Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 51(1): 99-116, 2023 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-36535377

RESUMO

Numerous cancers, including prostate cancer (PCa), are addicted to transcription programs driven by specific genomic regions known as super-enhancers (SEs). The robust transcription of genes at such SEs is enabled by the formation of phase-separated condensates by transcription factors and coactivators with intrinsically disordered regions. The androgen receptor (AR), the main oncogenic driver in PCa, contains large disordered regions and is co-recruited with the transcriptional coactivator mediator complex subunit 1 (MED1) to SEs in androgen-dependent PCa cells, thereby promoting oncogenic transcriptional programs. In this work, we reveal that full-length AR forms foci with liquid-like properties in different PCa models. We demonstrate that foci formation correlates with AR transcriptional activity, as this activity can be modulated by changing cellular foci content chemically or by silencing MED1. AR ability to phase separate was also validated in vitro by using recombinant full-length AR protein. We also demonstrate that AR antagonists, which suppress transcriptional activity by targeting key regions for homotypic or heterotypic interactions of this receptor, hinder foci formation in PCa cells and phase separation in vitro. Our results suggest that enhanced compartmentalization of AR and coactivators may play an important role in the activation of oncogenic transcription programs in androgen-dependent PCa.


Assuntos
Neoplasias da Próstata , Receptores Androgênicos , Masculino , Humanos , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Androgênios , Fatores de Transcrição/metabolismo , Regulação da Expressão Gênica , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Expressão Gênica , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica
2.
Int J Mol Sci ; 22(5)2021 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-33801338

RESUMO

The inhibition of the androgen receptor (AR) is an established strategy in prostate cancer (PCa) treatment until drug resistance develops either through mutations in the ligand-binding domain (LBD) portion of the receptor or its deletion. We previously identified a druggable pocket on the DNA binding domain (DBD) dimerization surface of the AR and reported several potent inhibitors that effectively disrupted DBD-DBD interactions and consequently demonstrated certain antineoplastic activity. Here we describe further development of small molecule inhibitors of AR DBD dimerization and provide their broad biological characterization. The developed compounds demonstrate improved activity in the mammalian two-hybrid assay, enhanced inhibition of AR-V7 transcriptional activity, and improved microsomal stability. These findings position us for the development of AR inhibitors with entirely novel mechanisms of action that would bypass most forms of PCa treatment resistance, including the truncation of the LBD of the AR.


Assuntos
Antagonistas de Receptores de Andrógenos/farmacologia , DNA de Neoplasias/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , Receptores Androgênicos/química , Bibliotecas de Moléculas Pequenas/farmacologia , Transcrição Gênica , Antagonistas de Receptores de Andrógenos/química , Simulação por Computador , DNA de Neoplasias/antagonistas & inibidores , Ensaios de Triagem em Larga Escala , Humanos , Masculino , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Conformação Proteica , Domínios Proteicos , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Bibliotecas de Moléculas Pequenas/química , Células Tumorais Cultivadas
3.
Med Res Rev ; 40(1): 413-430, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-30927317

RESUMO

The ETS family of proteins consists of 28 transcription factors, many of which have been implicated in development and progression of a variety of cancers. While one family member, ERG, has been rigorously studied in the context of prostate cancer where it plays a critical role, other ETS factors keep emerging as potential hallmark oncodrivers. In recent years, numerous studies have reported initial discoveries of small molecule inhibitors of ETS proteins and opened novel avenues for ETS-directed cancer therapies. This review summarizes the state of the art data on therapeutic targeting of ETS family members and highlights the corresponding drug discovery strategies.


Assuntos
Sistemas de Liberação de Medicamentos , Neoplasias/tratamento farmacológico , Proteínas Proto-Oncogênicas c-ets/metabolismo , Sequência de Aminoácidos , Animais , Humanos , Proteínas Proto-Oncogênicas c-ets/química , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Bibliotecas de Moléculas Pequenas/uso terapêutico
4.
Int J Mol Sci ; 21(12)2020 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-32545494

RESUMO

Breast cancer (BCa) is one of the most predominantly diagnosed cancers in women. Notably, 70% of BCa diagnoses are Estrogen Receptor α positive (ERα+) making it a critical therapeutic target. With that, the two subtypes of ER, ERα and ERß, have contrasting effects on BCa cells. While ERα promotes cancerous activities, ERß isoform exhibits inhibitory effects on the same. ER-directed small molecule drug discovery for BCa has provided the FDA approved drugs tamoxifen, toremifene, raloxifene and fulvestrant that all bind to the estrogen binding site of the receptor. These ER-directed inhibitors are non-selective in nature and may eventually induce resistance in BCa cells as well as increase the risk of endometrial cancer development. Thus, there is an urgent need to develop novel drugs with alternative ERα targeting mechanisms that can overcome the limitations of conventional anti-ERα therapies. Several functional sites on ERα, such as Activation Function-2 (AF2), DNA binding domain (DBD), and F-domain, have been recently considered as potential targets in the context of drug research and discovery. In this review, we summarize methods of computer-aided drug design (CADD) that have been employed to analyze and explore potential targetable sites on ERα, discuss recent advancement of ERα inhibitor development, and highlight the potential opportunities and challenges of future ERα-directed drug discovery.


Assuntos
Neoplasias da Mama/metabolismo , Receptor alfa de Estrogênio/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Sítios de Ligação/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Simulação por Computador , Desenho Assistido por Computador , Resistência a Medicamentos/efeitos dos fármacos , Receptor alfa de Estrogênio/química , Feminino , Humanos , Ligantes , Bibliotecas de Moléculas Pequenas/uso terapêutico
5.
Molecules ; 24(4)2019 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-30791548

RESUMO

The heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) is a versatile RNA-binding protein playing a critical role in alternative pre-mRNA splicing regulation in cancer. Emerging data have implicated hnRNP A1 as a central player in a splicing regulatory circuit involving its direct transcriptional control by c-Myc oncoprotein and the production of the constitutively active ligand-independent alternative splice variant of androgen receptor, AR-V7, which promotes castration-resistant prostate cancer (CRPC). As there is an urgent need for effective CRPC drugs, targeting hnRNP A1 could, therefore, serve a dual purpose of preventing AR-V7 generation as well as reducing c-Myc transcriptional output. Herein, we report compound VPC-80051 as the first small molecule inhibitor of hnRNP A1 splicing activity discovered to date by using a computer-aided drug discovery approach. The inhibitor was developed to target the RNA-binding domain (RBD) of hnRNP A1. Further experimental evaluation demonstrated that VPC-80051 interacts directly with hnRNP A1 RBD and reduces AR-V7 messenger levels in 22Rv1 CRPC cell line. This study lays the groundwork for future structure-based development of more potent and selective small molecule inhibitors of hnRNP A1⁻RNA interactions aimed at altering the production of cancer-specific alternative splice isoforms.


Assuntos
Biologia Computacional , Descoberta de Drogas , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Ribonucleoproteína Nuclear Heterogênea A1/genética , Neoplasias de Próstata Resistentes à Castração/genética , Splicing de RNA/efeitos dos fármacos , Sítios de Ligação , Linhagem Celular Tumoral , Biologia Computacional/métodos , Simulação por Computador , Descoberta de Drogas/métodos , Ribonucleoproteína Nuclear Heterogênea A1/química , Humanos , Masculino , Modelos Moleculares , Conformação Molecular , Relação Estrutura-Atividade
6.
Prostate ; 78(6): 457-468, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29450905

RESUMO

BACKGROUND: Patients with advanced prostate cancer have limited curative options, therefore new treatments are needed. Mouse models play a pivotal role in the discovery and development of new treatments. In the present study, a TRAMP-derived Orthotopic Prostate Syngeneic (TOPS) mouse model was developed and found to provide a consistent means of monitoring tumor and metastatic responses to novel treatments. METHODS: The mouse TOPS model was generated using luciferase transduced TRAMP-C2 prostate cancer cells that were orthotopically injected into Bl6 mice by ultrasound guidance. Tumor growth and development was monitored using ultrasound and bioluminescence imaging. RESULTS: Tumors and metastases were consistently established and increases in tumor size correlated with increases in bioluminescence. In addition, when mice with an established tumor were castrated, tumor progression mirrored clinical progression. We further treated the TOPS model with an oncolytic Herpes Simplex virus and showed that we were able to monitor the therapeutic effect of the orthotopic tumor after virus treatment through IVIS imaging system. CONCLUSION: We have developed a powerful animal model to advance the current selection of effective treatments for patients with advanced prostate cancer.


Assuntos
Adenocarcinoma/patologia , Antineoplásicos/uso terapêutico , Modelos Animais de Doenças , Neoplasias da Próstata/patologia , Adenocarcinoma/tratamento farmacológico , Animais , Progressão da Doença , Masculino , Camundongos , Camundongos Transgênicos , Próstata/patologia , Neoplasias da Próstata/tratamento farmacológico
7.
J Chem Inf Model ; 58(8): 1533-1543, 2018 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-30063345

RESUMO

The majority of computational methods for predicting toxicity of chemicals are typically based on "nonmechanistic" cheminformatics solutions, relying on an arsenal of QSAR descriptors, often vaguely associated with chemical structures, and typically employing "black-box" mathematical algorithms. Nonetheless, such machine learning models, while having lower generalization capacity and interpretability, typically achieve a very high accuracy in predicting various toxicity endpoints, as unambiguously reflected by the results of the recent Tox21 competition. In the current study, we capitalize on the power of modern AI to predict Tox21 benchmark data using merely simple 2D drawings of chemicals, without employing any chemical descriptors. In particular, we have processed rather trivial 2D sketches of molecules with a supervised 2D convolutional neural network (2DConvNet) and demonstrated that the modern image recognition technology results in prediction accuracies comparable to the state-of-the-art cheminformatics tools. Furthermore, the performance of the image-based 2DConvNet model was comparatively evaluated on an external set of compounds from the Prestwick chemical library and resulted in experimental identification of significant and previously unreported antiandrogen potentials for several well-established generic drugs.


Assuntos
Aprendizado Profundo , Descoberta de Drogas , Modelos Biológicos , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/toxicidade , Algoritmos , Gráficos por Computador , Bases de Dados de Produtos Farmacêuticos , Descoberta de Drogas/métodos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/etiologia , Humanos , Modelos Químicos , Preparações Farmacêuticas/química
8.
Int J Mol Sci ; 20(1)2018 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-30597997

RESUMO

Myc (avian myelocytomatosis viral oncogene homolog) represents one of the most sought after drug targets in cancer. Myc transcription factor is an essential regulator of cell growth, but in most cancers it is overexpressed and associated with treatment-resistance and lethal outcomes. Over 40 years of research and drug development efforts did not yield a clinically useful Myc inhibitor. Drugging the "undruggable" is problematic, as Myc inactivation may negatively impact its physiological functions. Moreover, Myc is a disordered protein that lacks effective binding pockets on its surface. It is well established that the Myc function is dependent on dimerization with its obligate partner, Max (Myc associated factor X), which together form a functional DNA-binding domain to activate genomic targets. Herein, we provide an overview of the knowledge accumulated to date on Myc regulation and function, its critical role in cancer, and summarize various strategies that are employed to tackle Myc-driven malignant transformation. We focus on important structure-function relationships of Myc with its interactome, elaborating structural determinants of Myc-Max dimer formation and DNA recognition exploited for therapeutic inhibition. Chronological development of small-molecule Myc-Max prototype inhibitors and corresponding binding sites are comprehensively reviewed and particular emphasis is placed on modern computational drug design methods. On the outlook, technological advancements may soon provide the so long-awaited Myc-Max clinical candidate.


Assuntos
Antineoplásicos/farmacologia , Descoberta de Drogas/métodos , Simulação de Acoplamento Molecular/métodos , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Animais , Antineoplásicos/química , Humanos , Ligação Proteica , Relação Quantitativa Estrutura-Atividade , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/metabolismo
9.
Int J Mol Sci ; 19(2)2018 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-29462880

RESUMO

Estrogen receptor-α positive (ERα⁺) breast cancers represent 75% of all invasive breast cancer cases, while de novo or acquired resistance to ER-directed therapy is also on the rise. Numerous factors contribute to this phenomenon including the recently-reported ESR1 gene mutations such as Y537S, which amplifies co-activator interactions with ERα and promotes constitutive activation of ERα function. Herein, we propose that direct targeting of the activation function-2 (AF2) site on ERα represents a promising alternative therapeutic strategy to overcome mutation-driven resistance in breast cancer. A systematic computer-guided drug discovery approach was employed to develop a potent ERα inhibitor that was extensively evaluated by a series of experiments to confirm its AF2-specific activity. We demonstrate that the developed small-molecule inhibitor effectively prevents ERα-coactivator interactions and exhibits a strong anti-proliferative effect against tamoxifen-resistant cells, as well as downregulates ERα-dependent genes and effectively diminishes the receptor binding to chromatin. Notably, the identified lead compound successfully inhibits known constitutively-active, resistance-associated mutant forms of ERα observed in clinical settings. Overall, this study reports the development of a novel class of ERα AF2 inhibitors, which have the potential to effectively inhibit ERα activity by a unique mechanism and to circumvent the issue of mutation-driven resistance in breast cancer.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/genética , Receptor alfa de Estrogênio/genética , Tiofenos/administração & dosagem , Sítios de Ligação/efeitos dos fármacos , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Cromatina/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Mutação , Ligação Proteica , Tamoxifeno/administração & dosagem , Tamoxifeno/efeitos adversos
10.
Molecules ; 23(11)2018 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-30441799

RESUMO

Orphan nuclear receptor TLX (NR2E1) plays a critical role in the regulation of neural stem cells (NSC) as well as in the development of NSC-derived brain tumors. In the last years, new data have emerged implicating TLX in prostate and breast cancer. Therefore, inhibitors of TLX transcriptional activity may have a significant impact on the treatment of several critical malignancies. However, the TLX protein possesses a non-canonical ligand-binding domain (LBD), which lacks a ligand-binding pocket (conventionally targeted in case of nuclear receptors) that complicates the development of small molecule inhibitors of TLX. Herein, we utilized a rational structure-based design approach to identify small molecules targeting the Atro-box binding site of human TLX LBD. As a result of virtual screening of ~7 million molecular structures, 97 compounds were identified and evaluated in the TLX-responsive luciferase reporter assay. Among those, three chemicals demonstrated 40⁻50% inhibition of luciferase-detected transcriptional activity of the TLX orphan nuclear receptor at a dose of 35 µM. The identified compounds represent the first class of small molecule inhibitors of TLX transcriptional activity identified via methods of computer-aided drug discovery.


Assuntos
Desenho Assistido por Computador , Desenho de Fármacos , Modelos Moleculares , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/genética , Ativação Transcricional/efeitos dos fármacos , Sítios de Ligação , Regulação da Expressão Gênica , Genes Reporter , Humanos , Conformação Molecular , Estrutura Molecular , Receptores Nucleares Órfãos , Ligação Proteica , Relação Quantitativa Estrutura-Atividade
11.
J Chem Inf Model ; 57(5): 1018-1028, 2017 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-28441481

RESUMO

Small-molecule drug design is a complex and iterative decision-making process relying on pre-existing knowledge and driven by experimental data. Low-molecular-weight chemicals represent an attractive therapeutic option, as they are readily accessible to organic synthesis and can easily be characterized.1 Their potency as well as pharmacokinetic and pharmacodynamic properties can be systematically and rationally investigated and ultimately optimized via expert science behind medicinal chemistry and methods of computer-aided drug design (CADD). In recent years, significant advances in molecular modeling techniques have afforded a variety of tools to effectively identify potential binding pockets on prospective targets, to map key interactions between ligands and their binding sites, to construct and assess energetics of the resulting complexes, to predict ADMET properties of candidate compounds, and to systematically analyze experimental and computational data to derive meaningful structure-activity relationships leading to the creation of a drug candidate. This Perspective describes a real case of a drug discovery campaign accomplished in a relatively short time with limited resources. The study integrated an arsenal of available molecular modeling techniques with an array of experimental tools to successfully develop a novel class of potent and selective androgen receptor inhibitors with a novel mode of action. It resulted in the largest academic licensing deal in Canadian history, totaling $142M. This project exemplifies the importance of team science, an integrative approach to drug discovery, and the use of best practices in CADD. We posit that the lessons learned and best practices for executing an effective CADD project can be applied, with similar success, to many drug discovery projects in both academia and industry.


Assuntos
Biologia Computacional , Descoberta de Drogas , Neoplasias da Próstata , Bibliotecas de Moléculas Pequenas/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Sítios de Ligação/efeitos dos fármacos , Humanos , Masculino , Modelos Moleculares , Neoplasias da Próstata/tratamento farmacológico , Receptores Androgênicos/metabolismo , Alinhamento de Sequência , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/uso terapêutico
12.
J Chem Inf Model ; 56(12): 2507-2516, 2016 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-28024400

RESUMO

The human androgen receptor (AR) is a ligand-activated transcription factor that plays a pivotal role in the development and progression of prostate cancer (PCa). Many forms of castration-resistant prostate cancer (CRPC) still rely on the AR for survival. Currently used antiandrogens face clinical limitations as drug resistance develops in patients over time since they all target the mutation-prone androgen binding site (ABS), where gain-of-function mutations eventually convert antagonists into agonists. With a significant number of reported distinct mutations located across the ABS, it is imperative to develop a prognostic platform which would equip clinicians with prior knowledge and actionable strategies if cases of previously unreported AR mutations are encountered. The goal of this study is to develop a theoretical approach that can predict such previously unreported AR mutants in response to current treatment options for PCa. The expected drug response by these mutants has been modeled using cheminformatics methodology. The corresponding QSAR pipeline has been created, which extracts key protein-ligand interactions and quantifies them by 4D molecular descriptors. The developed models reported with an accuracy reaching 90% and enable prediction of activation of AR mutants by its native ligand as well as assess whether known antiandrogens will act on them as agonists or antagonists. As a result, a previously uncharacterized mutant, T878G, has been predicted to be activated by the latest antiandrogen enzalutamide, and the corresponding experimental evaluation confirmed this prediction. Overall, the developed cheminformatics pipeline provides useful insights toward understanding the changing genomic landscape of advanced PCa.


Assuntos
Antagonistas de Receptores de Andrógenos/farmacologia , Feniltioidantoína/análogos & derivados , Mutação Puntual/efeitos dos fármacos , Receptores Androgênicos/metabolismo , Antagonistas de Receptores de Andrógenos/química , Androgênios/química , Androgênios/farmacologia , Benzamidas , Humanos , Masculino , Modelos Moleculares , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Nitrilas , Feniltioidantoína/química , Feniltioidantoína/farmacologia , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Receptores Androgênicos/genética
13.
J Biol Chem ; 289(38): 26417-26429, 2014 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-25086042

RESUMO

The androgen receptor (AR) is a transcription factor that has a pivotal role in the occurrence and progression of prostate cancer. The AR is activated by androgens that bind to its ligand-binding domain (LBD), causing the transcription factor to enter the nucleus and interact with genes via its conserved DNA-binding domain (DBD). Treatment for prostate cancer involves reducing androgen production or using anti-androgen drugs to block the interaction of hormones with the AR-LBD. Eventually the disease changes into a castration-resistant form of PCa where LBD mutations render anti-androgens ineffective or where constitutively active AR splice variants, lacking the LBD, become overexpressed. Recently, we identified a surfaced exposed pocket on the AR-DBD as an alternative drug-target site for AR inhibition. Here, we demonstrate that small molecules designed to selectively bind the pocket effectively block transcriptional activity of full-length and splice variant AR forms at low to sub-micromolar concentrations. The inhibition is lost when residues involved in drug interactions are mutated. Furthermore, the compounds did not impede nuclear localization of the AR and blocked interactions with chromatin, indicating the interference of DNA binding with the nuclear form of the transcription factor. Finally, we demonstrate the inhibition of gene expression and tumor volume in mouse xenografts. Our results indicate that the AR-DBD has a surface site that can be targeted to inhibit all forms of the AR, including enzalutamide-resistant and constitutively active splice variants and thus may serve as a potential avenue for the treatment of recurrent and metastatic prostate cancer.


Assuntos
Antagonistas de Receptores de Andrógenos/farmacologia , Imidazóis/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Receptores Androgênicos/fisiologia , Tiazóis/farmacologia , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Animais , Sítios de Ligação , Núcleo Celular/metabolismo , Cromatina/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Masculino , Camundongos Nus , Dados de Sequência Molecular , Terapia de Alvo Molecular , Neoplasias da Próstata/patologia , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/fisiologia , Receptores Androgênicos/química , Transcrição Gênica , Ativação Transcricional , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Breast Cancer Res ; 17: 27, 2015 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-25848700

RESUMO

INTRODUCTION: Current approaches to inhibit oestrogen receptor-alpha (ERα) are focused on targeting its hormone-binding pocket and have limitations. Thus, we propose that inhibitors that bind to a coactivator-binding pocket on ERα, called activation function 2 (AF2), might overcome some of these limitations. METHODS: In silico virtual screening was used to identify small-molecule ERα AF2 inhibitors. These compounds were screened for inhibition of ERα transcriptional activity using stably transfected T47D-KBluc cell line. A direct physical interaction between the AF2 binders and the ERα protein was measured using biolayer interferometry (BLI) and an ERα coactivator displacement assay. Cell viability was assessed by MTS assay in ERα-positive MCF7 cells, tamoxifen-resistant (TamR) cell lines TamR3 and TamR6, and ERα-negative MDA-MB-453 and HeLa cell lines. In addition, ERα inhibition in TamR cells and the effect of compounds on mRNA and protein expression of oestrogen-dependent genes, pS2, cathepsin D and cell division cycle 2 (CDC2) were determined. RESULTS: Fifteen inhibitors from two chemical classes, derivatives of pyrazolidine-3,5-dione and carbohydrazide, were identified. In a series of in vitro assays, VPC-16230 of the carbohydrazide chemical class emerged as a lead ERα AF2 inhibitor that significantly downregulated ERα transcriptional activity (half-maximal inhibitory concentration = 5.81 µM). By directly binding to the ERα protein, as confirmed by BLI, VPC-16230 effectively displaced coactivator peptides from the AF2 pocket, confirming its site-specific action. VPC-16230 selectively suppressed the growth of ERα-positive breast cancer cells. Furthermore, it significantly inhibited ERα mediated transcription in TamR cells. More importantly, it reduced mRNA and protein levels of pS2, cathepsin D and CDC2, validating its ER-directed activity. CONCLUSION: We identified VPC-16230 as an ERα AF2-specific inhibitor that demonstrated promising antiproliferative effects in breast cancer cell lines, including TamR cells. VPC-16230 reduced the expression of ERα-inducible genes, including CDC2, which is involved in cell division. We anticipate that the application of ERα AF2 inhibitors will provide a novel approach that can act as a complementary therapeutic to treat ERα-positive, tamoxifen-resistant and metastatic breast cancers.


Assuntos
Antineoplásicos/química , Domínio Catalítico , Descoberta de Drogas , Moduladores de Receptor Estrogênico/química , Receptor alfa de Estrogênio/química , Antineoplásicos/farmacologia , Sítios de Ligação , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Simulação por Computador , Descoberta de Drogas/métodos , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Estradiol/metabolismo , Moduladores de Receptor Estrogênico/farmacologia , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/genética , Estrogênios/metabolismo , Feminino , Humanos , Ligantes , Células MCF-7 , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Reprodutibilidade dos Testes , Bibliotecas de Moléculas Pequenas , Transcrição Gênica/efeitos dos fármacos
15.
Prostate ; 75(10): 1043-50, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25833156

RESUMO

BACKGROUND: The progesterone receptor, like the androgen receptor, belongs to the steroid receptor superfamily. Our previous studies have reported that the PR is expressed specifically in prostate stroma. PR inhibits proliferation of, and regulates cytokine secretion by stromal cells. However, PR protein expression in cancer-associated stroma during prostate cancer progression has not been profiled. Since the phenotypes of prostate stromal cells change dynamically as tumors progress, whether the PR plays a role in regulating stromal cell differentiation needs to be investigated. METHODS: Immunohistochemistry assays measured PR protein levels on human prostate tissue microarrays containing 367 tissue cores from benign prostate, prostate tumors with different Gleason scores, tumors under various durations of castration therapy, and tumors at the castration-resistant stage. Immunoblotting assays determined whether PR regulated the expression of alpha smooth muscle actin (α-SMA), vimentin, and fibroblast specific protein (FSP) in human prostate stromal cells. RESULTS: PR protein levels decreased in cancer-associated stroma when compared with that in benign prostate stroma. This reduction in PR expression was not correlated with Gleason scores. PR protein levels were elevated by castration therapy, but reduced to pre-castration levels when tumors progressed to the castration-resistant stage. Enhanced PR expression in human prostate stromal cells increased α-SMA, but decreased vimentin and FSP protein levels ligand-independently. CONCLUSION: These results suggest that PR plays an active role in regulating stromal cell phenotypes during prostate cancer progression.


Assuntos
Diferenciação Celular/fisiologia , Neoplasias da Próstata/patologia , Receptores de Progesterona/metabolismo , Células Estromais/patologia , Actinas/análise , Adulto , Idoso , Proteínas de Ligação ao Cálcio/análise , Humanos , Masculino , Pessoa de Meia-Idade , Orquiectomia , Próstata/química , Neoplasias da Próstata/química , Vimentina/análise
16.
EMBO J ; 30(13): 2719-33, 2011 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-21602788

RESUMO

The androgen receptor (AR) is a key regulator of prostate growth and the principal drug target for the treatment of prostate cancer. Previous studies have mapped AR targets and identified some candidates which may contribute to cancer progression, but did not characterize AR biology in an integrated manner. In this study, we took an interdisciplinary approach, integrating detailed genomic studies with metabolomic profiling and identify an anabolic transcriptional network involving AR as the core regulator. Restricting flux through anabolic pathways is an attractive approach to deprive tumours of the building blocks needed to sustain tumour growth. Therefore, we searched for targets of the AR that may contribute to these anabolic processes and could be amenable to therapeutic intervention by virtue of differential expression in prostate tumours. This highlighted calcium/calmodulin-dependent protein kinase kinase 2, which we show is overexpressed in prostate cancer and regulates cancer cell growth via its unexpected role as a hormone-dependent modulator of anabolic metabolism. In conclusion, it is possible to progress from transcriptional studies to a promising therapeutic target by taking an unbiased interdisciplinary approach.


Assuntos
Carcinoma/genética , Carcinoma/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/fisiologia , Animais , Sequência de Bases , Sítios de Ligação/genética , Vias Biossintéticas/genética , Carcinoma/patologia , Linhagem Celular Tumoral , Proliferação de Células , Análise por Conglomerados , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Metabolismo/genética , Metabolismo/fisiologia , Camundongos , Modelos Biológicos , Neoplasias da Próstata/patologia , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Elementos de Resposta/genética , Transplante Heterólogo
17.
Arch Ital Urol Androl ; 87(2): 121-9, 2015 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-26150028

RESUMO

OBJECTIVE: We performed a systematic review of the literature to assess the efficacy and the safety of second-line agents targeting metastatic castration-resistant prostate cancer (mCRPC) that has progressed after docetaxel. Pooled-analysis was also performed, to assess the effectiveness of agents targeting the androgen axis via identical mechanisms of action (abiraterone acetate, orteronel). MATERIALS AND METHODS: We included phase III randomized controlled trials that enrolled patients with mCRPC progressing during or after first-line docetaxel treatment. Trials were identified by electronic database searching. The primary outcome of the review was overall survival. Secondary outcomes were radiographic progression-free survival (rPFS) and severe adverse effects (grade 3 or higher). RESULTS: Ten articles met the inclusion criteria for the review. These articles reported the results of five clinical trials, enrolling in total 5047 patients. The experimental interventions tested in these studies were enzalutamide, ipilimumab, abiraterone acetate, orteronel and cabazitaxel. Compared to control cohorts (active drug-treated or placebo-treated), the significant overall survival advantages achieved were 4.8 months for enzalutamide (hazard ratio for death vs. placebo: 0.63; 95% CI 0.53 to 0.75, P < 0.0001), 4.6 months for abiraterone (hazard ratio for death vs. placebo: 0.66, 95% CI 0.58 to 0.75, P < 0.0001) and 2.4 months for cabazitaxel (hazard ratio for death vs. mitoxantrone-prednisone: 0.70, 95% CI 0.59 to 0.83, p < 0.0001). Pooled analysis of androgen synthesis inhibitors orteronel and abiraterone resulted in significantly increased overall and progression-free survival for anti-androgen agents, compared to placebo (hazard ratio for death: 0.76, 95% CI 0.67 to 0.87, P < 0.0001; hazard ratio for radiographic progression: 0.7, 95% CI 0.63 to 0.77, P < 0.00001). Androgen synthesis inhibitors induced significant increases in risk ratios for adverse effects linked to elevated mineralocorticoid secretion, compared to placebo (risk ratio for hypokalemia: 5.75, 95% CI 2.08 to 15.90; P = 0.0008; risk-ratio for hypertension: 2.29, 95% CI 1.02 to 5.17; P = 0.05). CONCLUSIONS: In docetaxel-pretreated patients enzalutamide, abiraterone-prednisone and cabazitaxel-prednisone can improve overall survival of patients, compared to placebo or to best of care at the time of study (mitoxantrone-prednisone). Agents targeting the androgen axis (enzalutamide, abiraterone, orteronel) significantly prolonged rPFS, compared to placebo. Further investigation is warranted to evaluate the benefit of combination or sequential administration of these agents. Large-scale studies are also necessary to evaluate the impact of relevant toxic effects observed in a limited number of patients (e.g., enzalutamide-induced seizures, orteronel-induced pancreatitis, and others).


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Acetato de Abiraterona , Androstenos/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Benzamidas , Progressão da Doença , Medicina Baseada em Evidências , Humanos , Imidazóis/administração & dosagem , Ipilimumab , Masculino , Naftalenos/administração & dosagem , Nitrilas , Feniltioidantoína/administração & dosagem , Feniltioidantoína/análogos & derivados , Neoplasias de Próstata Resistentes à Castração/mortalidade , Neoplasias de Próstata Resistentes à Castração/patologia , Neoplasias de Próstata Resistentes à Castração/secundário , Ensaios Clínicos Controlados Aleatórios como Assunto , Fatores de Risco , Análise de Sobrevida , Taxoides/administração & dosagem , Fatores de Tempo , Resultado do Tratamento
18.
Hum Mol Genet ; 21(14): 3112-27, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22505583

RESUMO

MicroRNAs (miRs) play an important role in the development of many complex human diseases and may have tumour suppressor or oncogenic (oncomir) properties. Prostate cancer is initially an androgen-driven disease, and androgen receptor (AR) remains a key driver of growth even in castration-resistant tumours. However, AR-mediated oncomiR pathways remain to be elucidated. We demonstrate that miR-27a is an androgen-regulated oncomir in prostate cancer, acting via targeting the tumour suppressor and AR corepressor, Prohibitin (PHB). Increasing miR-27a expression results in reduced PHB mRNA and protein levels, and increased expression of AR target genes and prostate cancer cell growth. This involves a novel mechanism for androgen-mediated miR regulation, whereby AR induces a transient increase in miR-23a27a24-2 transcription, but more significantly accelerates processing of the primiR-23a27a24-2 cluster. Androgens therefore regulate miR-27a expression both transcriptionally (via AR binding to the cluster promoter) and post-transcriptionally (accelerating primiR processing to the mature form). We further show that a miR-27a anti-sense oligonucleotide, by opposing the effects of mir-27a, has therapeutic potential in prostate cancer.


Assuntos
Androgênios/metabolismo , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Proteínas Oncogênicas/genética , Neoplasias da Próstata/genética , Processamento Pós-Transcricional do RNA , Proteínas Repressoras/genética , Sequência de Bases , Linhagem Celular Tumoral , Humanos , Masculino , MicroRNAs/metabolismo , Dados de Sequência Molecular , Proteínas Oncogênicas/metabolismo , Proibitinas , Neoplasias da Próstata/metabolismo , Ligação Proteica , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Proteínas Repressoras/metabolismo
19.
Mol Ther ; 21(4): 842-8, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23337981

RESUMO

Vesicular stomatitis virus (VSV) is an oncolytic virus which selectively infects and kills cancer cells. The goal of the present study was to determine whether VSV is capable of targeting metastatic lesions that arise in situ in the transgenic adenocarcinoma of the mouse prostate (TRAMP) model. The interferon (IFN)-responsive luciferase containing VSV(AV3) strain was injected intraprostatically into both control and TRAMP mice. Distribution, infectivity, apoptosis, and status of the IFN response were evaluated at the site of viral injection (prostate), as well as in metastatic lesions (lymph nodes), through plaque, polymerase chain reaction (PCR), and immunohistochemical analysis. Bioluminescence analyses demonstrated that VSV(AV3) persisted at high levels in the prostate region of TRAMP mice for up to 96 hours, but at relatively low levels and for only 48 hours in control mice. Live virus was discovered in the lymph nodes of TRAMP mice, but not in control mice. TUNEL staining revealed increased cell death in VSV(AV3) infected metastatic cells present in the lymph nodes of TRAMP mice. There was an evidence of IFN activation in lymph nodes containing metastatic cells. Our results indicate that intraprostatic injections of VSV(AV3) can be used as a means to infect and kill metastatic lesions associated with advanced prostate cancer.


Assuntos
Adenocarcinoma/terapia , Metástase Neoplásica/prevenção & controle , Terapia Viral Oncolítica/métodos , Neoplasias da Próstata/terapia , Vírus da Estomatite Vesicular Indiana/fisiologia , Animais , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Vírus da Estomatite Vesicular Indiana/genética
20.
Sci Rep ; 14(1): 7082, 2024 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-38528115

RESUMO

FOXA1 is a pioneer transcription factor that is frequently mutated in prostate, breast, bladder, and salivary gland malignancies. Indeed, metastatic castration-resistant prostate cancer (mCRPC) commonly harbour FOXA1 mutations with a prevalence of 35%. However, despite the frequent recurrence of FOXA1 mutations in prostate cancer, the mechanisms by which FOXA1 variants drive its oncogenic effects are still unclear. Semaphorin 3C (SEMA3C) is a secreted autocrine growth factor that drives growth and treatment resistance of prostate and other cancers and is known to be regulated by both AR and FOXA1. In the present study, we characterize FOXA1 alterations with respect to its regulation of SEMA3C. Our findings reveal that FOXA1 alterations lead to elevated levels of SEMA3C both in prostate cancer specimens and in vitro. We further show that FOXA1 negatively regulates SEMA3C via intronic cis elements, and that mutations in FOXA1 forkhead domain attenuate its inhibitory function in reporter assays, presumably by disrupting DNA binding of FOXA1. Our findings underscore the key role of FOXA1 in prostate cancer progression and treatment resistance by regulating SEMA3C expression and suggest that SEMA3C may be a driver of growth and tumor vulnerability of mCRPC harboring FOXA1 alterations.


Assuntos
Fator 3-alfa Nuclear de Hepatócito , Neoplasias de Próstata Resistentes à Castração , Semaforinas , Humanos , Masculino , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Fator 3-alfa Nuclear de Hepatócito/genética , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Mutação , Próstata/patologia , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/patologia , Fatores de Transcrição/metabolismo , Semaforinas/genética , Semaforinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA