Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Arch Pharm (Weinheim) ; 349(2): 104-11, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26686665

RESUMEN

A series of novel pyridazin-6-one-1-acetylhydrazone hybrids were rationally designed to inhibit phosphodiesterase 4 (PDE4B). The prepared compounds were evaluated for their in vitro ability to inhibit the PDE4B enzyme; several of these compounds showed moderate activity compared to the reference drug, rolipram. Compounds 6, 12, and 14 emerged as the most potent inhibitors in this series. The [3-(4-methoxyphenyl)-6-oxo-5,6-dihydro-4H-pyridazin-1-yl]acetic acid [1-(3,4,5-trimethoxyphenyl)ethylidene]hydrazide (12) showed an IC50 value of 13 µM against PDE4B. Docking of 6, 12, and 14 into the active site of PDE4B illustrates their possible binding mode and provides insights for further optimization of this drug scaffold.


Asunto(s)
Hidrazonas/química , Inhibidores de Fosfodiesterasa 4/química , Piridazinas/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/química , Humanos , Hidrazonas/síntesis química , Simulación del Acoplamiento Molecular , Inhibidores de Fosfodiesterasa 4/síntesis química , Piridazinas/síntesis química , Relación Estructura-Actividad
2.
Arch Pharm (Weinheim) ; 347(9): 650-7, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24985336

RESUMEN

A novel series of quinazolin-4(3H)-one/Schiff base hybrids was rationally designed and synthesized. The prepared compounds were evaluated for in vitro activity to inhibit phosphodiesterase 4 (PDE4), where several of them showed good-to-moderate activity compared to rolipram. Compound 7 showed potent PDE4 inhibition in this series, with an IC50 of 1.60 µM. Compounds that showed PDE4 inhibition were further assessed for antiproliferative activity using different human tumor cell lines. Compound 10 exhibited significant antiproliferative activity with IC50 values of 140, 79, and 320 nM in breast, lung, and colon tumor cells, respectively. Docking of compound 7 in the active site of PDE4B illustrates its possible binding mode and provides insight for further optimizations of this novel scaffold for inhibiting PDE4.


Asunto(s)
Antineoplásicos/síntesis química , Proliferación Celular/efectos de los fármacos , Diseño de Fármacos , Inhibidores de Fosfodiesterasa 4/síntesis química , Quinazolinonas/síntesis química , Bases de Schiff/química , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Humanos , Concentración 50 Inhibidora , Simulación del Acoplamiento Molecular , Estructura Molecular , Inhibidores de Fosfodiesterasa 4/química , Inhibidores de Fosfodiesterasa 4/farmacología , Quinazolinonas/química , Quinazolinonas/farmacología
3.
Med Chem Res ; 23(9): 4177-4188, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27559271

RESUMEN

A structure-based medicinal chemistry strategy was applied to design new naproxen derivatives that show growth inhibitory activity against human colon tumor cells through a cyclooxygenase (COX)-independent mechanism. In vitro testing of the synthesized compounds against the human HT-29 colon tumor cell line revealed enhanced growth inhibitory activity compared to the parent naproxen with 3a showing IC50 of 11.4 µM (two orders of magnitude more potent than naproxen). Selectivity of 3a was investigated against a panel of three tumor and one normal colon cell lines and showed up to six times less toxicity against normal colonocytes. Compound 3a was shown to induce dose-dependent apoptosis of HT116 colon tumor cells as evidenced by measuring the activity of caspases-3 and 7. None of the synthesized compounds showed activity against COX-1 or COX-2 isozymes, confirming a COX-independent mechanism of action. Compound 3k was found to have no ulcerogenic effect in rats as indicated by electron microscope scanning of the stomach after oral administration. A pharmacophore model was developed for elucidating structure-activity relationships and subsequent chemical optimization for this series of compounds as colorectal cancer chemopreventive drugs.

4.
Bioorg Med Chem ; 21(23): 7343-56, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24139845

RESUMEN

Derivatives with scaffolds of 1,3,5-tri-substituted pyrazoline and 1,3,4,5-tetra-substituted pyrazoline were synthesized and tested for their inhibitory effects versus the p53(+/+) HCT116 and p53(-/-) H1299 human tumor cell lines. Several compounds were active against the two cell lines displaying IC50 values in the low micromolar range with a clearly more pronounced effect on the p53(+/+) HCT116 cells. The compound class shows excellent developability due to the modular synthesis, allowing independent optimization of all three to four key substituents to improve the properties of the molecules.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Inhibidores de Crecimiento/química , Inhibidores de Crecimiento/farmacología , Neoplasias/tratamiento farmacológico , Proteína p53 Supresora de Tumor/genética , Línea Celular Tumoral , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/genética , Relación Estructura-Actividad
5.
Arch Pharm (Weinheim) ; 346(1): 23-33, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23307609

RESUMEN

New derivatives with the tetrahydro-ß-carboline-imidazolidinedione and tetrahydro-ß-carboline-piperazinedione scaffolds and a pendant bromothienyl moiety at C-5/C-6 were synthesized and tested for their ability to inhibit PDE5 in vitro. The following SAR can be concluded: The tetracyclic scaffold is essential for PDE5 inhibition; the ethyl group is the most suitable among the adopted N-substituents on the terminal ring (hydantoin/piperazinedione); the appropriate stereochemistry of C-5/C-6 derived from the aldehyde rather than C-11a/C-12a derived from tryptophan appears crucial for inhibition of PDE5; surprisingly, derivatives with the hydantoin terminal ring are more active than their analogs with the piperazinedione ring; the selectivity versus PDE5 relative to PDE11 with cGMP as a substrate is mainly a function of the substitution and stereochemistry pattern of the external ring, in other words of the interaction with the H-loop residues of the isozymes. Thirteen derivatives showed PDE5 inhibitory activity with IC(50) values in the range of 0.16-5.4 µm. Compound 8 was the most potent PDE5 inhibitor and showed selectivity towards PDE5 versus other PDEs, with a selectivity index of 49 towards PDE5 rather than PDE11 with cGMP as the substrate.


Asunto(s)
Carbolinas/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/efectos de los fármacos , Inhibidores de Fosfodiesterasa 5/farmacología , Carbolinas/síntesis química , Carbolinas/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Dicetopiperazinas/síntesis química , Dicetopiperazinas/química , Dicetopiperazinas/farmacología , Diseño de Fármacos , Humanos , Imidazolidinas/síntesis química , Imidazolidinas/química , Imidazolidinas/farmacología , Concentración 50 Inhibidora , Estructura Molecular , Inhibidores de Fosfodiesterasa 5/síntesis química , Inhibidores de Fosfodiesterasa 5/química , Relación Estructura-Actividad
6.
Cancers (Basel) ; 15(3)2023 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-36765604

RESUMEN

The nonsteroidal anti-inflammatory drug (NSAID) sulindac demonstrates attractive anticancer activity, but the toxicity resulting from cyclooxygenase (COX) inhibition and the suppression of physiologically important prostaglandins precludes its long-term, high dose use in the clinic for cancer prevention or treatment. While inflammation is a known tumorigenic driver, evidence suggests that sulindac's antineoplastic activity is partially or fully independent of its COX inhibitory activity. One COX-independent target proposed for sulindac is cyclic guanosine monophosphate phosphodiesterase (cGMP PDE) isozymes. Sulindac metabolites, i.e., sulfide and sulfone, inhibit cGMP PDE enzymatic activity at concentrations comparable with those associated with cancer cell growth inhibitory activity. Additionally, the cGMP PDE isozymes PDE5 and PDE10 are overexpressed during the early stages of carcinogenesis and appear essential for cancer cell proliferation and survival based on gene silencing experiments. Here, we describe a novel amide derivative of sulindac, sulindac sulfide amide (SSA), which was rationally designed to eliminate COX-inhibitory activity while enhancing cGMP PDE inhibitory activity. SSA was 68-fold and 10-fold less potent than sulindac sulfide (SS) in inhibiting COX-1 and COX-2, respectively, but 10-fold more potent in inhibiting growth and inducing apoptosis in breast cancer cells. The pro-apoptotic activity of SSA was associated with inhibition of cGMP PDE activity, elevation of intracellular cGMP levels, and activation of cGMP-dependent protein kinase (PKG) signaling, as well as the inhibition of ß-catenin/Tcf transcriptional activity. SSA displayed promising in vivo anticancer activity, resulting in a 57% reduction in the incidence and a 62% reduction in the multiplicity of tumors in the N-methyl-N-nitrosourea (MNU)-induced model of breast carcinogenesis. These findings provide strong evidence for cGMP/PKG signaling as a target for breast cancer prevention or treatment and the COX-independent anticancer properties of sulindac. Furthermore, this study validates the approach of optimizing off-target effects by reducing the COX-inhibitory activity of sulindac for future targeted drug discovery efforts to enhance both safety and efficacy.

7.
Arch Pharm (Weinheim) ; 344(3): 149-57, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21384413

RESUMEN

Starting from tadalafil as a template, a series of functionalized tetrahydro-ß-carboline derivatives have been prepared and identified as novel potent and selective PDE5 inhibitors. Replacing the 3,4-methylenedioxyphenyl at position 6 of tadalafil, together with elongation of the N2-methyl substituent and manipulation of the stereochemical aspects of the two chiral carbons led to the identification of compound XXI, a highly potent PDE5 inhibitor (IC(50) = 3 nM). Compound XXI was also highly selective for PDE5 versus PDE3B, PDE4B, and PDE11A, with a selectivity index of 52 and 235 towards PDE5 rather than PDE11 with both cAMP and cGMP as substrate, respectively.


Asunto(s)
Carbolinas/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/efectos de los fármacos , Inhibidores de Fosfodiesterasa 5/farmacología , Carbolinas/síntesis química , Carbolinas/química , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Diseño de Fármacos , Humanos , Concentración 50 Inhibidora , Inhibidores de Fosfodiesterasa 5/síntesis química , Inhibidores de Fosfodiesterasa 5/química , Relación Estructura-Actividad , Tadalafilo
8.
Bioorg Med Chem ; 17(16): 5974-82, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19628397

RESUMEN

Two series of 4,6-diaryl-2-imino-1,2-dihydropyridine-3-carbonitriles and their isosteric 4,6-diaryl-2-oxo-1,2-dihydropyridine-3-carbonitriles were synthesized through a combinatorial approach. The prepared analogues were evaluated for their in vitro capacity to inhibit PDE3A and the growth of the human HT-29 colon adenocarcinoma tumor cell line. Compound 6-(4-bromophenyl)-4-(2-ethoxyphenyl)-2-imino-1,2-dihydropyridine-3-carbonitrile (Id) exhibited the strongest PDE3 inhibition when cGMP but not cAMP is the substrate with a IC(50)of 27microM, which indicates a highly selective mechanism of enzyme inhibition. On the other hand, compound 6-(1,3-benzodioxol-5-yl)-4-(2-ethoxyphenyl)-2-imino-1,2-dihydropyridine-3-carbonitrile (Ii) was the most active in inhibiting colon tumor cell growth with a IC(50) of 3microM. The electronic effects, steric effects and conformational aspects of Id seem to be the most crucial for the PDE3 inhibition. Meanwhile, steric factors and the H-bonding capability seem to be the most important factors for tumor cell growth inhibitory activity. Conversely, there is no direct correlation between PDE3 inhibition and anticancer activity for the prepared compounds. An in silico docking experiment indicates the potential involvement of other potential molecular targets such as PIM-1 kinase to explain its tumor cell growth inhibitory activity.


Asunto(s)
Antineoplásicos/síntesis química , Inhibidores de Fosfodiesterasa 3 , Inhibidores de Fosfodiesterasa/síntesis química , Piridinas/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Sitios de Unión , Línea Celular Tumoral , Simulación por Computador , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Diseño de Fármacos , Humanos , Inhibidores de Fosfodiesterasa/química , Inhibidores de Fosfodiesterasa/farmacología , Piridinas/química , Piridinas/farmacología , Relación Estructura-Actividad
9.
Med Chem ; 12(5): 472-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26648332

RESUMEN

In view of the emerging clinical indications for Phosphodiesterase 9 inhibitors e.g. treatment of Alzheimer, diabetes, cancer, and the limited number of its selective inhibitors which possess a single chemical scaffolds, a structure-based approach was undertaken to mine the ZINC database by virtual screening to identify novel PDE9 inhibitors. The database, which was never reported to have been used before for discovery of PDE9 inhibitors, was screened against the ligand binding pocket of the PDE9 complex (PDB:4GH6) using molecular docking programs, MOE and AutoDock Vina in PyRx. Three different scoring functions were used to evaluate the docking poses and scores of the compounds, and the compounds were selected through consensus selection, thus reducing the margin of error in docking. The highest scoring compounds were then selected and purchased for in vitro testing as PDE9 inhibitors and cancer growth inhibitory agents. This led to the discovery of three previously unreported potent PDE 9 inhibitory compounds with two unique chemical scaffolds. Consistent with the role of PDE9 in cancer cell growth, the compounds also inhibited the growth of breast tumor cell lines, MCF-7 and MDA-468 at concentrations comparable to those that inhibited PDE9.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Compuestos Azo/farmacología , Furocumarinas/farmacología , Naftalenosulfonatos/farmacología , Inhibidores de Fosfodiesterasa/farmacología , 3',5'-AMP Cíclico Fosfodiesterasas/química , 3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Compuestos Azo/química , Minería de Datos , Bases de Datos de Compuestos Químicos , Diseño de Fármacos , Furocumarinas/química , Humanos , Ligandos , Células MCF-7 , Simulación del Acoplamiento Molecular , Estructura Molecular , Naftalenosulfonatos/química , Inhibidores de Fosfodiesterasa/química
10.
J Biomed Res ; 30(2): 120-133, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28276667

RESUMEN

ATP-binding cassette (ABC) transporters ABCC1 (MRP1), ABCB1 (P-gp), and ABCG2 (BCRP) contribute to chemotherapy failure. The primary goals of this study were to characterize the efficacy and mechanism of the nonsteroidal anti-inflammatory drug (NSAID), sulindac sulfide, to reverse ABCC1 mediated resistance to chemotherapeutic drugs and to determine if sulindac sulfide can influence sensitivity to chemotherapeutic drugs independently of drug efflux. Cytotoxicity assays were performed to measure resistance of ABC-expressing cell lines to doxorubicin and other chemotherapeutic drugs. NSAIDs were tested for the ability to restore sensitivity to resistance selected tumor cell lines, as well as a large panel of standard tumor cell lines. Other experiments characterized the mechanism by which sulindac sulfide inhibits ABCC1 substrate and co-substrate (GSH) transport in isolated membrane vesicles and intact cells. Selective reversal of multi-drug resistance (MDR), decreased efflux of doxorubicin, and fluorescent substrates were demonstrated by sulindac sulfide and a related NSAID, indomethacin, in resistance selected and engineered cell lines expressing ABCC1, but not ABCB1 or ABCG2. Sulindac sulfide also inhibited transport of leukotriene C4 into membrane vesicles. Sulindac sulfide enhanced the sensitivity to doxorubicin in 24 of 47 tumor cell lines, including all melanoma lines tested (7-7). Sulindac sulfide also decreased intracellular GSH in ABCC1 expressing cells, while the glutathione synthesis inhibitor, BSO, selectively increased sensitivity to sulindac sulfide induced cytotoxicity. Sulindac sulfide potently and selectively reverses ABCC1-mediated MDR at clinically achievable concentrations. ABCC1 expressing tumors may be highly sensitive to the direct cytotoxicity of sulindac sulfide, and in combination with chemotherapeutic drugs that induce oxidative stress.

11.
PLoS One ; 9(6): e98370, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24896091

RESUMEN

BACKGROUND: Triple-negative breast cancer (TNBC) is a highly diverse group that is associated with an aggressive phenotype. Its treatment has been challenging due to its heterogeneity and absence of well-defined molecular targets. Thus, there is an urgent need to identify novel agents with therapeutic application. NF-κB is over-expressed in many breast cancers; thus, inactivation of the NF-κB pathway could serve as a therapeutic target. Here we report for the first time the anti-tumor activity of panepoxydone (PP), a NF-κB inhibitor isolated from an edible mushroom, in several breast cancer cell lines. METHODS: We investigated the effects of PP on cell growth, migration-invasion, apoptosis and EMT-related proteins expression in MCF-7 and TNBC cell lines MDA-MB-231, MDA-MB-468 and MDA-MB-453. RESULTS: Significant antitumor activity was seen in all cell lines, with differential responses noted in cell-line specific manner. Treatment with PP resulted in significant cytotoxicity, decreased invasion, migration and increased apoptosis in all cell lines tested. Up-regulation of Bax and cleaved PARP and down-regulation of Bcl-2, survivin, cyclin D1 and caspase 3 were noted in PP-treated breast cancer cells. The antitumor effect of PP appeared related to its ability to inhibit the phosphorylation of inhibitor of NF-κB (IκBα) with cytoplasmic accumulation. PP treatment also down-regulated FOXM1 which resulted in a reversal of EMT. Similar results were obtained after silencing of NF-kB and FOXM1. CONCLUSION: Altogether, these studies show, for the first time the antitumor activity of PP against breast cancer cells, in particular TNBC cells. Furthermore, it highlights the concept that optimal treatment of TNBC warrants attention to the differential sensitivity of various TNBC subtypes to therapeutic agents. These results suggest that the PP may be a potentially effective chemopreventive or therapeutic agent against breast cancer. However, additional studies are required to more fully elucidate the mechanism of antitumor effect of PP.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Proliferación Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Factores de Transcripción Forkhead/metabolismo , FN-kappa B/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Antineoplásicos/uso terapéutico , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Proteína Forkhead Box M1 , Humanos , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Regulación hacia Arriba/efectos de los fármacos
12.
Eur J Med Chem ; 70: 273-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24161704

RESUMEN

Two series of 2-(3,5-diaryl-4,5-dihydropyrazol-1-yl)-1-methyl-6-oxo-4-phenyl-1,6-dihydropyrimidine-5-carbonitriles 5a-h and 4-(4-chlorophenyl)-2-(3,5-diaryl-4,5-dihydropyrazol-1-yl)-1-methyl-6-oxo-1,6-dihydropyrimidine-5-carbonitriles 6a-h were synthesized via a cyclocondensation reaction of the corresponding 2-hydrazinopyrimidines 3a,b with the appropriate 2-propen-1-ones 4a-h. The target compounds were screened for their antiproliferative activity against A 549 (lung), HT 29 (colon), MCF 7 and MDA-MB 231 (breast) cell lines. The two most susceptible cell lines were the colon (HT 29) and breast (MDA-MB 231). Generally, the 4-unsubstitutedphenylpyrimidine derivatives 5a-h were more active than their 4-chlorophenylpyrimidine analogs 6a-h. Compounds 5e and 5g, showed high activity against three of the cell lines. The most active compound 5c possessed IC50 = 1.76 µM against A 549 cell line.


Asunto(s)
Antineoplásicos/farmacología , Pirazoles/química , Pirimidinas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HT29 , Humanos , Células MCF-7 , Estructura Molecular , Pirimidinas/síntesis química , Pirimidinas/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
13.
Mol Cancer Ther ; 12(9): 1848-59, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23804703

RESUMEN

Nonsteroidal anti-inflammatory drugs (NSAID) display promising antineoplastic activity for colorectal and other cancers, but toxicity from COX inhibition limits their long-term use for chemoprevention. Previous studies have concluded that the basis for their tumor cell growth inhibitory activity does not require COX inhibition, although the underlying mechanism is poorly understood. Here, we report that the NSAID sulindac sulfide inhibits cyclic guanosine 3',5'-monophosphate phosphodiesterase (cGMP PDE) activity to increase intracellular cGMP levels and activate cGMP-dependent protein kinase (PKG) at concentrations that inhibit proliferation and induce apoptosis of colon tumor cells. Sulindac sulfide did not activate the cGMP/PKG pathway, nor affect proliferation or apoptosis in normal colonocytes. Knockdown of the cGMP-specific PDE5 isozyme by siRNA and PDE5-specific inhibitors tadalafil and sildenafil also selectively inhibited the growth of colon tumor cells that expressed high levels of PDE5 compared with colonocytes. The mechanism by which sulindac sulfide and the cGMP/PKG pathway inhibits colon tumor cell growth involves the transcriptional suppression of ß-catenin to inhibit Wnt/ß-catenin T-cell factor transcriptional activity, leading to downregulation of cyclin D1 and survivin. These observations suggest that safer and more efficacious sulindac derivatives can be developed for colorectal cancer chemoprevention by targeting PDE5 and possibly other cGMP-degrading isozymes.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/patología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Sulindac/análogos & derivados , Vía de Señalización Wnt/efectos de los fármacos , Antineoplásicos/análisis , Apoptosis/efectos de los fármacos , Células CACO-2 , Carbolinas/farmacología , Línea Celular , Neoplasias del Colon/metabolismo , GMP Cíclico/genética , Ciclina D1/metabolismo , Células HCT116 , Células HT29 , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Inhibidores de Fosfodiesterasa 5/farmacología , Piperazinas/farmacología , Purinas/farmacología , Citrato de Sildenafil , Sulfonas/farmacología , Sulindac/análisis , Sulindac/farmacología , Survivin , Tadalafilo
14.
Eur J Med Chem ; 57: 329-43, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23117589

RESUMEN

By studying the co-crystal information of interactions between PDE5 and its inhibitors, forty new tetrahydro-ß-carbolines based-analogues were synthesized, and tested for their PDE5 inhibition. Some compounds were as active as tadalafil in inhibiting PDE5 and of better selectivity profile particularly versus PDE11A, the nature of the terminal ring and its nitrogen substituent are the main determinants of selectivity. Ensemble docking confirmed the role of H-loop closed conformer in activity versus its occluded and open forms. Conformational studies showed the effect of bulkiness of the terminal ring N-alkyl substituent on the formation of stable enzyme ligands conformers. The difference in potencies of hydantoin and piperazinedione analogues, together with the necessity of C-5/C-6 R-absolute configuration has been revealed through molecular docking.


Asunto(s)
Carbolinas/síntesis química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/química , Dicetopiperazinas/síntesis química , Hidantoínas/síntesis química , Inhibidores de Fosfodiesterasa 5/síntesis química , 3',5'-GMP Cíclico Fosfodiesterasas , Carbolinas/química , Proliferación Celular , Dicetopiperazinas/química , Diseño de Fármacos , Pruebas de Enzimas , Humanos , Hidantoínas/química , Enlace de Hidrógeno , Concentración 50 Inhibidora , Cinética , Simulación del Acoplamiento Molecular , Inhibidores de Fosfodiesterasa 5/química , Hidrolasas Diéster Fosfóricas/química , Estructura Secundaria de Proteína , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/química , Relación Estructura-Actividad , Tadalafilo , Termodinámica
15.
Med Chem ; 8(3): 392-400, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22530887

RESUMEN

Two series of compounds with the general formula of 4,6-diaryl-2-oxo-1,2 dihydropyridine-3-carbonitriles and their isosteric imino derivatives were synthesized through a one pot reaction of acetophenone, aldehyde and ammonium acetate with ethyl cyanoacetate or malononitrile, respectively. The synthesized compounds were evaluated for tumor cell growth inhibitory using the human HT-29 colon and MDA-MB-231 breast tumor cell lines. Compound 4-(2- Ethoxyphenyl)-2-imino-6-(4-fluorophenyl)-1,2-dihydropyridine-3 carbonitrile (6) showed IC50 value of 0.70 µM versus HT-29. Meanwhile, compound 4-(2-Hydroxyphenyl)-2-imino-6-(4-fluorophenyl)-1,2-dihydropyridine-3-carbonitrile (4) showed IC50 value of 4.6 µM versus MDA-MB-231. Docking compound 10 to possible molecular targets, survivin and PIM1 kinase showed appreciable interactions with both, which suggest possible targets for the antitumor activity of this novel class of anticancer compounds.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Dihidropiridinas/farmacología , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Dihidropiridinas/síntesis química , Dihidropiridinas/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Modelos Moleculares , Estructura Molecular , Estereoisomerismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
16.
Cancer Prev Res (Phila) ; 5(6): 822-33, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22556201

RESUMEN

Nonsteroidal anti-inflammatory drugs (NSAIDs) have been widely reported to inhibit tumor growth by a COX-independent mechanism, although alternative targets have not been well defined or used to develop improved drugs for cancer chemoprevention. Here, we characterize a novel sulindac derivative referred to as sulindac benzylamine (SBA) that does not inhibit COX-1 or COX-2, yet potently inhibits the growth and induces the apoptosis of human colon tumor cells. The basis for this activity appears to involve cyclic guanosine 3',5',-monophosphate phosphodiesterase (cGMP PDE) inhibition as evident by its ability to inhibit cGMP hydrolysis in colon tumor cell lysates and purified cGMP-specific PDE5, increase intracellular cGMP levels, and activate cGMP-dependent protein kinase G at concentrations that suppress tumor cell growth. PDE5 was found to be essential for colon tumor cell growth as determined by siRNA knockdown studies, elevated in colon tumor cells as compared with normal colonocytes, and associated with the tumor selectivity of SBA. SBA activation of PKG may suppress the oncogenic activity of ß-catenin as evident by its ability to reduce ß-catenin nuclear levels, Tcf (T-cell factor) transcriptional activity, and survivin levels. These events preceded apoptosis induction and appear to result from a rapid elevation of intracellular cGMP levels following cGMP PDE inhibition. We conclude that PDE5 and possibly other cGMP degrading isozymes can be targeted to develop safer and more efficacious NSAID derivatives for colorectal cancer chemoprevention.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias del Colon/patología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Inhibidores de Fosfodiesterasa 5/farmacología , Sulindac/farmacología , Activación Transcripcional/efectos de los fármacos , beta Catenina/genética , Apoptosis/efectos de los fármacos , Western Blotting , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/enzimología , Neoplasias del Colon/genética , GMP Cíclico/metabolismo , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Humanos , Modelos Moleculares , ARN Interferente Pequeño/genética , Sulindac/análogos & derivados , Factores de Transcripción TCF/genética , Factores de Transcripción TCF/metabolismo
17.
Cancer Prev Res (Phila) ; 4(8): 1275-84, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21505183

RESUMEN

Nonsteroidal anti-inflammatory drugs (NSAID) such as sulindac sulfide (SS) display promising antineoplastic properties, but toxicities resulting from COX inhibition limit their clinical use. Although COX inhibition is responsible for the anti-inflammatory activity of SS, recent studies suggest that phosphodiesterase (PDE) 5 inhibition and activation of cyclic guanosine monophosphate (cGMP) signaling are closely associated with its ability to induce apoptosis of tumor cells. However, the underlying mechanisms responsible for apoptosis induction, factors that influence sensitivity of tumor cells to SS, and the importance of PDE5 for breast tumor cell growth have not been established. Here we show that SS can induce apoptosis of breast tumor cells, which predominantly rely on PDE5 for cGMP hydrolysis but not normal mammary epithelial cells, which rely on PDE isozymes other than PDE5 for cGMP hydrolysis. Inhibition of PDE5 and activation of protein kinase G (PKG) by SS was associated with increased ß-catenin phosphorylation, decreased ß-catenin mRNA and protein levels, reduced ß-catenin nuclear localization, decreased T-cell factor/lymphoid enhancer factor (Tcf/Lef) promoter activity, and decreased expression of Wnt/ß-catenin-regulated proteins. Suppression of PDE5 with siRNA or known PDE5 inhibitors was sufficient to selectively induce apoptosis and attenuate ß-catenin-mediated transcription in breast tumor cells with minimal effects on normal mammary epithelial cells. These findings provide evidence that SS induces apoptosis of breast tumor cells through a mechanism involving inhibition of PDE5 and attenuation of oncogenic Wnt/ß-catenin-mediated transcription. We conclude that PDE5 represents a novel molecular target for the discovery of safer and more efficacious drugs for breast cancer chemoprevention.


Asunto(s)
Apoptosis , Neoplasias de la Mama/tratamiento farmacológico , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/genética , Inhibidores de Fosfodiesterasa 5/farmacología , Sulindac/análogos & derivados , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Línea Celular Tumoral , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Femenino , Humanos , Isoenzimas , Oncogenes , Transducción de Señal , Sulindac/farmacología , Transcripción Genética , Proteínas Wnt/antagonistas & inhibidores , beta Catenina/antagonistas & inhibidores
18.
J Med Chem ; 54(2): 495-509, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21189023

RESUMEN

New derivatives based upon the tetrahydro-ß-carboline-hydantoin and tetrahydro-ß-carboline-piperazinedione scaffolds were synthesized. All compounds were evaluated for their ability to inhibit PDE5 in vitro, and numerous compounds with IC(50) values in the low nanomolar range were identified including compounds derived from l-tryptophan. Compounds with high potency versus PDE5 were then evaluated for inhibitory activity against other PDEs to assess isozyme selectivity. Compound 5R,11aS-5-(3,4-dichlorophenyl)-2-ethyl-5,6,11,11a-tetrahydro-1H-imidazo[1',5':1,6]pyrido[3,4-b]indole-1,3(2H)dione 14 showed a selectivity index of >200 for cGMP hydrolysis by PDE5 versus PDE11. Meanwhile, 6R,12aR-6-(2,4-dichlorophenyl)-2-ethyl-2,3,6,7,12,12a-hexahydropyrazino[1',2':1,6]pyrido[3,4-b]indole-1,4dione 45 demonstrated strong potency for inhibition of PDE11 with an IC(50) value of 11 nM, representing the most potent PDE11 inhibitor thus far reported. Docking experiments differentiated between active and inactive analogues and revealing the conformational, steric, and lipophilic necessities for potent PDE5 inhibition. Many derivatives, including potent PDE5 inhibitors, were able to inhibit the growth of the MDA-MB-231 breast tumor cell line with low micromolar potency.


Asunto(s)
Antineoplásicos/síntesis química , Carbolinas/síntesis química , Modelos Moleculares , Inhibidores de Fosfodiesterasa 5/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Carbolinas/química , Carbolinas/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Inhibidores de Fosfodiesterasa 5/química , Inhibidores de Fosfodiesterasa 5/farmacología , Proteínas Recombinantes/antagonistas & inhibidores , Estereoisomerismo , Relación Estructura-Actividad
19.
Cancer Prev Res (Phila) ; 3(7): 885-95, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20587701

RESUMEN

Nonsteroidal anti-inflammatory drugs including sulindac are well documented to be highly effective for cancer chemoprevention. However, their cyclooxygenase (COX)-inhibitory activities cause severe gastrointestinal, renal, and cardiovascular toxicities, limiting their chronic use. Recent studies suggest that COX-independent mechanisms may be responsible for the chemopreventive benefits of nonsteroidal anti-inflammatory drugs and support the potential for the development of a novel generation of sulindac derivatives lacking COX inhibition for cancer chemoprevention. A prototypic sulindac derivative with a N,N-dimethylammonium substitution called sulindac sulfide amide (SSA) was recently identified to be devoid of COX-inhibitory activity yet displays much more potent tumor cell growth-inhibitory activity in vitro compared with sulindac sulfide. In this study, we investigated the androgen receptor (AR) signaling pathway as a potential target for its COX-independent antineoplastic mechanism and evaluated its chemopreventive efficacy against prostate carcinogenesis using the transgenic adenocarcinoma of mouse prostate model. The results showed that SSA significantly suppressed the growth of human and mouse prostate cancer cells expressing AR in strong association with G(1) arrest, and decreased AR level and AR-dependent transactivation. Dietary SSA consumption dramatically attenuated prostatic growth and suppressed AR-dependent glandular epithelial lesion progression through repressing cell proliferation in the transgenic adenocarcinoma of mouse prostate mice, whereas it did not significantly affect neuroendocrine carcinoma growth. Overall, the results suggest that SSA may be a chemopreventive candidate against prostate glandular epithelial carcinogenesis.


Asunto(s)
Adenocarcinoma/prevención & control , Anticarcinógenos/uso terapéutico , Neoplasias de la Próstata/prevención & control , Sulindac/análogos & derivados , Adenocarcinoma/genética , Animales , Anticarcinógenos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimioprevención , Modelos Animales de Enfermedad , Inhibidores de Crecimiento/química , Inhibidores de Crecimiento/uso terapéutico , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Prostaglandina-Endoperóxido Sintasas/metabolismo , Receptores Androgénicos/metabolismo , Sulindac/química , Sulindac/uso terapéutico
20.
Med Chem ; 6(6): 374-87, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21054274

RESUMEN

Starting from a previously reported lead compound GR30040X (a hydantoin tetrahydro-ß-carboline derivative with a 4- pyridinyl ring at C- 5), a series of structurally related tetrahydro-ß-carboline derivatives were prepared. The tetrahydro-ß-carboline skeleton was fused either to a hydantoin or to a piperazindione ring, the pendant aryl group attached to C-5 or C-6 was changed to a 3, 4-dimethoxyphenyl or a 3-pyridinyl ring; different N-substituents on the terminal ring were introduced, a straight chain ethyl group, a branched tert. butyl and P-chlorophenyl group rather than n-butyl group of the lead compound. All four possible diastereomers of target tetrahydro-ß-carboline derivatives were prepared, separated by column chromatography and the significance of these stereochemical manipulations were studied. Synthesized compounds were evaluated for their inhibitory effect versus PDE5. Seven hits were obtained with appreciable inhibitory activity versus PDE5 with IC50s 0.14 - 4.99 µM.


Asunto(s)
Carbolinas/síntesis química , Inhibidores de Fosfodiesterasa 5/síntesis química , Carbolinas/química , Carbolinas/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Humanos , Concentración 50 Inhibidora , Inhibidores de Fosfodiesterasa 5/química , Estereoisomerismo , Relación Estructura-Actividad , Tadalafilo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA