EMT and stem cell properties in PC cells.


Subject(s)
Epithelial-Mesenchymal Transition , Janus Kinase 2/metabolism , Prostatic Neoplasms/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Animals , Cadherins/metabolism , Humans , Male , Mice , Neoplastic Stem Cells/pathology , Nuclear Proteins/metabolism , Prostatic Neoplasms/pathology , Recurrence , Signal Transduction/physiology , Twist-Related Protein 1/metabolism
3.
Am J Pathol ; 182(6): 2264-75, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23660011

ABSTRACT

The molecular mechanisms underlying progression of prostate cancer (PCa) to castrate-resistant (CR) and metastatic disease are poorly understood. Our previous mechanistic work shows that inhibition of transcription factor Stat5 by multiple alternative methods induces extensive rapid apoptotic death of Stat5-positive PCa cells in vitro and inhibits PCa xenograft tumor growth in nude mice. Furthermore, STAT5A/B induces invasive behavior of PCa cells in vitro and in vivo, suggesting involvement of STAT5A/B in PCa progression. Nuclear STAT5A/B protein levels are increased in high-grade PCas, CR PCas, and distant metastases, and high nuclear STAT5A/B expression predicts early disease recurrence and PCa-specific death in clinical PCas. Based on these findings, STAT5A/B represents a therapeutic target protein for advanced PCa. The mechanisms underlying increased Stat5 protein levels in PCa are unclear. Herein, we demonstrate amplification at the STAT5A/B gene locus in a significant fraction of clinical PCa specimens. STAT5A/B gene amplification was more frequently found in PCas of high histologic grades and in CR distant metastases. Quantitative in situ analysis revealed that STAT5A/B gene amplification was associated with increased STAT5A/B protein expression in PCa. Functional studies showed that increased STAT5A/B copy numbers conferred growth advantage in PCa cells in vitro and as xenograft tumors in vivo. The work presented herein provides the first evidence of somatic STAT5A/B gene amplification in clinical PCas.


Subject(s)
Gene Amplification , Prostatic Neoplasms/genetics , STAT5 Transcription Factor/genetics , Tumor Suppressor Proteins/genetics , Animals , DNA Copy Number Variations , DNA, Neoplasm/genetics , Disease Progression , Gene Expression Regulation, Neoplastic/physiology , Humans , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Nude , Neoplasm Grading , Neoplasm Transplantation , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Recurrence , STAT5 Transcription Factor/biosynthesis , Transplantation, Heterologous , Tumor Cells, Cultured , Tumor Suppressor Proteins/biosynthesis
4.
Mol Cancer Ther ; 19(1): 231-246, 2020 01.
Article in English | MEDLINE | ID: mdl-31548294

ABSTRACT

The second-generation antiandrogen, enzalutamide, is approved for castrate-resistant prostate cancer (CRPC) and targets androgen receptor (AR) activity in CRPC. Despite initial clinical activity, acquired resistance to enzalutamide arises rapidly and most patients develop terminal disease. Previous work has established Stat5 as a potent inducer of prostate cancer growth. Here, we investigated the significance of Jak2-Stat5 signaling in resistance of prostate cancer to enzalutamide. The levels of Jak2 and Stat5 mRNA, proteins and activation were evaluated in prostate cancer cells, xenograft tumors, and clinical prostate cancers before and after enzalutamide therapy. Jak2 and Stat5 were suppressed by genetic knockdown using lentiviral shRNA or pharmacologic inhibitors. Responsiveness of primary and enzalutamide-resistant prostate cancer to pharmacologic inhibitors of Jak2-Stat5 signaling was assessed in vivo in mice bearing prostate cancer xenograft tumors. Patient-derived prostate cancers were tested for responsiveness to Stat5 blockade as second-line treatment after enzalutamide ex vivo in tumor explant cultures. Enzalutamide-liganded AR induces sustained Jak2-Stat5 phosphorylation in prostate cancer leading to the formation of a positive feed-forward loop, where activated Stat5, in turn, induces Jak2 mRNA and protein levels contributing to further Jak2 activation. Mechanistically, enzalutamide-liganded AR induced Jak2 phosphorylation through a process involving Jak2-specific phosphatases. Stat5 promoted prostate cancer growth during enzalutamide treatment. Jak2-Stat5 inhibition induced death of prostate cancer cells and patient-derived prostate cancers surviving enzalutamide treatment and blocked enzalutamide-resistant tumor growth in mice. This work introduces a novel concept of a pivotal role of hyperactivated Jak2-Stat5 signaling in enzalutamide-resistant prostate cancer, which is readily targetable by Jak2 inhibitors in clinical development.


Subject(s)
Janus Kinase 2/antagonists & inhibitors , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms, Castration-Resistant/drug therapy , Animals , Benzamides , Humans , Male , Mice , Mice, Nude , Nitriles , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms, Castration-Resistant/pathology , Signal Transduction , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 24(8): 1917-1931, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29483142

ABSTRACT

Purpose: The standard treatment for organ-confined prostate cancer is surgery or radiation, and locally advanced prostate cancer is typically treated with radiotherapy alone or in combination with androgen deprivation therapy. Here, we investigated whether Stat5a/b participates in regulation of double-strand DNA break repair in prostate cancer, and whether Stat5 inhibition may provide a novel strategy to sensitize prostate cancer to radiotherapy.Experimental Design: Stat5a/b regulation of DNA repair in prostate cancer was evaluated by comet and clonogenic survival assays, followed by assays specific to homologous recombination (HR) DNA repair and nonhomologous end joining (NHEJ) DNA repair. For HR DNA repair, Stat5a/b regulation of Rad51 and the mechanisms underlying the regulation were investigated in prostate cancer cells, xenograft tumors, and patient-derived prostate cancers ex vivo in 3D explant cultures. Stat5a/b induction of Rad51 and HR DNA repair and responsiveness to radiation were evaluated in vivo in mice bearing prostate cancer xenograft tumors.Results: Stat5a/b is critical for Rad51 expression in prostate cancer via Jak2-dependent mechanisms by inducing Rad51 mRNA levels. Consistent with this, genetic knockdown of Stat5a/b suppressed HR DNA repair while not affecting NHEJ DNA repair. Pharmacologic Stat5a/b inhibition potently sensitized prostate cancer cell lines and prostate cancer tumors to radiation, while not inducing radiation sensitivity in the neighboring tissues.Conclusions: This work introduces a novel concept of a pivotal role of Jak2-Stat5a/b signaling for Rad51 expression and HR DNA repair in prostate cancer. Inhibition of Jak2-Stat5a/b signaling sensitizes prostate cancer to radiation and, therefore, may provide an adjuvant therapy for radiation to reduce radiation-induced damage to the neighboring tissues. Clin Cancer Res; 24(8); 1917-31. ©2018 AACR.


Subject(s)
DNA Repair , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Rad51 Recombinase/metabolism , Radiation Tolerance/genetics , STAT5 Transcription Factor/genetics , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Disease Models, Animal , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gene Expression , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Neoplasm Grading , Neoplasm Staging , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , RNA, Small Interfering/genetics , Rad51 Recombinase/genetics , Radiation Tolerance/drug effects , Radiation, Ionizing , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/metabolism , Xenograft Model Antitumor Assays
6.
Oncotarget ; 8(2): 3724-3745, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-27741508

ABSTRACT

Despite aggressive treatment for localized cancer, prostate cancer (PC) remains a leading cause of cancer-related death for American men due to a subset of patients progressing to lethal and incurable metastatic castrate-resistant prostate cancer (CRPC). Organ-confined PC is treated by surgery or radiation with or without androgen deprivation therapy (ADT), while options for locally advanced and disseminated PC include radiation combined with ADT, or systemic treatments including chemotherapy. Progression to CRPC results from failure of ADT, which targets the androgen receptor (AR) signaling axis and inhibits AR-driven proliferation and survival pathways. The exact mechanisms underlying the transition from androgen-dependent PC to CRPC remain incompletely understood. Reactivation of AR has been shown to occur in CRPC despite depletion of circulating androgens by ADT. At the same time, the presence of AR-negative cell populations in CRPC has also been identified. While AR signaling has been proposed as the primary driver of CRPC, AR-independent signaling pathways may represent additional mechanisms underlying CRPC progression. Identification of new therapeutic strategies to target both AR-positive and AR-negative PC cell populations and, thereby, AR-driven as well as non-AR-driven PC cell growth and survival mechanisms would provide a two-pronged approach to eliminate CRPC cells with potential for synthetic lethality. In this review, we provide an overview of AR-dependent and AR-independent molecular mechanisms which drive CRPC, with special emphasis on the role of the Jak2-Stat5a/b signaling pathway in promoting castrate-resistant growth of PC through both AR-dependent and AR-independent mechanisms.


Subject(s)
Prostatic Neoplasms/etiology , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Androgen Receptor Antagonists/pharmacology , Androgen Receptor Antagonists/therapeutic use , Animals , Cell Transformation, Neoplastic/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Humans , Janus Kinase 2/metabolism , Ligands , Male , Molecular Targeted Therapy , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/etiology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/genetics , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects
7.
Mol Cancer Ther ; 14(3): 713-26, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25552366

ABSTRACT

Although poorly understood, androgen receptor (AR) signaling is sustained despite treatment of prostate cancer with antiandrogens and potentially underlies development of incurable castrate-resistant prostate cancer. However, therapies targeting the AR signaling axis eventually fail when prostate cancer progresses to the castrate-resistant stage. Stat5a/b, a candidate therapeutic target protein in prostate cancer, synergizes with AR to reciprocally enhance the signaling of both proteins. In this work, we demonstrate that Stat5a/b sequesters antiandrogen-liganded (MDV3100, bicalutamide, flutamide) AR in prostate cancer cells and protects it against proteasomal degradation in prostate cancer. Active Stat5a/b increased nuclear levels of both unliganded and antiandrogen-liganded AR, as demonstrated in prostate cancer cell lines, xenograft tumors, and clinical patient-derived prostate cancer samples. Physical interaction between Stat5a/b and AR in prostate cancer cells was mediated by the DNA-binding domain of Stat5a/b and the N-terminal domain of AR. Moreover, active Stat5a/b increased AR occupancy of the prostate-specific antigen promoter and AR-regulated gene expression in prostate cancer cells. Mechanistically, both Stat5a/b genetic knockdown and antiandrogen treatment induced proteasomal degradation of AR in prostate cancer cells, with combined inhibition of Stat5a/b and AR leading to maximal loss of AR protein and prostate cancer cell viability. Our results indicate that therapeutic targeting of AR in prostate cancer using antiandrogens may be substantially improved by targeting of Stat5a/b.


Subject(s)
Androgen Antagonists/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptors, Androgen/metabolism , STAT5 Transcription Factor/antagonists & inhibitors , Tumor Suppressor Proteins/antagonists & inhibitors , Androgens/metabolism , Anilides/pharmacology , Benzamides , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Flutamide/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Ligands , Male , Nitriles/pharmacology , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Prostate-Specific Antigen/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Tosyl Compounds/pharmacology
8.
Mol Cancer Ther ; 14(8): 1777-93, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26026053

ABSTRACT

Bypassing tyrosine kinases responsible for Stat5a/b phosphorylation would be advantageous for therapy development for Stat5a/b-regulated cancers. Here, we sought to identify small molecule inhibitors of Stat5a/b for lead optimization and therapy development for prostate cancer and Bcr-Abl-driven leukemias. In silico screening of chemical structure databases combined with medicinal chemistry was used for identification of a panel of small molecule inhibitors to block SH2 domain-mediated docking of Stat5a/b to the receptor-kinase complex and subsequent phosphorylation and dimerization. We tested the efficacy of the lead compound IST5-002 in experimental models and patient samples of two known Stat5a/b-driven cancers, prostate cancer and chronic myeloid leukemia (CML). The lead compound inhibitor of Stat5-002 (IST5-002) prevented both Jak2 and Bcr-Abl-mediated phosphorylation and dimerization of Stat5a/b, and selectively inhibited transcriptional activity of Stat5a (IC50 = 1.5µmol/L) and Stat5b (IC50 = 3.5 µmol/L). IST5-002 suppressed nuclear translocation of Stat5a/b, binding to DNA and Stat5a/b target gene expression. IST5-002 induced extensive apoptosis of prostate cancer cells, impaired growth of prostate cancer xenograft tumors, and induced cell death in patient-derived prostate cancers when tested ex vivo in explant organ cultures. Importantly, IST5-002 induced robust apoptotic death not only of imatinib-sensitive but also of imatinib-resistant CML cell lines and primary CML cells from patients. IST5-002 provides a lead structure for further chemical modifications for clinical development for Stat5a/b-driven solid tumors and hematologic malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Prostatic Neoplasms/metabolism , Quantitative Structure-Activity Relationship , STAT5 Transcription Factor/chemistry , Tumor Suppressor Proteins/chemistry , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cluster Analysis , Databases, Factual , Disease Models, Animal , Drug Resistance, Neoplasm , Gene Expression , Gene Expression Profiling , Genes, Reporter , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Male , Mice , Models, Molecular , Molecular Conformation , Phosphorylation , Prostatic Neoplasms/drug therapy , Protein Multimerization , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Small Molecule Libraries , Tissue Culture Techniques , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
9.
Mol Cancer Ther ; 13(5): 1246-58, 2014 May.
Article in English | MEDLINE | ID: mdl-24577942

ABSTRACT

Metastatic prostate cancer is lethal and lacks effective strategies for prevention or treatment, requiring novel therapeutic approaches. Interleukin-6 (IL-6) is a cytokine that has been linked with prostate cancer pathogenesis by multiple studies. However, the direct functional roles of IL-6 in prostate cancer growth and progression have been unclear. In the present study, we show that IL-6 is produced in distant metastases of clinical prostate cancers. IL-6-activated signaling pathways in prostate cancer cells induced a robust 7-fold increase in metastases formation in nude mice. We further show that IL-6 promoted migratory prostate cancer cell phenotype, including increased prostate cancer cell migration, microtubule reorganization, and heterotypic adhesion of prostate cancer cells to endothelial cells. IL-6-driven metastasis was predominantly mediated by Stat3 and to lesser extent by ERK1/2. Most importantly, pharmacologic inhibition of Jak1/2 by AZD1480 suppressed IL-6-induced signaling, migratory prostate cancer cell phenotypes, and metastatic dissemination of prostate cancer in vivo in nude mice. In conclusion, we demonstrate that the cytokine IL-6 directly promotes prostate cancer metastasis in vitro and in vivo via Jak-Stat3 signaling pathway, and that IL-6-driven metastasis can be effectively suppressed by pharmacologic targeting of Jak1/2 using Jak1/2 inhibitor AZD1480. Our results therefore provide a strong rationale for further development of Jak1/2 inhibitors as therapy for metastatic prostate cancer.


Subject(s)
Interleukin-6/metabolism , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Disease Models, Animal , Gene Expression , Humans , Interleukin-6/genetics , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Nude , Neoplasm Metastasis , Phenotype , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , STAT3 Transcription Factor/metabolism
10.
Clin Cancer Res ; 19(20): 5658-74, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23942095

ABSTRACT

PURPOSE: Progression of prostate cancer to the lethal castrate-resistant stage coincides with loss of responsiveness to androgen deprivation and requires development of novel therapies. We previously provided proof-of-concept that Stat5a/b is a therapeutic target protein for prostate cancer. Here, we show that pharmacologic targeting of Jak2-dependent Stat5a/b signaling by the Jak2 inhibitor AZD1480 blocks castrate-resistant growth of prostate cancer. EXPERIMENTAL DESIGN: Efficacy of AZD1480 in disrupting Jak2-Stat5a/b signaling and decreasing prostate cancer cell viability was evaluated in prostate cancer cells. A unique prostate cancer xenograft mouse model (CWR22Pc), which mimics prostate cancer clinical progression in patients, was used to assess in vivo responsiveness of primary and castrate-resistant prostate cancer (CRPC) to AZD1480. Patient-derived clinical prostate cancers, grown ex vivo in organ explant cultures, were tested for responsiveness to AZD1480. RESULTS: AZD1480 robustly inhibited Stat5a/b phosphorylation, dimerization, nuclear translocation, DNA binding, and transcriptional activity in prostate cancer cells. AZD1480 reduced prostate cancer cell viability sustained by Jak2-Stat5a/b signaling through induction of apoptosis, which was rescued by constitutively active Stat5a/b. In mice, pharmacologic targeting of Stat5a/b by AZD1480 potently blocked growth of primary androgen-dependent as well as recurrent castrate-resistant CWR22Pc xenograft tumors, and prolonged survival of tumor-bearing mice versus vehicle or docetaxel-treated mice. Finally, nine of 12 clinical prostate cancers responded to AZD1480 by extensive apoptotic epithelial cell loss, concurrent with reduced levels of nuclear Stat5a/b. CONCLUSIONS: We report the first evidence for efficacy of pharmacologic targeting of Stat5a/b as a strategy to inhibit castrate-resistant growth of prostate cancer, supporting further clinical development of Stat5a/b inhibitors as therapy for advanced prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Janus Kinase 2/antagonists & inhibitors , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , STAT5 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , Aged , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Humans , Janus Kinase 2/metabolism , Male , Mice , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Orchiectomy , Phosphorylation/drug effects , Prostatic Neoplasms/therapy , Protein Binding/drug effects , Protein Multimerization/drug effects , Protein Transport/drug effects , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Receptors, Androgen/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/chemistry , STAT5 Transcription Factor/metabolism , Transcriptional Activation , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Endocr Relat Cancer ; 19(1): 1-12, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21903859

ABSTRACT

Ionizing radiation (IR) is used frequently in the management of multiple tumor types, including both organ-confined and locally advanced prostate cancer (PCa). Enhancing tumor radiosensitivity could both reduce the amount of radiation required for definitive treatment and improve clinical outcome. Androgen suppression therapy improves clinical outcomes when combined with radiation therapy but is associated with significant acute and chronic toxicities; hence, there is a clear need for alternative means to increase the therapeutic window of radiotherapy. Herein, it is demonstrated that the mammalian target of rapamycin (mTOR) inhibitors rapamycin (sirolimus) and temsirolimus limit both hormone therapy (HT)-sensitive and castration-resistant PCa (CRPC) cell proliferation as single agents and have a profound radiosensitization effect when used in combination with IR. Importantly, the observed radiosensitization was influenced by the treatment schedule, in which adjuvant administration of mTOR inhibitors was most effective in limiting PCa cell population doubling. This schedule-dependent influence on in vitro treatment outcome was determined to be the result of relative effects on the cell cycle kinetics. Finally, adjuvant administration of either mTOR inhibitor tested after IR significantly decreased clonogenic cell survival of both HT-sensitive and CRPC cells compared with IR alone. Taken together, these data demonstrate that inhibition of mTOR confers a radiosensitization phenotype that is dependent on relative cell cycle kinetics and provide a foundation for clinical assessment.


Subject(s)
Prostatic Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Cell Cycle , Cell Line, Tumor , Cell Survival , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , X-Rays
12.
Nat Rev Urol ; 8(11): 597-607, 2011 Oct 04.
Article in English | MEDLINE | ID: mdl-21971318

ABSTRACT

Prolactin is best known for its actions on the mammary gland. However, circulating prolactin is also detected in males and its receptor (PRLR) is expressed in the prostate, suggesting that the prostate is a target of prolactin. Germline knockout of prolactin or its receptor has failed to reveal a key role for prolactin signaling in mouse prostate physiology. However, several studies involving rodent models and human prostate cell lines and specimens have supported the contribution of the canonical PRLR-Jak2-Stat5a/b pathway to prostate cancer tumorigenesis and progression. Increased expression of prolactin in the prostate itself (rather than changes in circulating prolactin levels) and crosstalk with androgen receptor (AR) signaling are potential mechanisms for increased Stat5a/b signaling in prostate cancer. In the mouse prostate, prolactin overexpression results in disorganized expansion of the basal/stem cell compartment, which has been proposed to house putative prostate tumor-initiating cells. These findings provide new insight into the molecular and cellular targets by which locally produced prolactin could contribute to prostate cancer initiation and progression. A number of pharmacological inhibitors targeting various levels of the PRLR-Jak2-Stat5a/b pathway have been developed and are entering clinical trials for advanced prostate cancer.


Subject(s)
Prolactin/physiology , Prostate/physiology , Prostatic Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Female , Gene Targeting , Humans , Male , Prolactin/biosynthesis , Prolactin/deficiency , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/blood , Prostatic Neoplasms/drug therapy , Receptors, Prolactin/biosynthesis , Receptors, Prolactin/deficiency , Receptors, Prolactin/physiology , Signal Transduction/drug effects , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL