Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cancer Ther ; 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38894678

RESUMEN

Polo-like kinase 1 (PLK1) inhibitors have had limited antitumor efficacy as single agents, and a focus of current efforts is on combination therapies. We initially confirmed that the PLK1 specific inhibitor onvansertib (ONV) could enhance responses to a PARP inhibitor (olaparib) in prostate cancer xenografts. To identify more effective combinations we screened a library of bioactive compounds for efficacy in combination with ONV in LNCaP prostate cancer cells, which identified a series of compounds including multiple AKT inhibitors. We confirmed in vitro synergy between ONV and the AKT inhibitor ipatasertib (IPA) and found that the combination increased apoptosis. Mechanistic studies showed that ONV increased expression of the anti-apoptotic protein SURVIVIN, and that this was mitigated by IPA. Studies in three PTEN deficient prostate cancer xenograft models showed that co-treatment with IPA and ONV led to significant tumor growth inhibition compared to monotherapies. Together these in vitro and in vivo studies demonstrate that the efficacy of PLK1 antagonists can be enhanced by PARP or AKT inhibition, and support further development of these combination therapies.

2.
Prostate ; 84(10): 909-921, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38619005

RESUMEN

INTRODUCTION: Lysine-specific demethylase 1 (LSD1) is emerging as a critical mediator of tumor progression in metastatic castration-resistant prostate cancer (mCRPC). Neuroendocrine prostate cancer (NEPC) is increasingly recognized as an adaptive mechanism of resistance in mCRPC patients failing androgen receptor axis-targeted therapies. Safe and effective LSD1 inhibitors are necessary to determine antitumor response in prostate cancer models. For this reason, we characterize the LSD1 inhibitor bomedemstat to assess its clinical potential in NEPC as well as other mCRPC pathological subtypes. METHODS: Bomedemstat was characterized via crystallization, flavine adenine dinucleotide spectrophotometry, and enzyme kinetics. On-target effects were assessed in relevant prostate cancer cell models by measuring proliferation and H3K4 methylation using western blot analysis. In vivo, pharmacokinetic (PK) and pharmacodynamic (PD) profiles of bomedemstat are also described. RESULTS: Structural, biochemical, and PK/PD properties of bomedemstat, an irreversible, orally-bioavailable inhibitor of LSD1 are reported. Our data demonstrate bomedemstat has >2500-fold greater specificity for LSD1 over monoamine oxidase (MAO)-A and -B. Bomedemstat also demonstrates activity against several models of advanced CRPC, including NEPC patient-derived xenografts. Significant intra-tumoral accumulation of orally-administered bomedemstat is measured with micromolar levels achieved in vivo (1.2 ± 0.45 µM at the 7.5 mg/kg dose and 3.76 ± 0.43 µM at the 15 mg/kg dose). Daily oral dosing of bomedemstat at 40 mg/kg/day is well-tolerated, with on-target thrombocytopenia observed that is rapidly reversible following treatment cessation. CONCLUSIONS: Bomedemstat provides enhanced specificity against LSD1, as revealed by structural and biochemical data. PK/PD data display an overall safety profile with manageable side effects resulting from LSD1 inhibition using bomedemstat in preclinical models. Altogether, our results support clinical testing of bomedemstat in the setting of mCRPC.


Asunto(s)
Histona Demetilasas , Neoplasias de la Próstata Resistentes a la Castración , Histona Demetilasas/antagonistas & inhibidores , Histona Demetilasas/metabolismo , Masculino , Humanos , Animales , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Ratones , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/farmacocinética , Benzamidas , Piperazinas , Triazoles
3.
Prostate ; 83(15): 1470-1493, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37559436

RESUMEN

BACKGROUND: The quinoline-3-carboxamide, Tasquinimod (TasQ), is orally active as a maintenance therapy with an on-target mechanism-of-action via allosteric binding to HDAC4. This prevents formation of the HDAC4/NCoR1/HDAC3 complex, disrupting HIF-1α transcriptional activation and repressing MEF-2 target genes needed for adaptive survival signaling in the compromised tumor micro environment. In phase 3 clinical testing against metastatic castration-resistant prostate cancer(mCRPC), TasQ (1 mg/day) increased time-to-progression, but not overall survival. METHODS: TasQ analogs were chemically synthesized and tested for activity compared to the parental compound. These included HDAC4 enzymatic assays, qRT-PCR and western blot analyses of gene and protein expression following treatment, in vitro and in vivo efficacy against multiple prostate cancer models including PDXs, pharmacokinetic analyses,AHR binding and agonist assays, SPR analyses of binding to HDAC4 and NCoR1, RNAseq analysis of in vivo tumors, 3D endothelial sprouting assays, and a targeted kinase screen. Genetic knockout or knockdown controls were used when appropriate. RESULTS: Here, we document that, on this regimen (1 mg/day), TasQ blood levels are 10-fold lower than the optimal concentration (≥2 µM) needed for anticancer activity, suggesting higher daily doses are needed. Unfortunately, we also demonstrate that TasQ is an arylhydrocarbon receptor (AHR) agonist, which binds with an EC50 of 1 µM to produce unwanted off-target side effects. Therefore, we screened a library of TasQ analogsto maximize on-target versus off-target activity. Using this approach, we identified ESATA-20, which has ~10-fold lower AHR agonism and 5-fold greater potency against prostate cancer patient-derived xenografts. CONCLUSION: This increased therapeuticindex nominates ESATA-20 as a lead candidate forclinical development as an orally active third generation quinoline-3-carboxamide analog thatretains its on-target ability to disrupt HDAC4/HIF-1α/MEF-2-dependent adaptive survival signaling in the compromisedtumor microenvironment found in mCRPC.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Masculino , Humanos , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Western Blotting , Línea Celular Tumoral , Microambiente Tumoral , Histona Desacetilasas/metabolismo , Proteínas Represoras/metabolismo
4.
J Clin Invest ; 132(23)2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36194476

RESUMEN

Testosterone is the canonical growth factor of prostate cancer but can paradoxically suppress its growth when present at supraphysiological levels. We have previously demonstrated that the cyclical administration of supraphysiological androgen (SPA), termed bipolar androgen therapy (BAT), can result in tumor regression and clinical benefit for patients with castration-resistant prostate cancer. However, predictors and mechanisms of response and resistance have been ill defined. Here, we show that growth inhibition of prostate cancer models by SPA required high androgen receptor (AR) activity and were driven in part by downregulation of MYC. Using matched sequential patient biopsies, we show that high pretreatment AR activity predicted downregulation of MYC, improved clinical response, and prolonged progression-free and overall survival for patients on BAT. BAT induced strong downregulation of AR in all patients, which is shown to be a primary mechanism of acquired resistance to SPA. Acquired resistance was overcome by alternating SPA with the AR inhibitor enzalutamide, which induced adaptive upregulation of AR and resensitized prostate cancer to SPA. This work identifies high AR activity as a predictive biomarker of response to BAT and supports a treatment paradigm for prostate cancer involving alternating between AR inhibition and activation.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Receptores Androgénicos , Masculino , Humanos , Receptores Androgénicos/metabolismo , Andrógenos/farmacología , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Antagonistas de Receptores Androgénicos/farmacología , Antagonistas de Receptores Androgénicos/uso terapéutico , Nitrilos , Testosterona/farmacología , Resistencia a Antineoplásicos , Línea Celular Tumoral
6.
JCI Insight ; 6(8)2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33724955

RESUMEN

Resistance to AR signaling inhibitors (ARSis) in a subset of metastatic castration-resistant prostate cancers (mCRPCs) occurs with the emergence of AR- neuroendocrine prostate cancer (NEPC) coupled with mutations/deletions in PTEN, TP53, and RB1 and the overexpression of DNMTs, EZH2, and/or SOX2. To resolve whether the lack of AR is the driving factor for the emergence of the NE phenotype, molecular, cell, and tumor biology analyses were performed on 23 xenografts derived from patients with PC, recapitulating the full spectrum of genetic alterations proposed to drive NE differentiation. Additionally, phenotypic response to CRISPR/Cas9-mediated AR KO in AR+ CRPC cells was evaluated. These analyses document that (a) ARSi-resistant NEPC developed without androgen deprivation treatment; (b) ARS in ARSi-resistant AR+/NE+ double-positive "amphicrine" mCRPCs did not suppress NE differentiation; (c) the lack of AR expression did not necessitate acquiring a NE phenotype, despite concomitant mutations/deletions in PTEN and TP53, and the loss of RB1 but occurred via emergence of an AR-/NE- double-negative PC (DNPC); (d) despite DNPC cells having homogeneous genetic driver mutations, they were phenotypically heterogeneous, expressing basal lineage markers alone or in combination with luminal lineage markers; and (e) AR loss was associated with AR promoter hypermethylation in NEPCs but not in DNPCs.


Asunto(s)
Carcinoma Neuroendocrino/genética , Transformación Celular Neoplásica/genética , Resistencia a Antineoplásicos/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Acetato de Abiraterona/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Benzamidas/uso terapéutico , Carcinoma Neuroendocrino/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Masculino , Ratones , Trasplante de Neoplasias , Nitrilos/uso terapéutico , Fosfohidrolasa PTEN/genética , Feniltiohidantoína/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/genética , Proteínas de Unión a Retinoblastoma/genética , Factores de Transcripción SOXB1/genética , Tiohidantoínas/uso terapéutico , Proteína p53 Supresora de Tumor/genética , Ubiquitina-Proteína Ligasas/genética
8.
Oncogene ; 39(45): 6935-6949, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32989253

RESUMEN

The role of truncated androgen receptor splice variant-7 (AR-V7) in prostate cancer biology is an unresolved question. Is it simply a marker of resistance to 2nd-generation androgen receptor signaling inhibitors (ARSi) like abiraterone acetate (Abi) and enzalutamide (Enza) or a functional driver of lethal resistance via its ligand-independent transcriptional activity? To resolve this question, the correlation between resistance to ARSi and genetic chances and expression of full length AR (AR-FL) vs. AR-V7 were evaluated in a series of independent patient-derived xenografts (PDXs). While all PDXs lack PTEN expression, there is no consistent requirement for mutation in TP53, RB1, BRCA2, PIK3CA, or MSH2, or expression of SOX2 or ERG and ARSi resistance. Elevated expression of AR-FL alone is sufficient for Abi but not Enza resistance, even if AR-FL is gain-of-function (GOF) mutated. Enza resistance is consistently correlated with enhanced AR-V7 expression. In vitro and in vivo growth responses of Abi-/Enza-resistant LNCaP-95 cells in which CRISPR-Cas9 was used to knockout AR-FL or AR-V7 alone or in combination were evaluated. Combining these growth responses with RNAseq analysis demonstrates that both AR-FL- and AR-V7-dependent transcriptional complementation are needed for Abi/Enza resistance.


Asunto(s)
Antagonistas de Andrógenos/farmacología , Resistencia a Antineoplásicos/genética , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Receptores Androgénicos/genética , Acetato de Abiraterona/farmacología , Acetato de Abiraterona/uso terapéutico , Anciano , Antagonistas de Andrógenos/uso terapéutico , Animales , Benzamidas , Biopsia , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Masculino , Ratones , Persona de Mediana Edad , Nitrilos , Feniltiohidantoína/análogos & derivados , Feniltiohidantoína/farmacología , Feniltiohidantoína/uso terapéutico , Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Empalme del ARN , RNA-Seq , Receptores Androgénicos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Mol Cancer Ther ; 19(11): 2353-2362, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32943549

RESUMEN

PRX302 is a highly potent, mutant bacterial pore-forming biologic protoxin engineered for selective activation by PSA, a serine protease expressed by benign and malignant prostate epithelial cells. Although being developed as a local therapy for benign prostatic hyperplasia and localized prostate cancer, PRX302 cannot be administered systemically as a treatment for metastatic disease due to binding to ubiquitously expressed glycosylphosphatidylinositol (GPI)-anchored proteins, which leads to poor accumulation within the tumor microenvironment. To overcome this limitation, poly-lactic-co-glycolic acid (PLGA) microparticles encapsulating the protoxin were developed, which are known to accumulate in the liver, a major site of metastasis for prostate cancer and other solid tumors. A highly sensitive and reproducible sandwich ELISA to quantify PRX302 released from microparticles was developed. Utilizing this assay, PRX302 release from different microparticle formulations was assessed over multiple days. Hemolysis assays documented PSA-dependent pore formation and lytic potential (i.e., function) of the released protoxin. MTT assays demonstrated that conditioned supernatant from PRX302-loaded, but not blank (i.e., unloaded), PLGA microparticles was highly cytotoxic to PC3 and DU145 human prostate cancer cells in the presence of exogenous PSA. Microparticle encapsulation prevented PRX302 from immediately interacting with GPI-anchored proteins as demonstrated in a competition assay, which resulted in an increased therapeutic index and significant antitumor efficacy following a single dose of PRX302-loaded microparticles in a preclinical model of prostate cancer liver metastasis with no obvious toxicity. These results document that PRX302 released from PLGA microparticles demonstrate in vivo antitumor efficacy in a clinically relevant preclinical model of metastatic prostate cancer.


Asunto(s)
Antineoplásicos/farmacología , Productos Biológicos/farmacología , Composición de Medicamentos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/secundario , Neoplasias de la Próstata Resistentes a la Castración/patología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Toxinas Bacterianas/metabolismo , Productos Biológicos/administración & dosificación , Productos Biológicos/química , Línea Celular Tumoral , Modelos Animales de Enfermedad , Composición de Medicamentos/métodos , Hemólisis/efectos de los fármacos , Humanos , Masculino , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Unión Proteica , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Oncotarget ; 7(44): 71298-71308, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27542256

RESUMEN

Accurate modeling of angiogenesis in vitro is essential for guiding the preclinical development of novel anti-angiogenic agents and treatment strategies. The formation of new blood vessels is a multifactorial and multi-stage process dependent upon paracrine factors produced by stromal cells in the local microenvironment. Mesenchymal stem cells (MSCs) are multipotent cells in adults that can be recruited to sites of inflammation and tissue damage where they aid in wound healing through regenerative, trophic, and immunomodulatory properties. Primary stromal cultures derived from human bone marrow, normal prostate, or prostate cancer tissue are highly enriched in MSCs and stromal progenitors. Using conditioned media from these primary cultures, a robust pro-angiogenic response was observed in a physiologically-relevant three-dimensional fibrin matrix assay. To evaluate the utility of this assay, the allosteric HDAC4 inhibitor tasquinimod and the anti-VEGF monoclonal antibody bevacizumab were used as model compounds with distinct mechanisms of action. While both agents had a profound inhibitory effect on endothelial sprouting, only bevacizumab induced significant regression of established vessels. Additionally, the pro-angiogenic properties of MSCs derived from prostate cancer patients provides further evidence that selective targeting of this population may be of therapeutic benefit.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Neovascularización Fisiológica/fisiología , Próstata/citología , Inhibidores de la Angiogénesis/farmacología , Bevacizumab/farmacología , Células Cultivadas , Fibrina , Humanos , Masculino , Quinolonas/farmacología , Nicho de Células Madre
11.
Oncotarget ; 5(18): 8093-106, 2014 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-25193858

RESUMEN

Tasquinimod, an orally active quinoline-3-carboxamide, binds with high affinity to HDAC4 and S100A9 in cancer and infiltrating host cells within compromised tumor microenvironment inhibiting adaptive survival pathways needed for an angiogenic response. Clinical trials document that as low as 0.5-1mg tasquinimod/day is therapeutic against castrate resistant metastatic prostate cancer. Tasquinimod is metabolized via cytochrome P4503A4, but ketoconazole at a dose which completely inhibits CYP3A metabolism does not affect tasquinimod's ability to inhibit endothelial "sprouting" in vitro or anti-cancer efficacy against human prostate cancer xenografts in vivo. Tasquinimod's potency is facilitated by its reversible binding (Kd < 35 µM) to the IIA subdomain of albumin (Sudlow's site I). As blood vessels within the compromised cancer microenvironment are characterized by a higher degree of leakiness than those in normal tissues, this results in an enhanced uptake of tasquinimod bound to albumin in cancer tissue via a tumor specific process known as the "enhanced permeability and retention" (i.e., EPR) effect. Thus, despite plasma levels of < 1 µM, the EPR effect results in intracellular drug concentrations of 2-3 µM, levels several-fold higher than needed for inhibition of endothelial sprouting (IC50 ~ 0.5 µM) or for inhibition of HDAC4 and S100A9 mediated tumor growth.


Asunto(s)
Inhibidores de la Angiogénesis/farmacocinética , Neoplasias de la Próstata Resistentes a la Castración/irrigación sanguínea , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Quinolinas/farmacocinética , Albúmina Sérica/metabolismo , Microambiente Tumoral , Administración Oral , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/sangre , Animales , Disponibilidad Biológica , Biotransformación , Permeabilidad Capilar , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Desnudos , Neovascularización Fisiológica/efectos de los fármacos , Orquiectomía , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Unión Proteica , Quinolinas/administración & dosificación , Quinolinas/sangre , Quinolonas , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Prostate ; 74(11): 1118-31, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24913829

RESUMEN

INTRODUCTION: Physiologic testosterone continuously stimulates prostate stromal cell secretion of paracrine growth factors (PGFs), which if unopposed would induce hyperplastic overgrowth of normal prostate epithelial cells (PrECs). METHODS: Lentiviral shRNA stable knock down of c-MYC, ß-catenin, or TCF-4 completely inhibits normal (i.e., non-transformed) human PrECs growth. c-MYC enhancer driven reporter expression and growth is inhibited by two chemically distinct molecules, which prevent ß-catenin signaling either by blocking TCF-4 binding (i.e., toxoflavin) or by stimulating degradation (i.e., AVX939). Recombinant DKK1 protein at a dose, which inhibits activation of canonical Wnt signaling does not inhibit PrEC growth. Nuclear ß-catenin translocation and PrEC growth is prevented by both lack of PGFs or Akt inhibitor-I. Growth inhibition induced by lack of PGFs, toxoflavin, or Akt inhibitor-I is overcome by constitutive c-MYC transcription. RESULTS: In the presence of continuous PGF signaling, PrEC hyperplasia is prevented by androgen binding to AR suppressing c-MYC transcription, resulting in G0 arrest/terminal differentiation independent of Rb, p21, p27, FoxP3, or down regulation of growth factors receptors and instead involves androgen-induced formation of AR/ß-catenin/TCF-4 complexes, which suppress c-MYC transcription. Such suppression does not occur when AR is mutated in its zinc-finger binding domain. DISCUSSION: Proliferation of non-transformed human PrECs is dependent upon c-MYC transcription via formation/binding of ß-catenin/TCF-4 complexes at both 5' and 3' c-MYC enhancers stimulated by Wnt-independent, PGF induced Akt signaling. In the presence of continuous PGF signaling, PrEC hyperplasia is prevented by androgen-induced formation of AR/ß-catenin/TCF-4 complexes, which retains binding to 3' c-MYC enhancer, but now suppresses c-MYC transcription.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Proliferación Celular , Células Epiteliales/patología , Próstata/patología , Proteínas Proto-Oncogénicas c-myc/fisiología , Receptores Androgénicos/fisiología , Factores de Transcripción/fisiología , beta Catenina/fisiología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/efectos de los fármacos , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Células Cultivadas , Factores de Transcripción Forkhead/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Masculino , Hiperplasia Prostática/prevención & control , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Proto-Oncogénicas c-myc/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Transducción de Señal/fisiología , Factor de Transcripción 4 , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Transcripción Genética/fisiología , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/efectos de los fármacos , beta Catenina/genética
13.
Cancer Res ; 73(4): 1386-99, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23149916

RESUMEN

Tasquinimod is an orally active antiangiogenic drug that is currently in phase III clinical trials for the treatment of castration-resistant prostate cancer. However, the target of this drug has remained unclear. In this study, we applied diverse strategies to identify the histone deacetylase HDAC4 as a target for the antiangiogenic activity of tasquinimod. Our comprehensive analysis revealed allosteric binding (Kd 10-30 nmol/L) to the regulatory Zn(2+) binding domain of HDAC4 that locks the protein in a conformation preventing HDAC4/N-CoR/HDAC3 complex formation. This binding inhibited colocalization of N-CoR/HDAC3, thereby inhibiting deacetylation of histones and HDAC4 client transcription factors, such as HIF-1α, which are bound at promoter/enhancers where epigenetic reprogramming is required for cancer cell survival and angiogenic response. Through this mechanism, tasquinimod is effective as a monotherapeutic agent against human prostate, breast, bladder, and colon tumor xenografts, where its efficacy could be further enhanced in combination with a targeted thapsigargin prodrug (G202) that selectively kills tumor endothelial cells. Together, our findings define a mechanism of action of tasquinimod and offer a perspective on how its clinical activity might be leveraged in combination with other drugs that target the tumor microenvironment. Cancer Res; 73(4); 1386-99. ©2012 AACR.


Asunto(s)
Histona Desacetilasas/metabolismo , Neoplasias/tratamiento farmacológico , Quinolinas/farmacología , Proteínas Represoras/metabolismo , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Acetilación/efectos de los fármacos , Regulación Alostérica , Animales , Western Blotting , Hipoxia de la Célula , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Histona Desacetilasas/química , Histona Desacetilasas/genética , Histonas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Desnudos , Modelos Moleculares , Neoplasias/genética , Neoplasias/patología , Profármacos/farmacología , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Quinolonas , Interferencia de ARN , Proteínas Represoras/química , Proteínas Represoras/genética , Tapsigargina/farmacología , Microambiente Tumoral/genética
14.
Prostate ; 72(6): 638-48, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21837778

RESUMEN

BACKGROUND: Tasquinimod is a novel inhibitor of tumor angiogenesis which enhances therapeutic efficacy when combined with androgen ablation and/or taxane-based chemotherapies in pre-clinical prostate cancer models. It has entered registration Phase III evaluation for the treatment of castration resistant prostate cancer. Since tasquinimod suppresses the angiogenic switch induced by tumor hypoxia as prostate cancers outgrow their blood supply, this raises the issue of whether tasquinimod also suppresses the angiogenic rebound induced by fractionated radiation thereby enhancing therapeutic response to fractionated radiation. METHODS: Human endothelial and prostate cancer cells in culture and human prostate cancer xenografts growing in castrated male nude mice were evaluated for their response to radiation alone and in combination with tasquinimod. RESULTS: At clinically relevant drug levels, tasquinimod significantly (P < 0.05) enhances anti-cancer efficacy of fractionated radiation with optimal timing for initiating daily tasquinimod treatment being after completion of the fractionated radiation. CONCLUSIONS: Based upon cell culture studies and tumor tissue oxygenation (i.e., pO(2)), tumor vascular volume, and tumor blood vessel density measurements, the mechanism for such enhancement and optimal timing involves tasquinimod's ability to prevent the angiogenic rebound induced by fractionated radiation.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neovascularización Patológica/terapia , Neoplasias de la Próstata/terapia , Quinolinas/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Animales , Terapia Combinada , Humanos , Masculino , Ratones , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/radioterapia , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/radioterapia , Quinolonas , Trasplante Heterólogo , Células Tumorales Cultivadas
15.
PLoS One ; 5(7): e11475, 2010 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-20628607

RESUMEN

The conversion of androgen receptor (AR) signaling as a mechanism of growth suppression of normal prostate epithelial cells to that of growth stimulation in prostate cancer cells is often associated with AR mutation, amplification and over-expression. Thus, down-regulation of AR signaling is commonly therapeutic for prostate cancer. The E006AA cell line was established from a hormone naïve, localized prostate cancer. E006AA cells are genetically aneuploid and grow equally well when xenografted into either intact or castrated male NOG but not nude mice. These cells exhibit: 1) X chromosome duplication and AR gene amplification, although paradoxically not coupled with increased AR expression, and 2) somatic, dominant-negative Serine-599-Glycine loss-of-function mutation within the dimerization surface of the DNA binding domain of the AR gene. No effect on the growth of E006AA cells is observed using targeted knockdown of endogenous mutant AR, ectopic expression of wild-type AR, or treatment with androgens or anti-androgens. E006AA cells represent a prototype for a newly identified subtype of prostate cancer cells that exhibit a dominant-negative AR loss-of-function in a hormonally naïve patient. Such loss-of-function eliminates AR-mediated growth suppression normally induced by normal physiological levels of androgens, thus producing a selective growth advantage for these malignant cells in hormonally naïve patients. These data highlight that loss of AR-mediated growth suppression is an independent process, and that, without additional changes, is insufficient for acquiring oncogene addiction to AR signaling. Thus, patients with prostate cancer cells harboring such AR loss-of-function mutations will not benefit from aggressive hormone or anti-AR therapies even though they express AR protein.


Asunto(s)
Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Andrógenos/farmacología , Animales , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Hibridación Fluorescente in Situ , Lentivirus/genética , Masculino , Ratones , Ratones SCID , Mutación , Reacción en Cadena de la Polimerasa , Neoplasias de la Próstata/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/fisiología , Receptores Androgénicos/genética , Análisis de Secuencia de ADN
16.
Mol Cancer Ther ; 8(5): 1340-9, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19417145

RESUMEN

There are quantitative and/or qualitative mechanisms allowing androgen receptor (AR) growth signaling in androgen ablation refractory prostate cancer cells. Regardless of the mechanism, agents that deplete AR protein expression prevent such AR growth signaling. Thapsigargin (TG) is a highly cell-penetrant sequiterpene-lactone that once inside cells inhibits (IC(50), ∼ 10 nmol/L) critically important housekeeping SERCA 2b calcium pumps in the endoplasmic reticulum. Using a series of five genetically diverse androgen ablation refractory human prostate cancer lines (LNCaP, LAPC-4, VCaP, MDA-PCa-2b, and CWR22Rv1), TG inhibition of SERCA pumps consistently results in depletion of the endoplasmic reticulum Ca(+2) coupled with µmol/L elevation in the intracellular free Ca(+2) initiating a molecular cascade that: (a) inhibits Cap-dependent AR protein synthesis resulting in 90% depletion of AR protein by 24 hours of TG exposure, (b) arrests the cells in G(0), and (c) induces their apoptotic death. Unfortunately, due to its highly lipophilic nature, TG is not deliverable as a systemic agent without host toxicity. Therefore, TG analogues containing amino acids were developed, which retain ability to deplete AR protein and induce cell death and which can be covalently linked to peptide carriers producing water soluble prodrugs for systemic delivery. Specific amino acid sequences are used to restrict the liberation of cytotoxic amino acid containing TG analogues from the peptide prodrug by prostate-specific proteases, such as prostate-specific antigen and prostate-specific membrane antigen, or cancer-specific proteases, such as fibroblast activation protein, so that toxicity of these prodrugs is selectively targeted to metastatic sites of prostate cancer. Based on these results, these prodrugs are undergoing clinical development.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Tapsigargina/farmacología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Calcio/metabolismo , Proteínas de Ciclo Celular , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Retículo Endoplásmico/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Conejos , Receptores Androgénicos/genética , Transducción de Señal/efectos de los fármacos , Tapsigargina/química , Factores de Transcripción/metabolismo
17.
J Nucl Med ; 50(5): 774-80, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19372481

RESUMEN

UNLABELLED: Apoptosis is a key mechanism in numerous pathologies. However, there are no effective noninvasive means available for an early detection and quantitative assessment of evolution dynamics of the apoptotic process. Here, we have characterized the ability of the novel PET voltage sensor (18)F-fluorobenzyl triphenyl phosphonium ((18)F-FBnTP) to quantify the time-dependent apoptotic action of the taxanes paclitaxel and docetaxel in vitro and in vivo. METHODS: The duration-dependent treatment effect of paclitaxel on (18)F-FBnTP uptake was assayed in human MDA-MB-231 breast carcinoma cells. The expression of the proapoptotic Bax and antiapoptotic Bcl-2 mitochondrial proteins, release of the apoptogen cytochrome c, and activation of executioner caspase-3 were determined by Western blotting. The fraction of viable cells was determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide. The effect of docetaxel on (18)F-FBnTP and (18)F-FDG uptake in orthotopic prostate tumors in mice was compared. RESULTS: (18)F-FBnTP cellular uptake in viable cells declined linearly with the increasing duration of paclitaxel treatment, from 3 to 24 h, and plateaued at 48 h. The extent of decrease of (18)F-FBnTP correlated strongly with the Bax-to-Bcl-2 ratio (R(2) = 0.83) and release of cytochrome c (R(2) = 0.92), but preceded in time the caspase-3 cleavage. The P-glycoprotein blocker verapamil did not interfere with (18)F-FBnTP cellular uptake. (18)F-FBnTP prostate tumor contrast was greater than (18)F-FDG prostate tumor contrast. Docetaxel caused a marked decrease (52.4%) of (18)F-FBnTP tumor uptake, within 48 h, whereas (18)F-FDG was much less affected (12%). CONCLUSION: The voltage sensor (18)F-FBnTP is a viable means for quantification of paclitaxel pharmacodynamics. (18)F-FBnTP permits the detection of paclitaxel apoptotic action in vivo earlier than does (18)F-FDG. (18)F-FBnTP may afford a novel approach for early detection and quantitative assessment of the cumulative-effect kinetics of proapoptotic drugs and conditions using PET.


Asunto(s)
Apoptosis/fisiología , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/fisiopatología , Potencial de la Membrana Mitocondrial , Compuestos Organofosforados , Tomografía de Emisión de Positrones/métodos , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Citocromos c/metabolismo , Docetaxel , Genes bcl-2 , Humanos , Paclitaxel/farmacología , Radiofármacos , Taxoides/farmacología , Moduladores de Tubulina/farmacología , Proteína X Asociada a bcl-2/metabolismo
18.
Prostate ; 67(7): 790-7, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17373719

RESUMEN

PURPOSE: Tasquinimod is a second-generation orally active quinoline-3-carboxamide analog with enhanced potency against prostate cancer via its anti-angiogenic activity. It is presently undergoing clinical trials. Androgen ablation and taxanes are standard therapies for metastatic prostate cancer. This raises the issue of whether combining tasquinimod with either of these approaches enhances therapeutic efficacy. EXPERIMENTAL DESIGN: The tumor growth of a series of human prostate cancer xenografts (CWR-22Rv1, CWR-22R-H, LAPC-4, LNCaP, PC-3 and DU-145) in male nude mice given nothing versus tasquinimod alone or in combination with androgen ablation or with androgen ablation plus taxotere were evaluated as model systems to resolve these issues. RESULTS: These studies documented that daily oral treatment with tasquinimod consistently, statistically (P < 0.05) inhibited the tumor growth of each of the xenografts in a dose-dependent manner via an anti-angiogenic response as monitored by a significant (P < 0.05) decrease in the tumor blood vessel density. Tasquinimod's anti-prostate cancer efficacy is enhanced when combined with androgen ablation and this enhancement was observed even when androgen ablation was either subsequent to or proceeded by tasquinimod treatment. In addition, tasquinimod also enhanced the tumor growth inhibition and survival when combined with androgen ablation plus taxotere. Companion studies documented that tasquinimod has no direct effect on serum PSA in these xenografts. CONCLUSIONS: These results documented that differences in serum PSA in tasquinimod-treated hosts are related to inhibition in tumor growth. This suggests that in clinical trials, changes in PSA slope or doubling time are indicative of therapeutic response to tasquinimod.


Asunto(s)
Andrógenos/fisiología , Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/uso terapéutico , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/tratamiento farmacológico , Quinolinas/uso terapéutico , Taxoides/uso terapéutico , Animales , Línea Celular Tumoral , Docetaxel , Sinergismo Farmacológico , Quimioterapia Combinada , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/irrigación sanguínea , Quinolonas , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Cancer Res ; 66(17): 8598-607, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16951173

RESUMEN

Stage-specific differentiation markers were used to evaluate the cellular composition and the origin of nonimmortalized (PrEC) and immortalized (PZ-HPV7, CA-HPV10, RWPE-1, and 957E/hTERT) human prostate cell lines. These studies documented that immortalized and nonimmortalized prostate epithelial cells established and maintained in low (i.e., <300 micromol/L) Ca(2+) serum-free defined (SFD) medium were all derived from normal nonmalignant prostate tissues and contain CD133(+)/ABCG2(+)/alpha(2)beta(1)(Hi)/p63(-)/PSCA(-)/AR(-)/PSA(-) prostate stem cells. In these cultures, prostate stem cells are able to self-renew and generate two distinct cell lineages: the minor proliferatively quiescent neuroendocrine lineage and the major transit-amplifying cell lineage. Subsequently, CD133(-)/ABCG2(-)/alpha(2)beta(1)(Hi)/p63(+)/PSCA(-)/AR(-)/PSA(-) transit-amplifying cells proliferate frequently and eventually mature into proliferatively quiescent CD133(-)/ABCG2(-)/alpha(2)beta(1)(Lo)/p63(-)/PSCA(+)/AR(-)/PSA(-) intermediate cells. Such proliferatively quiescent intermediate cells, however, do not complete their full maturation into CD133(-)/ABCG2(-)/alpha(2)beta(1)(Lo)/p63(-)/PSCA(-)/AR(+)/PSA(+) luminal-secretory cells in low Ca(2+) SFD medium. Addition of universal type I IFN and synthetic androgen (R1881) to culture medium resulted in up-regulation of androgen receptor protein expression. However, it failed to induce full differentiation of intermediate cells into AR(+)/PSA(+) luminal-secretory cells. Our results indicate that such inability of prostate epithelial cells to complete their differentiation is due to continuous expression of Notch-1 receptor and its downstream effector, Hey-1 protein, which actively suppresses differentiation via its ability to transcriptionally repress a series of genes, including the GATA family of transcription factors.


Asunto(s)
Calcio/farmacología , Células Epiteliales/citología , Próstata/citología , Células Madre/citología , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Medio de Cultivo Libre de Suero , Células Epiteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Metribolona/farmacología , Próstata/efectos de los fármacos , Antígeno Prostático Específico/genética , Receptores Androgénicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/efectos de los fármacos
20.
Prostate ; 66(16): 1768-78, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16955399

RESUMEN

BACKGROUND: Linomide, Figure 1, produces robust and consistent in vivo growth inhibition of prostate cancer models via its anti-angiogenic activity and inhibition of autoimmune encephalomyelitis models of multiple sclerosis (MS). MS clinical trials were discontinued because of unacceptable toxicity, due to dose-dependent induction of proinflammation. METHODS: Therefore, linomide analogs were initially screened to determine their in vivo potency to inhibit growth of the Dunning R-3327 AT-1 rat prostate cancer model in rats and their potency to inhibit angiogenesis in a Matrigel assay in mice. RESULTS: Based upon its superior potency (i.e., 30- to 60-fold more potent than linomide) in these assays and its lack of a proinflammation in the Beagle-dog, ABR-215050 (tasquinimod), Figure 1, was characterized for dose-response ability to inhibit the growth of a series of four additional human and rodent prostate cancer models in mice. Pharmacokinetic analysis following oral dosing documented that blood and tumor tissue levels of ABR-215050 as low as 0.5-1 microM are therapeutically effective. This efficacy is correlated with inhibition of angiogenesis in a variety of assays (endothelial capillary tube formation, aortic ring assay, chorioallantoic membrane assay, real-time tumor blood flow and PO(2) measurements, tumor blood vessel density, and tumor hypoxic and apoptotic fractions). CONCLUSIONS: Based upon its robust and consistent anti-angiogenic activity and thus tumor growth, ABR-215050 has entered clinical trials for the treatment of prostate cancer.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/tratamiento farmacológico , Quinolinas/farmacología , Animales , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Perros , Humanos , Hidroxiquinolinas/farmacología , Masculino , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neoplasias de la Próstata/patología , Quinolonas , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA