Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Ann Oncol ; 26(10): 2173-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26202595

RESUMEN

BACKGROUND: Veliparib (ABT-888) is a potent, orally bioavailable, small-molecule inhibitor of the DNA repair enzymes poly ADP-ribose polymerase-1 and -2. Veliparib enhances the efficacy of temozolomide (TMZ) and other cytotoxic agents in preclinical tumor models. PATIENTS AND METHODS: In this multicenter, double-blind trial, adults with unresectable stage III or IV metastatic melanoma were randomized 1:1:1 to TMZ plus veliparib 20 or 40 mg, or placebo twice daily. Efficacy end points included progression-free survival (PFS), overall survival (OS), and objective response rate (ORR). RESULTS: Patients (N = 346) were randomized between February 2009 and January 2010. Median [95% confidence interval (CI)] PFS was 3.7 (3.0-5.5), 3.6 (1.9-4.1), and 2 (1.9-3.7) months in the 20-mg, 40-mg, and placebo arms, respectively. Median (95% CI) OS was 10.8 (9.0-13.1), 13.6 (11.4-15.9), and 12.9 (9.8-14.3) months, respectively; ORR was 10.3%, 8.7%, and 7.0%. Exploratory analyses showed patients with low ERCC1 expression had longer PFS when TMZ was combined with veliparib. Toxicities were as expected for TMZ. The frequencies of thrombocytopenia, neutropenia, and leukopenia were significantly increased in the veliparib groups. Grade 3 or 4 adverse events, mainly hematologic toxicities, were seen in 55%, 63%, and 41% of patients in the 20-mg, 40-mg, and placebo arms, respectively. CONCLUSIONS: Median PFS with 20 and 40 mg veliparib almost doubled numerically compared with placebo, but the improvements did not reach statistical significance. OS was not increased with veliparib. Toxicities were similar to TMZ monotherapy, but with increased frequency.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Bencimidazoles/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Dacarbazina/análogos & derivados , Melanoma/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/secundario , Dacarbazina/uso terapéutico , Método Doble Ciego , Quimioterapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Masculino , Melanoma/mortalidad , Melanoma/patología , Persona de Mediana Edad , Metástasis de la Neoplasia , Estadificación de Neoplasias , Pronóstico , Tasa de Supervivencia , Temozolomida , Adulto Joven
2.
Oncogene ; 26(38): 5655-61, 2007 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-17334390

RESUMEN

Rapamycin, a natural product inhibitor of the Raptor-mammalian target of rapamycin complex (mTORC1), is known to induce Protein kinase B (Akt/PKB) Ser-473 phosphorylation in a subset of human cancer cell lines through inactivation of S6K1, stabilization of insulin receptor substrate (IRS)-1, and increased signaling through the insulin/insulin-like growth factor-I/phosphatidylinositol 3-kinase (PI3K) axis. We report that A-443654, a potent small-molecule inhibitor of Akt serine/threonine kinases, induces Akt Ser-473 phosphorylation in all human cancer cell lines tested, including PTEN- and TSC2-deficient lines. This phenomenon is dose-dependent, manifests coincident with Akt inhibition and likely represents an alternative, rapid-feedback pathway that can be functionally dissociated from mTORC1 inhibition. Experiments performed in TSC2-/- cells indicate that TSC2 and IRS-1 cooperate with, but are dispensable for, A-443654-mediated Akt phosphorylation. This feedback event does require PI3K activity, however, as it can be inhibited by LY294002 or wortmannin. Small interfering RNA-mediated knockdown of mTOR or Rictor, components of the rapamycin-insensitive mTORC2 complex, but not the mTORC1 component Raptor, also inhibited Akt Ser-473 phosphorylation induced by A-443654. Our data thus indicate that Akt phosphorylation and activity are coupled in a manner not previously appreciated and provide a novel mode of Akt regulation that is distinct from the previously described rapamycin-induced IRS-1 stabilization mechanism.


Asunto(s)
Indazoles/farmacología , Indoles/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Línea Celular Tumoral , Cromonas/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Humanos , Immunoblotting , Diana Mecanicista del Complejo 1 de la Rapamicina , Morfolinas/farmacología , Complejos Multiproteicos , Mutación , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Serina/metabolismo , Serina-Treonina Quinasas TOR , Factores de Tiempo , Factores de Transcripción/genética , Transfección , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
3.
Oncogene ; 25(9): 1340-8, 2006 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-16247451

RESUMEN

Tumors comprise genetically heterogeneous cell populations, whose growth and survival depend on multiple signaling pathways. This has spurred the development of multitargeted therapies, including small molecules that can inhibit multiple kinases. A major challenge in designing such molecules is to determine which kinases to inhibit in each cancer to maximize efficacy and therapeutic index. We describe an approach to this problem implementing RNA interference technology. In order to identify Akt-cooperating kinases, we screened a library of kinase-directed small interfering RNAs (siRNAs) for enhanced cancer cell killing in the presence of Akt inhibitor A-443654. siRNAs targeting casein kinase I gamma 3 (CSNK1G3) or the inositol polyphosphate multikinase (IPMK) significantly enhanced A-443654-mediated cell killing, and caused decreases in Akt Ser-473 and ribosomal protein S6 phosphorylation. Small molecules targeting CSNK1G3 and/or IPMK in addition to Akt may thus exhibit increased efficacy and have the potential for improved therapeutic index.


Asunto(s)
Quinasa de la Caseína I/biosíntesis , Fosfotransferasas (Aceptor de Grupo Alcohol)/biosíntesis , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Antineoplásicos/farmacología , Quinasa de la Caseína I/genética , Muerte Celular , Pruebas Genéticas/métodos , Humanos , Indazoles/farmacología , Indoles/farmacología , Isoenzimas , Neoplasias/genética , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , ARN Interferente Pequeño , Transducción de Señal
4.
Curr Pharm Des ; 8(28): 2541-58, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12369939

RESUMEN

Human urokinase-type plasminogen activator (uPA or uPA) has been implicated in the regulation and control of basement membrane and interstitial protein degradation. Since Urokinase plays a role in tissue remodeling, it may be responsible, in part, for the disease progression of cancer. Inhibitors of urokinase may then be useful in the treatment of cancer by retarding tumor growth and metastasis. Urokinase is a multidomain protein, two regions of the protein are most responsible for the observed proteolytic activity in cancer disease and progression. The N-terminal domain or ATF binds to a Urokinase receptor (uPAR) on the cell surface and the C-terminal serine protease domain, then, activates plasminogen to plasmin, beginning a cascade of events leading to the progression of cancer. Investigations of urokinase inhibition has been an area of ongoing research for the past 3 decades. It began with the discovery of small natural and unnatural amino acid derivatives or peptide analogs which exhibited weak inhibition of uPA. The last decade has seen the generation of several classes of potent and selective Urokinase inhibitor directed to the serine protease domain of the protein which have shown potential anti-cancer effects. The availability of structural information of enzyme-inhibitor complexes either by nuclear magnetic spectroscopy (NMR) or crystallography has allowed a detailed analysis of inhibitor protein interactions that contribute to observed inhibitor potency. Structural studies of specific inhibitor-uPA complexes will be discussed as well as the contributions of specific inhibitor protein interactions that are important for overall inhibitor potency. These data were used to discover a class of urokinase inhibitor based on the 2-Naphthamidine template that exhibits potent urokinase inhibition and excellent selectivity for urokinase over similar trypsin family serine proteases.


Asunto(s)
Inhibidores de Proteasas/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Animales , Humanos , Modelos Moleculares , Inhibidores de Proteasas/química , Estructura Terciaria de Proteína/efectos de los fármacos , Activador de Plasminógeno de Tipo Uroquinasa/química , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
5.
Neoplasia ; 3(5): 411-9, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11687952

RESUMEN

Checkpoint kinase 1 (Chk1) is a checkpoint gene that is activated after DNA damage. It phosphorylates and inactivates the Cdc2 activating phosphatase Cdc25C. This in turn inactivates Cdc2, which leads to G2/M arrest. We report that blocking Chk1 expression by antisense or ribozymes in mammalian cells induces apoptosis and interferes with the G2/M arrest induced by adriamycin. The Chk1 inhibitor UCN-01 also blocks the G2 arrest after DNA damage and renders cells more susceptible to adriamycin. These results indicate that Chk1 is an essential gene for the checkpoint mechanism during normal cell proliferation as well as in the DNA damage response.


Asunto(s)
Apoptosis , Inhibidores Enzimáticos/farmacología , Fase G2/fisiología , Oxazinas , Inhibidores de Proteínas Quinasas , Xantenos , Alcaloides/farmacología , Antineoplásicos/farmacología , Antineoplásicos Fitogénicos/farmacología , Western Blotting , Caspasas/metabolismo , Ciclo Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Colorantes , Cartilla de ADN/química , ADN sin Sentido/farmacología , Doxorrubicina/farmacología , Resistencia a Medicamentos , Etopósido/farmacología , Citometría de Flujo , Humanos , Neoplasias Pulmonares/metabolismo , Mitosis , Proteínas Quinasas/metabolismo , ARN Catalítico/farmacología , Estaurosporina/análogos & derivados , Células Tumorales Cultivadas
6.
Neoplasia ; 3(4): 278-86, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11571628

RESUMEN

The serine/threonine kinases, Akt1/PKBalpha, Akt2/PKBbeta, and Akt3/PKBgamma, play a critical role in preventing cancer cells from undergoing apoptosis. However, the function of individual Akt isoforms in the tumorigenicity of cancer cells is still not well defined. In the current study, we used an Akt1 antisense oligonucleotide (AS) to specifically downregulate Akt1 protein in both cancer and normal cells. Our data indicate that Akt1 AS treatment inhibits the ability of MiaPaCa-2, H460, HCT-15, and HT1080 cells to grow in soft agar. The treatment also induces apoptosis in these cancer cells as demonstrated by FACS analysis and a caspase activity assay. Conversely, Akt1 AS treatment has little effect on the cell growth and survival of normal human cells including normal human fibroblast (NHF), fibroblast from muscle (FBM), and mammary gland epithelial 184B5 cells. In addition, Akt1 AS specifically sensitizes cancer cells to typical chemotherapeutic agents. Thus, Akt1 is indispensable for maintaining the tumorigenicity of cancer cells. Inhibition of Akt1 may provide a powerful sensitization agent for chemotherapy specifically in cancer cells.


Asunto(s)
Apoptosis , Oxazinas , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas , Células Tumorales Cultivadas/patología , Xantenos , Antineoplásicos/farmacología , Western Blotting , Caspasas/metabolismo , Adhesión Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Colorantes , Grupo Citocromo c/metabolismo , Regulación hacia Abajo , Fibroblastos/citología , Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Oligonucleótidos Antisentido/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Células Tumorales Cultivadas/metabolismo
7.
Antimicrob Agents Chemother ; 45(9): 2563-70, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11502530

RESUMEN

In an effort to discover novel, noncarbohydrate inhibitors of influenza virus neuraminidase we hypothesized that compounds which contain positively charged amino groups in an appropriate position to interact with the Asp 152 or Tyr 406 side chains might be bound tightly by the enzyme. Testing of 300 alpha- and beta-amino acids led to the discovery of two novel neuraminidase inhibitors, a phenylglycine and a pyrrolidine, which exhibited K(i) values in the 50 microM range versus influenza virus A/N2/Tokyo/3/67 neuraminidase but which exhibited weaker activity against influenza virus B/Memphis/3/89 neuraminidase. Limited optimization of the pyrrolidine series resulted in a compound which was about 24-fold more potent than 2-deoxy-2,3-dehydro-N-acetylneuraminic acid in an anti-influenza cell culture assay using A/N2/Victoria/3/75 virus. X-ray structural studies of A/N9 neuraminidase-inhibitor complexes revealed that both classes of inhibitors induced the Glu 278 side chain to undergo a small conformational change, but these compounds did not show time-dependent inhibition. Crystallography also established that the alpha-amino group of the phenylglycine formed hydrogen bonds to the Asp 152 carboxylate as expected. Likewise, the beta-amino group of the pyrrolidine forms an interaction with the Tyr 406 hydroxyl group and represents the first compound known to make an interaction with this absolutely conserved residue. Phenylglycine and pyrrolidine analogs in which the alpha- or beta-amino groups were replaced with hydroxyl groups were 365- and 2,600-fold weaker inhibitors, respectively. These results underscore the importance of the amino group interactions with the Asp 152 and Tyr 406 side chains and have implications for anti-influenza drug design.


Asunto(s)
Aminoácidos/farmacología , Antivirales/farmacología , Glicina/análogos & derivados , Neuraminidasa/antagonistas & inhibidores , Orthomyxoviridae/enzimología , Aminoácidos/química , Antivirales/química , Cristalografía por Rayos X , Glicina/farmacología , Hidroxilación , Modelos Moleculares , Neuraminidasa/química , Orthomyxoviridae/efectos de los fármacos , Conformación Proteica , Pirrolidinas/farmacología
8.
Biochemistry ; 40(31): 9125-31, 2001 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-11478879

RESUMEN

Inhibition of the proteolytic activity of urokinase has been shown to inhibit the progression of tumors in rodent models and is being investigated for use in human disease. Understanding the rodent/human species-specificity of urokinase inhibitors is therefore critical for interpretation of rodent cancer progression models that use these inhibitors. We report here studies with a panel of 11 diverse urokinase inhibitors in both human and mouse enzymatic assays. Inhibitors such as amiloride, B428, and naphthamidine, that occupy only the S1 subsite pocket were found to be nearly equipotent between the human and the murine enzymes. Inhibitors that access additional, more distal, pockets were significantly more potent against the human enzyme but there was no corresponding potency increase against the murine enzyme. X-ray crystallographic structures of these compounds bound to the serine protease domain of human urokinase were solved and examined in order to explain the human/mouse potency differences. The differences in inhibitor potency could be attributed to four amino acid residues that differ between murine and human urokinases: 60, 99, 146, and 192. These residues are Asp, His, Ser, and Gln in human and Gln, Tyr, Glu, and Lys in mouse, respectively. Compounds bearing a cationic group that interacts with residue 60 will preferentially bind to the human enzyme because of favorable electrostatic interactions. The hydrogen bonding to residue 192 and steric considerations with residues 99 and 146 also contribute to the species specificity. The nonparallel human/mouse enzyme inhibition observations were extended to a cell-culture assay of urokinase-activated plasminogen-mediated fibronectin degradation with analogous results. These studies will aid the interpretation of in vivo evaluation of urokinase inhibitors.


Asunto(s)
Amidinas/química , Inhibidores de Serina Proteinasa/química , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Amidinas/síntesis química , Amilorida/síntesis química , Secuencia de Aminoácidos , Animales , Antineoplásicos/síntesis química , Sitios de Unión , Proteínas Sanguíneas , Carcinoma Pulmonar de Lewis , Cristalografía por Rayos X , Humanos , Ratones , Datos de Secuencia Molecular , Naftalenos/síntesis química , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Inhibidores de Serina Proteinasa/síntesis química , Especificidad de la Especie , Tiofenos/síntesis química , Células Tumorales Cultivadas
9.
J Med Chem ; 44(8): 1192-201, 2001 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-11312919

RESUMEN

The discovery of (+/-)-(2S,3R,4R)-2-(trifluoroacetamido)methyl-3-amino-1-(N'-ethyl-N'-isopropylcarbamyl)pyrrolidine-4-carboxylic acid (A-192558, 20e) as a potent inhibitor of influenza neuraminidase (NA) is described. Efficient syntheses of two core structures, cis-3-(allyloxycarbonyl)amino-1-(9'-fluorenylmethoxycarbonyl)pyrrolidine-4-carboxylic acid (7) and tert-butyl (+/-)-(2S,3R,4R)-2-aminomethyl-3-bis(tert-butyloxycarbonyl)amino-1-(N'-ethyl-N'-isopropylcarbamyl)pyrrolidine-4-carboxylate (18b), were developed. Starting with these core structures and using available structural information of the NA active site as the guide, analogues were synthesized in both the tri- and tetrasubstituted pyrrolidine series by means of high-throughput parallel synthesis in solid or solution phase for expeditious SAR. These studies accelerated the identification of (+/-)-(2S,3R,4R)-2-(trifluoroacetamido)methyl-3-amino-1-(N-ethyl-N-isopropylcarbamyl)pyrrolidine-4-carboxylate (20e, A-192558) as the most potent NA inhibitor in this series (IC50 = 0.2 microM against NA A and 8 microM against NA B). The X-ray crystallographic structure of A-192558 bound to NA revealed the predicted interaction of the carboxylic group with the positively charged pocket (Arg118, Arg292, Arg371) and interaction of the trifluoroacetamino residue with the hydrophobic pocket (Ile222, Trp178) of the enzyme active site. Surprisingly, the ethyl and isopropyl groups of the urea functionality induced a conformational change of Glu276, turning the Glu276/Glu277 hydrophilic pocket, which normally accommodates the triglycerol side chain of substrate sialic acid, into an induced hydrophobic pocket.


Asunto(s)
Antivirales/síntesis química , Inhibidores Enzimáticos/síntesis química , Neuraminidasa/antagonistas & inhibidores , Orthomyxoviridae/enzimología , Pirrolidinas/síntesis química , Antivirales/química , Cristalografía por Rayos X , Diseño de Fármacos , Inhibidores Enzimáticos/química , Modelos Moleculares , Estructura Molecular , Pirrolidinas/química
10.
Anticancer Res ; 21(1A): 23-8, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11299740

RESUMEN

BACKGROUND: Chkl is a checkpoint gene that is activated after DNA damage. It phosphorylates and inactivates Cdc25C at the late G2 phase. The inactivation of Cdc25C and consequently, the inactivation of Cdc2, are required for the G2 arrest induced by DNA damage. METHODS: We treated 184B5 cell line and its E6 transformed cell lines with adriamycin in the presence of staurosporine or UCNO1 and examined G2 arrest and cell death. RESULTS: We found that adriamycin induced a p53 and p21 response as well as a G1 arrest in 184B5 cells, but not in its E6 transformed cells. Staurosporine or UCNO1 abrogated the G2 arrest induced by adriamycin in both cell lines. In addition, staurosporine or UCNO1 specifically sensitized p53 incompetent cells to adriamycin. CONCLUSION: G2/M checkpoint abrogators can potentially enhance the cytotoxic effect of conventional chemotherapeutic reagents specifically to tumor cells.


Asunto(s)
Antineoplásicos/farmacología , Daño del ADN/efectos de los fármacos , Doxorrubicina/farmacología , Neoplasias/patología , Proteína p53 Supresora de Tumor/metabolismo , Alcaloides/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Transformada , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Inhibidores Enzimáticos/farmacología , Fase G2 , Humanos , Neoplasias/metabolismo , Proteínas Quinasas/metabolismo , Estaurosporina/farmacología , Células Tumorales Cultivadas
11.
Nat Biotechnol ; 18(10): 1105-8, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11017052

RESUMEN

The need to decrease the time scale for clinical compound discovery has led to innovations at several stages in the process, including genomics/proteomics for target identification, ultrahigh-throughput screening for lead identification, and structure-based drug design and combinatorial chemistry for lead optimization. A critical juncture in the process is the identification of a proper lead compound, because a poor choice may generate costly difficulties at later stages. Lead compounds are commonly identified from high-throughput screens of large compound libraries, derived from known substrates/inhibitors, or identified in computational prescreeusing X-ray crystal structures. Structural information is often consulted to efficiently optimize leads, but under the current paradigm, such data require preidentification and confirmation of compound binding. Here, we describe a new X-ray crystallography-driven screening technique that combines the steps of lead identification, structural assessment, and optimization. The method is rapid, efficient, and high-throughput, and it results in detailed crystallographic structure information. The utility of the method is demonstrated in the discovery and optimization of a new orally available class of urokinase inhibitors for the treatment of cancer.


Asunto(s)
Antineoplásicos/química , Cristalografía por Rayos X , Diseño de Fármacos , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/química , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Administración Oral , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Ligandos , Sustancias Macromoleculares , Conformación Molecular , Naftalenos/química , Naftalenos/farmacología , Quinolinas/química , Quinolinas/farmacología , Relación Estructura-Actividad , Factores de Tiempo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
12.
Eur J Biochem ; 267(13): 4165-70, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10866820

RESUMEN

Cathepsin B and in particular cell-surface and secreted cathepsin B has been implicated in the invasive and metastatic phenotype of numerous types of cancer. We describe here a method to easily survey cancer cell lines for cathepsin B activity using the highly selective substrate Z-Arg-Arg-AMC. Intact human U87 glioma cells hydrolyze Z-Arg-Arg-AMC with a Km of 460 microM at pH 7.0 and 37 degrees C. This is nearly the same as the Km of 430 microM obtained with purified cathepsin B assayed under the same conditions. The pericellular (i.e. both cell-surface and released) cathepsin B activity was inhibited by the cysteine protease inhibitors E-64, leupeptin, Mu-Np2-HphVS-2Np, Mu-Leu-HpHVSPh and the cathepsin B selective inhibitor Mu-Tyr(3,5 I2)-HphVSPh with IC50 values similar to those observed for the inhibition of purified human liver cathepsin B. Other human cancer cell lines with measurable pericellular cathepsin B activity included HT-1080 fibrosarcoma, MiaPaCa pancreatic, PC-3 prostate and HCT-116 colon. Cathepsin B activity correlated with protein levels of cathepsin B as determined by immunoblot analysis. Pericellular cathepsin B activity was also detected in the rat cell lines MatLyLu prostate and Mat B III adenocarcinoma and in the murine lines B16a melanoma and Lewis lung carcinoma. The ability to determine pericellular cathepsin B activity will be useful in selecting appropriate cell lines for use in vivo when analyzing the effects of inhibiting cathepsin B activity on tumor growth and metastasis.


Asunto(s)
Catepsina B/metabolismo , Neoplasias/enzimología , Animales , Catepsina B/antagonistas & inhibidores , Fluorescencia , Humanos , Ratones , Octoxinol/farmacología , Ratas , Células Tumorales Cultivadas
13.
Structure ; 8(5): 553-63, 2000 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-10801494

RESUMEN

BACKGROUND: Human urokinase-type plasminogen activator has been implicated in the regulation and control of basement membrane and interstitial protein degradation. Because of its role in tissue remodeling, urokinase is a central player in the disease progression of cancer, making it an attractive target for design of an anticancer clinical agent: Few urokinase inhibitors have been described, which suggests that discovery of such a compound is in the early stages. Towards integrating structural data into this process, a new human urokinase crystal form amenable to structure-based drug design has been used to discover potent urokinase inhibitors. RESULTS: On the basis of crystallographic data, 2-naphthamidine was chosen as the lead scaffold for structure-directed optimization. This co-crystal structure shows the compound binding at the primary specificity pocket of the trypsin-like protease and at a novel binding subsite that is accessible from the 8-position of 2-napthamidine. This novel subsite was characterized and used to design two compounds with very different 8-substituents that inhibit urokinase with K(i) values of 30-40 nM. CONCLUSIONS: Utilization of a novel subsite yielded two potent urokinase inhibitors even though this site has not been widely used in inhibitor optimization with other trypsin-like proteases, such as those reported for thrombin or factor Xa. The extensive binding pockets present at the substrate-binding groove of these other proteins are blocked by unique insertion loops in urokinase, thus necessitating the utilization of additional binding subsites. Successful implementation of this strategy and characterization of the novel site provides a significant step towards the discovery of an anticancer clinical agent.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/química , Modelos Moleculares , Naftalenos/química , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Activador de Plasminógeno de Tipo Uroquinasa/química , Sitios de Unión/efectos de los fármacos , Cristalografía por Rayos X , Inhibidores Enzimáticos/farmacología , Humanos , Sustancias Macromoleculares , Naftalenos/farmacología , Estructura Terciaria de Proteína/efectos de los fármacos , Especificidad por Sustrato , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
15.
Acta Crystallogr D Biol Crystallogr ; 54(Pt 6 Pt 2): 1367-76, 1998 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-10089498

RESUMEN

The papillomaviruses are a family of small double-stranded DNA viruses which exclusively infect epithelial cells and stimulate the proliferation of those cells. A key protein within the papillomavirus life-cycle is known as the E2 (Early 2) protein and is responsible for regulating viral transcription from all viral promoters as well as for replication of the papillomavirus genome in tandem with another protein known as E1. The E2 protein itself consists of three functional domains: an N-terminal trans-activation domain, a proline-rich linker, and a C-terminal DNA-binding domain. The first crystal structure of the human papillomavirus, serotype 31 (HPV-31), E2 DNA-binding domain has been determined at 2.4 A resolution. The HPV DNA-binding domain monomer consists of two beta-alpha-beta repeats of approximately equal length and is arranged as to have an anti-parallel beta-sheet flanked by the two alpha-helices. The monomers form the functional in vivo dimer by association of the beta-sheets of each monomer so as to form an eight-stranded anti-parallel beta-barrel at the center of the dimer, with the alpha-helices lining the outside of the barrel. The overall structure of HVP-31 E2 DNA-binding domain is similar to both the bovine papillomavirus E2-binding domain and the Epstein-Barr nuclear antigen-1 DNA-binding domain.


Asunto(s)
Proteínas de Unión al ADN/química , Papillomaviridae/química , Conformación Proteica , Proteínas Virales/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Bovinos , Cristalografía por Rayos X , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Datos de Secuencia Molecular , Papillomaviridae/clasificación , Proteínas Recombinantes de Fusión/química , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Serotipificación , Proteínas Virales/metabolismo
16.
Structure ; 4(10): 1205-20, 1996 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-8939746

RESUMEN

BACKGROUND: The over 100 serotypes of human rhinoviruses (HRV) are major causative agents of the common cold in humans. These HRVs can be roughly divided into a major and minor group according to their cellular receptors. They can also be divided into two antiviral groups, A and B, based on their sensitivity to different capsid-binding antiviral compounds. The crystal structures of HRV14 and HRV16, major-receptor group rhinoviruses, as well as HRV1A, a minor-receptor group rhinovirus, were determined previously. Sequence comparisons had shown that HRV14 seemed to be an outlier among rhinoviruses. Furthermore, HRV14 was the only virus with no cellular 'pocket factor' in a hydrophobic pocket which is targeted by many capsid-binding antiviral compounds and is thought to regulate viral stability. HRV3, another major-receptor group virus, was chosen for study because it is one of a subset of serotypes that best represents the drug sensitivity of most rhinovirus serotypes. Both HRV3 and HRV14 belong to antiviral group A, while HRV16 and HRV1A belong to antiviral group B. RESULTS: HRV3 was found to be very similar to HRV14 in sequence and structure. Like HRV14, crystallized HRV3 also has no bound pocket factor. The structure of HRV3 complexed with an antiviral compound, WIN56291, was also determined and found to be similar to the same antiviral compound complexed with HRV14. CONCLUSIONS: The amino-acid sequence and structural similarity between HRV3 and HRV14 suggests that rhinoviruses in the same antiviral group have similar amino-acid sequences and structures. The similar amino-acid composition in the pocket region and the viral protein VP1 N termini in all known group B HRV sequences suggests that these viruses may all contain pocket factors and ordered N-terminal amphipathic helices in VP1. Both of these factors contribute to viral stability, which is consistent with the observations that group B rhinoviruses have a higher chance of successful transmission from one host to another and is a possible explanation for the observed higher pathogenicity of these rhinoviruses.


Asunto(s)
Antígenos Virales/química , Cápside/química , ARN Viral/química , Rhinovirus/química , Antivirales/química , Sitios de Unión , Evolución Biológica , Simulación por Computador , Cristalografía , Humanos , Isoxazoles/química , Modelos Moleculares , Modelos Estructurales , Conformación Molecular , Datos de Secuencia Molecular , Picornaviridae/química , Poliovirus/clasificación , Rhinovirus/clasificación , Análisis de Secuencia de ADN , Especificidad de la Especie , Sincrotrones
17.
Acta Crystallogr D Biol Crystallogr ; 51(Pt 4): 496-503, 1995 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15299836

RESUMEN

Four novel antiviral WIN compounds, that contain a methyl tetrazole ring as well as isoxazole, pyridazine or acetylfuran rings, have had their structures determined in human rhinovirus serotype 14 at 2.9 A resolution. These compounds bind in the VP1 hydrophobic pocket, but are shifted significantly towards the pocket pore when compared to previously examined WIN compounds. A putative water network at the pocket pore is positioned to hydrogen bond with these four WIN compounds, and this network can account for potency differences seen in structurally similar WIN compounds.

18.
J Med Chem ; 37(24): 4177-84, 1994 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-7990117

RESUMEN

As a probe of the 3-methylisoxazole portion of our broad-spectrum antipicornaviral series, a panel of 2-acetylfuran analogues was prepared as replacements for the 3-methylisoxazole ring. Comparison of the two series showed remarkable similarity in potency, spectrum of activity, logP, and electrostatic parameters. X-ray studies of 21b bound to human rhinovirus-14 showed that the 2-acetyl group adopted a syn conformation and the carbonyl oxygen acts as a hydrogen bond acceptor with ASN219 in much the same way as the nitrogen of the isoxazole. The importance of the syn conformation and the hydrogen-bonding capability was confirmed by the reduced antiviral activity of the 2-methylfuran and 2-formylfuran analogues. From the results of this study, it is apparent that the syn-2-acetylfuran ring is acting as a bioisostere for the 3-methylisoxazole.


Asunto(s)
Antivirales/síntesis química , Furanos/síntesis química , Isoxazoles/síntesis química , Rhinovirus/efectos de los fármacos , Antivirales/farmacología , Furanos/farmacología , Humanos , Isoxazoles/farmacología , Conformación Molecular , Relación Estructura-Actividad
20.
J Med Chem ; 36(22): 3240-50, 1993 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-8230114

RESUMEN

A series of tetrazole analogues of Win 54954, a broad-spectrum antipicornavirus compound, has been synthesized to address the acid lability of the oxazoline ring of this series of compounds. The results of X-ray crystallography studies of several members of the oxazoline series bound to human rhinovirus type 1A and 14 have been used to design compounds in the tetrazole series with a broad spectrum of activity. Compound 16b, which has a three-carbon linkage between the isoxazole and phenyl rings and a propyl chain extending from the isoxazole ring, exhibiting an MIC80 for 15 rhinovirus serotypes of 0.20 microM as compared to 0.40 microM for Win 54954. X-ray studies of 16b bound to human rhinovirus-14 show that the propyl side chain extends into a pore in the binding site with the possibility of hydrophobic interactions with a pocket formed by Leu106 and a portion of Ser107.


Asunto(s)
Antivirales/síntesis química , Antivirales/farmacología , Isoxazoles/síntesis química , Isoxazoles/farmacología , Picornaviridae/efectos de los fármacos , Tetrazoles/síntesis química , Tetrazoles/farmacología , Secuencia de Aminoácidos , Sitios de Unión , Humanos , Isoxazoles/química , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular , Rhinovirus/efectos de los fármacos , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA