Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Biomed Chromatogr ; 38(1): e5754, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37750452

RESUMEN

Despite aggressive treatment approaches, the overall survival of glioblastoma (GBM) patients remained poor with a strong need for more effective chemotherapeutic agents. A previous study has shown that ARN14988 is more cytotoxic to GBM cells compared to US Food and Drug Administration-approved temozolomide. This finding makes ARN14988 a desirable candidate for further pharmacological assessment. Therefore, an efficient analytical method is needed to quantify ARN14988. Herein, we have developed and validated sample preparation and LC-MS/MS triple quadrupole (QQQ) method for quantification of ARN14988 in mouse plasma. In this method, the liquid-liquid extraction of ARN14988 from mouse plasma was performed using 5% ethyl acetate in hexane. The chromatographic separation was achieved using a C18 -column with mobile phases of 10 mm ammonium acetate (pH 5) and 0.1% formic acid in methanol, within a runtime of 10 min. The monitored transitions were m/z 391.20 → m/z 147.00 for ARN14988, and m/z 455.30 → m/z 165.00 for verapamil (internal standard) in positive electrospray ionization. The developed method for ARN14988 showed linearity over the range of 10-5,000 ng/ml (r2 > 0.99). The selectivity, sensitivity, matrix effect, recovery, stability, inter-day and intraday accuracy and precision were determined using four quality control samples. This validated method was successfully applied to the pharmacokinetic study of ARN14988 in mice.


Asunto(s)
Antineoplásicos , Cromatografía Líquida con Espectrometría de Masas , Animales , Ratones , Ceramidasa Ácida , Cromatografía Liquida/métodos , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem/métodos
2.
J Med Chem ; 65(8): 6039-6055, 2022 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-35404047

RESUMEN

Vitamin-D receptor (VDR) mRNA is overexpressed in neuroblastoma and carcinomas of lung, pancreas, and ovaries and predicts poor prognoses. VDR antagonists may be able to inhibit tumors that overexpress VDR. However, the current antagonists are arduous to synthesize and are only partial antagonists, limiting their use. Here, we show that the VDR antagonist MeTC7 (5), which can be synthesized from 7-dehydrocholesterol (6) in two steps, inhibits VDR selectively, suppresses the viability of cancer cell-lines, and reduces the growth of the spontaneous transgenic TH-MYCN neuroblastoma and xenografts in vivo. The VDR selectivity of 5 against RXRα and PPAR-γ was confirmed, and docking studies using VDR-LBD indicated that 5 induces major changes in the binding motifs, which potentially result in VDR antagonistic effects. These data highlight the therapeutic benefits of targeting VDR for the treatment of malignancies and demonstrate the creation of selective VDR antagonists that are easy to synthesize.


Asunto(s)
Neuroblastoma , Receptores de Calcitriol , Animales , Animales Modificados Genéticamente , Xenoinjertos , Humanos , Receptores de Calcitriol/antagonistas & inhibidores , Receptores de Calcitriol/metabolismo , Vitaminas
3.
J Med Chem ; 65(4): 3388-3403, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35133171

RESUMEN

Histone acetylation is a prominent epigenetic modification linked to the memory loss symptoms associated with neurodegenerative disease. The use of existing histone deacetylase inhibitor (HDACi) drugs for treatment is precluded by their weak blood-brain barrier (BBB) permeability and undesirable toxicity. Here, we address these shortcomings by developing a new class of disulfide-based compounds, inspired by the scaffold of the FDA-approved HDACi romidepsin (FK288). Our findings indicate that our novel compound MJM-1 increases the overall level of histone 3 (H3) acetylation in a prostate cancer cell line. In mice, MJM-1 injected intraperitoneally (i.p.) crossed the BBB and could be detected in the hippocampus, a brain region that mediates memory. Consistent with this finding, we found that the post-training i.p. administration of MJM-1 enhanced hippocampus-dependent spatial memory consolidation in male mice. Therefore, MJM-1 represents a potential lead for further optimization as a therapeutic strategy for ameliorating cognitive deficits in aging and neurodegenerative diseases.


Asunto(s)
Encéfalo/metabolismo , Inhibidores de Histona Desacetilasas/síntesis química , Memoria Espacial/efectos de los fármacos , Animales , Línea Celular Tumoral , Inhibidores de Histona Desacetilasas/farmacocinética , Inhibidores de Histona Desacetilasas/farmacología , Ratones , Ratones Endogámicos BALB C
4.
ACS Pharmacol Transl Sci ; 4(2): 687-702, 2021 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-33860194

RESUMEN

Triple-negative breast cancer (TNBC) has limited treatment options and the worst prognosis among all types of breast cancer. We describe two prodrugs, namely, CWB-20145 (1) and its methyl analogue FAN-NM-CH3 (2) that reduced the size of TNBC-derived tumors. The DNA cross-linking of nitrogen mustard prodrugs 1 and 2 was superior to that of chlorambucil and melphalan once activated in the presence of H2O2. The cellular toxicity of 1 and 2 was demonstrated in seven human cancer cell lines. The TNBC cell line MDA-MB-468 was particularly sensitive toward 1 and 2. Compound 2 was 10 times more cytotoxic than chlorambucil and 16 times more active than melphalan. An evaluation of the gene expression demonstrated an upregulation of the tumor suppressor genes p53 and p21 supporting a transcriptional mechanism of a reduced tumor growth. Pharmacokinetic studies with 1 showed a rapid conversion of the prodrug. The introduction of a methyl group generated 2 with an increased half-life. An in vivo toxicity study in mice demonstrated that both prodrugs were less toxic than chlorambucil. Compounds 1 and 2 reduced tumor growth with an inhibition rate of more than 90% in athymic nude mice xenografted with MDA-MB-468 cells. Together, the in vivo investigations demonstrated that treatment with 1 and 2 suppressed tumor growth without affecting normal tissues in mice. These phenylboronic acid nitrogen mustard prodrugs represent promising drug candidates for the treatment of TNBC. However, the mechanisms underlying their superior in vivo activity and selectivity as well as the correlation between H2O2 level and in vivo efficacy are not yet fully understood.

5.
ACS Med Chem Lett ; 10(1): 121-126, 2019 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-30655958

RESUMEN

A novel class of bivalent ligands targeting putative protease-activated receptor (PAR) heteromers has been prepared based upon reported antagonists for the subtypes PAR1 and PAR2. Modified versions of the PAR1 antagonist RWJ-58259 containing alkyne adapters were connected via cycloaddition reactions to azide-capped polyethylene glycol (PEG) spacers attached to imidazopyridazine-based PAR2 antagonists. Initial studies of the PAR1-PAR2 antagonists indicated that they inhibited G alpha q-mediated calcium mobilization in endothelial and cancer cells driven by both PAR1 and PAR2 agonists. Compounds of this novel class hold promise for the prevention of restenosis, cancer cell metastasis, and other proliferative disorders.

6.
Mol Cell Endocrinol ; 485: 1-8, 2019 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-30654005

RESUMEN

In recent decades, the majority of ligands developed for the vitamin D receptor (VDR) bind at its deeply buried genomic ligand binding pocket. Theses ligands can be categorized into agonists and partial agonists/antagonists. A limited number of ligands, most of them peptides, bind the VDR‒coactivator binding site that is formed in the presence of an agonist and inhibit coactivator recruitment, and therefore transcription. Another solvent exposed VDR‒ligand binding pocket was identified for lithocholic acid, improving the overall stability of the VDR complex. Additional proposed interactions with VDR are discussed herein that include the alternative VDR‒ligand binding pocket that may mediate both non-genomic cellular responses and binding function 3 that was identified for the androgen receptor. Many VDR ligands increase blood calcium levels at therapeutic concentrations in vivo, thus the identification of alternative VDR‒ligand binding pockets might be crucial to develop non-calcemic and potent ligands for VDR to treat cancer and inflammatory disease.


Asunto(s)
Péptidos/química , Péptidos/farmacología , Receptores de Calcitriol/química , Receptores de Calcitriol/metabolismo , Animales , Sitios de Unión , Calcio/sangre , Humanos , Ligandos , Modelos Moleculares , Unión Proteica , Estructura Terciaria de Proteína , Relación Estructura-Actividad
7.
J Med Chem ; 61(20): 9132-9145, 2018 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-30247905

RESUMEN

We describe several new aromatic nitrogen mustards with various aromatic substituents and boronic esters that can be activated with H2O2 to efficiently cross-link DNA. In vitro studies demonstrated the anticancer potential of these compounds at lower concentrations than those of other clinically used chemotherapeutics, such as melphalan and chlorambucil. In particular, compound 10, bearing an amino acid ester chain, is selectively cytotoxic toward breast cancer and leukemia cells that have inherently high levels of reactive oxygen species. Importantly, 10 was 10-14-fold more efficacious than melphalan and chlorambucil for triple-negative breast-cancer (TNBC) cells. Similarly, 10 is more toxic toward primary chronic-lymphocytic-leukemia cells than either chlorambucil or the lead compound, 9. The introduction of an amino acid side chain improved the solubility and permeability of 10. Furthermore, 10 inhibited the growth of TNBC tumors in xenografted mice without obvious signs of general toxicity, making this compound an ideal drug candidate for clinical development.


Asunto(s)
Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/farmacología , Diseño de Fármacos , Peróxido de Hidrógeno/metabolismo , Compuestos de Mostaza Nitrogenada/metabolismo , Compuestos de Mostaza Nitrogenada/farmacología , Antineoplásicos Alquilantes/química , Ácidos Borónicos/química , Línea Celular Tumoral , Ésteres/química , Humanos , Compuestos de Mostaza Nitrogenada/química
8.
Mol Pharm ; 15(5): 1766-1777, 2018 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-29578347

RESUMEN

We describe lead compound MIDD0301 for the oral treatment of asthma based on previously developed positive allosteric α5ß3γ2 selective GABAA receptor (GABAAR) ligands. MIDD0301 relaxed airway smooth muscle at single micromolar concentrations as demonstrated with ex vivo guinea pig tracheal rings. MIDD0301 also attenuated airway hyperresponsiveness (AHR) in an ovalbumin murine model of asthma by oral administration. Reduced numbers of eosinophils and macrophages were observed in mouse bronchoalveolar lavage fluid without changing mucous metaplasia. Importantly, lung cytokine expression of IL-17A, IL-4, and TNF-α were reduced for MIDD0301-treated mice without changing antiinflammatory cytokine IL-10 levels. Automated patch clamp confirmed amplification of GABA induced current mediated by α1-3,5ß3γ2 GABAARs in the presence of MIDD0301. Pharmacodynamically, transmembrane currents of ex vivo CD4+ T cells from asthmatic mice were potentiated by MIDD0301 in the presence of GABA. The number of CD4+ T cells observed in the lung of MIDD0301-treated mice were reduced by an oral treatment of 20 mg/kg b.i.d. for 5 days. A half-life of almost 14 h was demonstrated by pharmacokinetic studies (PK) with no adverse CNS effects when treated mice were subjected to sensorimotor studies using the rotarod. PK studies also confirmed very low brain distribution. In conclusion, MIDD0301 represents a safe and improved oral asthma drug candidate that relaxes airway smooth muscle and attenuates inflammation in the lung leading to a reduction of AHR at a dosage lower than earlier reported GABAAR ligands.


Asunto(s)
Asma/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Pulmón/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Receptores de GABA-A/metabolismo , Animales , Asma/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Líquido del Lavado Bronquioalveolar/química , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Constricción , Citocinas/metabolismo , Eosinófilos/efectos de los fármacos , Eosinófilos/metabolismo , Femenino , Cobayas , Inflamación/metabolismo , Ligandos , Pulmón/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Músculo Liso/metabolismo , Ovalbúmina/metabolismo , Hipersensibilidad Respiratoria/metabolismo
9.
Eur J Med Chem ; 133: 197-207, 2017 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-28388522

RESUMEN

Quinone methide (QM) formation induced by endogenously generated H2O2 is attractive for biological and biomedical applications. To overcome current limitations due to low biological activity of H2O2-activated QM precursors, we are introducing herein several new arylboronates with electron donating substituents at different positions of benzene ring and/or different neutral leaving groups. The reaction rate of the arylboronate esters with H2O2 and subsequent bisquinone methides formation and DNA cross-linking was accelerated with the application of Br as a leaving group instead of acetoxy groups. Additionally, a donating group placed meta to the nascent exo-methylene group of the quinone methide greatly improves H2O2-induced DNA interstrand cross-link formation as well as enhances the cellular activity. Multiple donating groups decrease the stability and DNA cross-linking capability, which lead to low cellular activity. A cell-based screen demonstrated that compounds 2a and 5a with a OMe or OH group dramatically inhibited the growth of various tissue-derived cancer cells while normal cells were less affected. Induction of H2AX phosphorylation by these compounds in CLL lymphocytes provide evidence for a correlation between cell death and DNA damage. The compounds presented herein showed potent anticancer activities and selectivity, which represent a novel scaffold for anticancer drug development.


Asunto(s)
Antineoplásicos/farmacología , Derivados del Benceno/farmacología , ADN/química , Peróxido de Hidrógeno/metabolismo , Indolquinonas/farmacología , Sustancias Intercalantes/farmacología , Antineoplásicos/química , Antineoplásicos/metabolismo , Secuencia de Bases/efectos de los fármacos , Derivados del Benceno/química , Derivados del Benceno/metabolismo , Ácidos Borónicos/química , Ácidos Borónicos/metabolismo , Ácidos Borónicos/farmacología , Línea Celular Tumoral , Humanos , Indolquinonas/química , Indolquinonas/metabolismo , Sustancias Intercalantes/química , Sustancias Intercalantes/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo
10.
Mol Pharm ; 13(6): 2026-38, 2016 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-27120014

RESUMEN

Recent studies have demonstrated that subtype-selective GABAA receptor modulators are able to relax precontracted human airway smooth muscle ex vivo and reduce airway hyper-responsiveness in mice upon aerosol administration. Our goal in this study was to investigate systemic administration of subtype-selective GABAA receptor modulators to alleviate bronchoconstriction in a mouse model of asthma. Expression of GABAA receptor subunits was identified in mouse lungs, and the effects of α4-subunit-selective GABAAR modulators, XHE-III-74EE and its metabolite XHE-III-74A, were investigated in a murine model of asthma (ovalbumin sensitized and challenged BALB/c mice). We observed that chronic treatment with XHE-III-74EE significantly reduced airway hyper-responsiveness. In addition, acute treatment with XHE-III-74A but not XHE-III-74EE decreased airway eosinophilia. Immune suppressive activity was also shown in activated human T-cells with a reduction in IL-2 expression and intracellular calcium concentrations [Ca(2+)]i in the presence of GABA or XHE-III-74A, whereas XHE-III-74EE showed only partial reduction of [Ca(2+)]i and no inhibition of IL-2 secretion. However, both compounds significantly relaxed precontracted tracheal rings ex vivo. Overall, we conclude that the systemic delivery of a α4-subunit-selective GABAAR modulator shows good potential for a novel asthma therapy; however, the pharmacokinetic properties of this class of drug candidates have to be improved to enable better beneficial systemic pharmacodynamic effects.


Asunto(s)
Asma/tratamiento farmacológico , Benzodiazepinas/farmacología , Receptores de GABA-A/metabolismo , Animales , Asma/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Cobayas , Humanos , Interleucina-2/metabolismo , Células Jurkat , Masculino , Ratones , Ratones Endogámicos BALB C , Músculo Liso/efectos de los fármacos , Músculo Liso/metabolismo , Ovalbúmina/administración & dosificación , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Xenopus laevis
11.
Eur J Med Chem ; 109: 238-46, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26774929

RESUMEN

A systematic study with phase 1 and phase 2 metabolites of cholesterol and vitamin D was conducted to determine whether their biological activity is mediated by the vitamin D receptor (VDR). The investigation necessitated the development of novel synthetic routes for lithocholic acid (LCA) glucuronides (Gluc). Biochemical and cell-based assays were used to demonstrate that hydroxylated LCA analogs were not able to bind VDR. This excludes VDR from mediating their biological and pharmacological activities. Among the synthesized LCA conjugates a novel VDR agonist was identified. LCA Gluc II increased the expression of CYP24A1 in DU145 cancer cells especially in the presence of the endogenous VDR ligand 1,25(OH)2D3. Furthermore, the methyl ester of LCA was identified as novel VDR antagonist. For the first time, we showed that calcitroic acid, the assumed inactive final metabolite of vitamin D, was able to activate VDR-mediated transcription to a higher magnitude than bile acid LCA. Due to a higher metabolic stability in comparison to vitamin D, a very low toxicity, and high concentration in bile and intestine, calcitroic acid is likely to be an important mediator of the protective vitamin D properties against colon cancer.


Asunto(s)
Calcitriol/análogos & derivados , Colesterol/metabolismo , Glucuronatos/farmacología , Ácido Litocólico/farmacología , Receptores de Calcitriol/metabolismo , Transcripción Genética/efectos de los fármacos , Vitamina D/metabolismo , Calcitriol/síntesis química , Calcitriol/química , Calcitriol/farmacología , Línea Celular Tumoral , Glucuronatos/síntesis química , Glucuronatos/química , Humanos , Ácido Litocólico/síntesis química , Ácido Litocólico/química , Masculino , Neoplasias de la Próstata/metabolismo , Receptores de Calcitriol/agonistas , Vitamina D3 24-Hidroxilasa/metabolismo
12.
Anticancer Res ; 35(11): 6001-7, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26504023

RESUMEN

AIM: To investigate the in vivo effects of 3-indolylmethanamines 31B and PS121912 in treating ovarian cancer and leukemia, respectively. MATERIALS AND METHODS: Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) and western blotting were applied to demonstrate the induction of apoptosis. Xenografted mice were investigated to show the antitumor effects of 3-indolylmethanamines. (13)C-Nuclear magnetic resource (NMR) and western blotting were used to demonstrate inhibition of glucose metabolism. RESULTS: 31B inhibited ovarian cancer cell proliferation and activated caspase-3, cleaved poly (ADP-ribose) polymerase 1 (PARP1), and phosphorylated mitogen-activated protein kinases (MAPK), JUN N-terminal kinase/stress-activated protein kinase (JNK/SAPK) and p38. 31B reduced ovarian cancer xenograft tumor growth and PS121912 inhibited the growth of HL-60-derived xenografts without any sign of toxicity. Compound 31B inhibited de novo glycolysis and lipogenesis mediated by the reduction of fatty acid synthase and lactate dehydrogenase-A expression. CONCLUSION: 3-Indolylmethanamines represent a new class of antitumor agents. We have shown for the first time the in vivo anticancer effects of 3-indolylmethanamines 31B and PS121912.


Asunto(s)
Aminas/química , Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Hidrocarburos Aromáticos/farmacología , Indoles/química , Neoplasias Ováricas/tratamiento farmacológico , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Femenino , Citometría de Flujo , Humanos , Indoles/farmacología , Espectroscopía de Resonancia Magnética , Ratones , Ratones Desnudos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Artículo en Inglés | MEDLINE | ID: mdl-25419525

RESUMEN

Herein, we described the development of two virtual screens to identify new vitamin D receptor (VDR) antagonists among nuclear receptor (NR) ligands. Therefore, a database of 14330 nuclear receptor ligands and their NR affinities was assembled using the online available "Binding Database". Two different virtual screens were carried out in conjunction with a reported VDR crystal structure applying a stringent and less stringent pharmacophore model to filter docked NR ligand conformations. The pharmacophore models were based on the spatial orientation of the hydroxyl functionalities of VDR's natural ligands 1,25(OH2)D3 and 25(OH2)D3. The first virtual screen identified 32 NR ligands with a calculate free energy of VDR binding of more than -6.0 kJ/mol. All but nordihydroguaiaretic acid (NDGA) are VDR ligands, which inhibited the interaction between VDR and coactivator peptide SRC2-3 with an IC50 value of 15.8 µM. The second screen identified 162 NR ligands with a calculate free energy of VDR binding of more than -6.0 kJ/mol. More than half of these ligands were developed to bind VDR followed by ERα/ß ligands (26%), TRα/ß ligands (7%) and LxRα/ß ligands (7%). The binding between VDR and ERα ligand H6036 as well as TRα/ß ligand triiodothyronine and a homoserine analog thereof was confirmed by fluorescence polarization.

14.
Cancer Chemother Pharmacol ; 74(4): 787-98, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25107568

RESUMEN

PURPOSE: PS121912 has been developed as selective vitamin D receptor (VDR)-coregulator inhibitor starting from a high throughput screening campaign to identify new agents that modulate VDR without causing hypercalcemia. Initial antiproliferative effects of PS121912 were observed that are characterized herein to enable future in vivo investigation with this molecule. METHODS: Antiproliferation and apoptosis were determined using four different cancer cell lines (DU145, Caco2, HL-60 and SKOV3) in the presence of PS121912, 1,25-(OH)2D3, or a combination of 1,25-(OH)2D3 and PS121912. VDR si-RNA was used to identify the role of VDR during this process. The application of ChIP enabled us to determine the involvement of coregulator recruitment during transcription, which was investigated by RT-PCR with VDR target genes and those affiliated with cell cycle progression. Translational changes of apoptotic proteins were determined with an antibody array. The preclinical characterization of PS121912 includes the determination of metabolic stability and CYP3A4 inhibition. RESULTS: PS121912 induced apoptosis in all four cancer cells, with HL-60 cells being the most sensitive. At sub-micromolar concentrations, PS121912 amplified the growth inhibition of cancer cells caused by 1,25-(OH)2D3 without being antiproliferative by itself. A knockout study with VDR si-RNA confirmed the mediating role of VDR. VDR target genes induced by 1,25-(OH)2D3 were down-regulated with the co-treatment of PS121912. This process was highly dependent on the recruitment of coregulators that in case of CYP24A1 was SRC2. The combination of PS121912 and 1,25-(OH)2D3 reduced the presence of SRC2 and enriched the occupancy of corepressor NCoR at the promoter site. E2F transcription factors 1 and 4 were down-regulated in the presence of PS121912 and 1,25-(OH)2D3 that in turn reduced the transcription levels of cyclin A and D, thus arresting HL-60 cells in the S or G2/M phase. In addition, proteins with hematopoietic functions such as cyclin-dependent kinase 6, histone deacetylase 9 and transforming growth factor beta 2 and 3 were down-regulated as well. Elevated levels of P21 and GADD45, in concert with cyclin D1, also mediated the antiproliferative response of HL-60 in the presence of 1,25-(OH)2D3 and PS121912. Studies at higher concentration of P121912 identified a VDR-independent pathway of antiproliferation that included the enzymatic and transcriptional activation of caspase 3/7. CONCLUSION: Overall, we conclude that PS121912 behaves like a VDR antagonist at low concentrations but interacts with more targets at higher concentrations leading to apoptosis mediated by caspase 3/7 activation. In addition, PS121912 showed an acceptable metabolic stability to enable in vivo cancer studies.


Asunto(s)
Antimetabolitos Antineoplásicos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Receptores de Calcitriol , Antimetabolitos Antineoplásicos/metabolismo , Antimetabolitos Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Células CACO-2 , Caspasas Efectoras/metabolismo , Citocromo P-450 CYP3A/metabolismo , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Activación Enzimática/efectos de los fármacos , Células HL-60 , Humanos , Receptores de Calcitriol/antagonistas & inhibidores , Receptores de Calcitriol/metabolismo , Activación Transcripcional/efectos de los fármacos , Células Tumorales Cultivadas
15.
Biochemistry ; 52(24): 4193-203, 2013 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-23713684

RESUMEN

A high-throughput screening campaign was conducted to identify small molecules with the ability to inhibit the interaction between the vitamin D receptor (VDR) and steroid receptor coactivator 2. These inhibitors represent novel molecular probes for modulating gene regulation mediated by VDR. Peroxisome proliferator-activated receptor (PPAR) δ agonist GW0742 was among the identified VDR-coactivator inhibitors and has been characterized herein as a pan nuclear receptor antagonist at concentrations of > 12.1 µM. The highest antagonist activity for GW0742 was found for VDR and the androgen receptor. Surprisingly, GW0742 behaved as a PPAR agonist and antagonist, activating transcription at lower concentrations and inhibiting this effect at higher concentrations. A unique spectroscopic property of GW0742 was identified as well. In the presence of rhodamine-derived molecules, GW0742 increased the fluorescence intensity and level of fluorescence polarization at an excitation wavelength of 595 nm and an emission wavelength of 615 nm in a dose-dependent manner. The GW0742-inhibited NR-coactivator binding resulted in a reduced level of expression of five different NR target genes in LNCaP cells in the presence of agonist. Especially VDR target genes CYP24A1, IGFBP-3, and TRPV6 were negatively regulated by GW0742. GW0742 is the first VDR ligand inhibitor lacking the secosteroid structure of VDR ligand antagonists. Nevertheless, the VDR-meditated downstream process of cell differentiation was antagonized by GW0742 in HL-60 cells that were pretreated with the endogenous VDR agonist 1,25-dihydroxyvitamin D3.


Asunto(s)
Núcleo Celular/metabolismo , Coactivador 2 del Receptor Nuclear/química , PPAR delta/agonistas , Receptores de Calcitriol/química , Tiazoles/farmacología , Línea Celular Tumoral , ADN/química , Relación Dosis-Respuesta a Droga , Células HEK293 , Células HL-60 , Humanos , Concentración 50 Inhibidora , Ligandos , Unión Proteica , Rodaminas/química , Espectrofotometría/métodos
16.
Genes Cancer ; 4(11-12): 524-34, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24386512

RESUMEN

Hypercalcemia remains a major impediment to the clinical use of vitamin D in cancer treatment. Approaches to remove hypercalcemia and development of nonhypercalcemic agents can lead to the development of vitamin D-based therapies for treatment of various cancers. In this report, in vitro and in vivo anticancer efficacy, safety, and details of vitamin D receptor (VDR) interactions of PT19c, a novel nonhypercalcemic vitamin D derived anticancer agent, are described. PT19c was synthesized by bromoacetylation of PTAD-ergocalciferol adduct. Broader growth inhibitory potential of PT19c was evaluated in a panel of chemoresistant breast, renal, ovarian, lung, colon, leukemia, prostate, melanoma, and central nervous system cancers cell line types of NCI60 cell line panel. Interactions of PT19c with VDR were determined by a VDR transactivation assay in a VDR overexpressing VDR-UAS-bla-HEK293 cells, in vitro VDR-coregulator binding, and molecular docking with VDR-ligand binding domain (VDR-LBD) in comparison with calcitriol. Acute toxicity of PT19c was determined in nontumored mice. In vivo antitumor efficacy of PT19c was determined via ovarian and endometrial cancer xenograft experiments. Effect of PT19c on actin filament organization and focal adhesion formation was examined by microscopy. PT19c treatment inhibited growth of chemoresistant NCI60 cell lines (log10GI50 ~ -4.05 to -6.73). PT19c (10 mg/kg, 35 days) reduced growth of ovarian and endometrial xenograft tumor without hypercalcemia. PT19c exerted no acute toxicity up to 400 mg/kg (QDx1) in animals. PT19c showed weak VDR antagonism, lack of VDR binding, and inverted spatial accommodation in VDR-LBD. PT19c caused actin filament dysfunction and inhibited focal adhesion in SKOV-3 cells. PT19c is a VDR independent nonhypercalcemic vitamin D-derived agent that showed noteworthy safety and efficacy in ovarian and endometrial cancer animal models and inhibited actin organization and focal adhesion in ovarian cancer cells.

17.
J Med Chem ; 55(10): 4640-51, 2012 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-22563729

RESUMEN

The vitamin D receptor (VDR) is a nuclear hormone receptor that regulates cell proliferation, cell differentiation, and calcium homeostasis. The receptor is activated by vitamin D analogues that induce the disruption of VDR-corepressor binding and promote VDR-coactivator interactions. The interactions between VDR and coregulators are essential for VDR-mediated transcription. Small molecule inhibition of VDR-coregulator binding represents an alternative method to the traditional ligand-based approach in order to modulate the expression of VDR target genes. A high throughput fluorescence polarization screen that quantifies the inhibition of binding between VDR and a fluorescently labeled steroid receptor coactivator 2 peptide was applied to discover the new small molecule VDR-coactivator inhibitors, 3-indolylmethanamines. Structure-activity relationship studies with 3-indolylmethanamine analogues were used to determine their mode of VDR-binding and to produce the first VDR-selective and irreversible VDR-coactivator inhibitors with the ability to regulate the transcription of the human VDR target gene TRPV6.


Asunto(s)
Indoles/síntesis química , Metilaminas/síntesis química , Coactivadores de Receptor Nuclear/antagonistas & inhibidores , Receptores de Calcitriol/antagonistas & inhibidores , Línea Celular , Ensayos Analíticos de Alto Rendimiento , Humanos , Indoles/química , Indoles/farmacología , Membranas Artificiales , Metilaminas/química , Metilaminas/farmacología , Coactivador 2 del Receptor Nuclear/antagonistas & inhibidores , Coactivador 2 del Receptor Nuclear/metabolismo , Coactivador 3 de Receptor Nuclear/antagonistas & inhibidores , Coactivador 3 de Receptor Nuclear/metabolismo , Coactivadores de Receptor Nuclear/metabolismo , Permeabilidad , Unión Proteica , Receptores de Calcitriol/metabolismo , Solubilidad , Relación Estructura-Actividad , Canales Catiónicos TRPV/genética , Transcripción Genética/efectos de los fármacos
18.
PLoS One ; 7(4): e34443, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22509304

RESUMEN

BACKGROUND: Numerous vitamin-D analogs exhibited poor response rates, high systemic toxicities and hypercalcemia in human trials to treat cancer. We identified the first non-hypercalcemic anti-cancer vitamin D analog MT19c by altering the A-ring of ergocalciferol. This study describes the therapeutic efficacy and mechanism of action of MT19c in both in vitro and in vivo models. METHODOLOGY/PRINCIPAL FINDING: Antitumor efficacy of MT19c was evaluated in ovarian cancer cell (SKOV-3) xenografts in nude mice and a syngenic rat ovarian cancer model. Serum calcium levels of MT19c or calcitriol treated animals were measured. In-silico molecular docking simulation and a cell based VDR reporter assay revealed MT19c-VDR interaction. Genomewide mRNA analysis of MT19c treated tumors identified drug targets which were verified by immunoblotting and microscopy. Quantification of cellular malonyl CoA was carried out by HPLC-MS. A binding study with PPAR-Y receptor was performed. MT19c reduced ovarian cancer growth in xenograft and syngeneic animal models without causing hypercalcemia or acute toxicity. MT19c is a weak vitamin-D receptor (VDR) antagonist that disrupted the interaction between VDR and coactivator SRC2-3. Genome-wide mRNA analysis and western blot and microscopy of MT19c treated xenograft tumors showed inhibition of fatty acid synthase (FASN) activity. MT19c reduced cellular levels of malonyl CoA in SKOV-3 cells and inhibited EGFR/phosphoinositol-3kinase (PI-3K) activity independently of PPAR-gamma protein. SIGNIFICANCE: Antitumor effects of non-hypercalcemic agent MT19c provide a new approach to the design of vitamin-D based anticancer molecules and a rationale for developing MT19c as a therapeutic agent for malignant ovarian tumors by targeting oncogenic de novo lipogenesis.


Asunto(s)
Ergocalciferoles/química , Ácidos Grasos/biosíntesis , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Secuencia de Aminoácidos , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Calcio/sangre , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Ácido Cítrico/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Receptores ErbB/metabolismo , Ergocalciferoles/efectos adversos , Ergocalciferoles/metabolismo , Ergocalciferoles/farmacología , Ácido Graso Sintasas/metabolismo , Femenino , Homeostasis/efectos de los fármacos , Humanos , Hipercalcemia/inducido químicamente , L-Lactato Deshidrogenasa/metabolismo , Malonil Coenzima A/biosíntesis , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Ováricas/genética , Ratas , Receptores de Calcitriol/antagonistas & inhibidores , Receptores de Calcitriol/química , Receptores de Calcitriol/metabolismo , Seguridad , Transducción de Señal/efectos de los fármacos
19.
Cancer Cell ; 20(3): 384-99, 2011 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-21907928

RESUMEN

Using a mouse model of ependymoma-a chemoresistant brain tumor-we combined multicell high-throughput screening (HTS), kinome-wide binding assays, and in vivo efficacy studies, to identify potential treatments with predicted toxicity against neural stem cells (NSC). We identified kinases within the insulin signaling pathway and centrosome cycle as regulators of ependymoma cell proliferation, and their corresponding inhibitors as potential therapies. FDA approved drugs not currently used to treat ependymoma were also identified that posses selective toxicity against ependymoma cells relative to normal NSCs both in vitro and in vivo, e.g., 5-fluorouracil. Our comprehensive approach advances understanding of the biology and treatment of ependymoma including the discovery of several treatment leads for immediate clinical translation.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Ensayos de Selección de Medicamentos Antitumorales , Ependimoma/tratamiento farmacológico , Ensayos Analíticos de Alto Rendimiento/métodos , Células-Madre Neurales/efectos de los fármacos , Animales , Ácidos Borónicos/farmacología , Bortezomib , Encéfalo/patología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular , Centrosoma/metabolismo , Ependimoma/metabolismo , Ependimoma/patología , Fluorouracilo/farmacología , Insulina/metabolismo , Ratones , Ratones Desnudos , Pirazinas/farmacología , Transducción de Señal , Células Tumorales Cultivadas
20.
Gynecol Oncol ; 123(2): 370-8, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21803404

RESUMEN

OBJECTIVE: In human trials calcitriol and its analogs displayed unacceptable systemic toxicities including hypercalcemia. This study was designed to evaluate a novel non-hypercalcemic vitamin-D derivative (MT19c) and its anticancer effects in cultured ovarian cancer cell model. METHODS: We modified the Ergocalciferol structure to generate MT19c, a heterocyclic vitamin-D derivative. Hypercalcemic liabilities of MT19c were assessed by estimating the blood calcium levels in drug treated animals. VDR agonistic or antagonistic properties of MT19c were determined via a VDR-coactivator binding assay. The anticancer effects of MT19c were evaluated by (i) cytotoxicity studies in cancer cell lines and the National Cancer Institute (NCI(60)) cell lines, (ii) identification of apoptosis markers by microscopy and western blots, (iii) cell cycle analysis, and (iv) by studying the insulin receptor substrate-1/2 (IRS1/2) signaling in ovarian cancer cells (SKOV-3) by western blotting. RESULTS: MT19c treatment did not cause hypercalcemia in mice and showed minor VDR antagonistic activity. In a NCI(60) screen MT19c revealed cell-type specific growth inhibition. MT19c displayed superior cytotoxicity to cisplatin, calcitriol, EB1089 and Iressa in SKOV-3 cell-lines and was comparable to Taxol in our in vitro assays. In SKOV-3 cells MT19c showed caspase dependent apoptosis, DNA fragmentation and cell cycle arrest. MT19c did not alter VDR but downregulated the IGFR/IRS-1/2-MEK-ras-ERK1/2-pathway via activated TNFα-receptor/SAPK/JNK component. CONCLUSION: Our results demonstrate how structural optimization of the vitamin-D scaffold leads to identification of a non-hypercalcemic compound MT19c which exerts cytotoxicity in vitro based on a VDR-independent signaling pathway and displays potent anti-cancer activity in ovarian cancer cell models.


Asunto(s)
Antineoplásicos/farmacología , Ergocalciferoles/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Calcio/sangre , Línea Celular Tumoral , Femenino , Humanos , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Neoplasias Ováricas/sangre , Neoplasias Ováricas/patología , Receptores de Calcitriol/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA