Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Nat Cancer ; 4(9): 1345-1361, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37743366

RESUMEN

RET receptor tyrosine kinase is activated in various cancers (lung, thyroid, colon and pancreatic, among others) through oncogenic fusions or gain-of-function single-nucleotide variants. Small-molecule RET kinase inhibitors became standard-of-care therapy for advanced malignancies driven by RET. The therapeutic benefit of RET inhibitors is limited, however, by acquired mutations in the drug target as well as brain metastasis, presumably due to inadequate brain penetration. Here, we perform preclinical characterization of vepafestinib (TAS0953/HM06), a next-generation RET inhibitor with a unique binding mode. We demonstrate that vepafestinib has best-in-class selectivity against RET, while exerting activity against commonly reported on-target resistance mutations (variants in RETL730, RETV804 and RETG810), and shows superior pharmacokinetic properties in the brain when compared to currently approved RET drugs. We further show that these properties translate into improved tumor control in an intracranial model of RET-driven cancer. Our results underscore the clinical potential of vepafestinib in treating RET-driven cancers.


Asunto(s)
Neoplasias Encefálicas , Mutación , Encéfalo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Solventes
3.
PLoS One ; 14(2): e0212233, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30779774

RESUMEN

Currently, there is no available therapy to eradicate hepatitis B virus (HBV) in chronically infected individuals. This is due to the difficulty in eliminating viral covalently closed circular (ccc) DNA, which is central to the gene expression and replication of HBV. We developed an assay system for nuclear circular DNA using an integration-deficient lentiviral vector. This vector produced non-integrated circular DNA in nuclei of infected cells. We engineered this vector to encode firefly luciferase to monitor the lentiviral episome DNA. We screened 3,840 chemicals by this assay for luciferase-reducing activity and identified dicumarol, which is known to have anticoagulation activity. We confirmed that dicumarol reduced lentiviral episome DNA. Furthermore, dicumarol inhibited HBV replication in cell culture using NTCP-expressing HepG2 and primary human hepatocytes. Dicumarol reduced intracellular HBV RNA, DNA, supernatant HBV antigens and DNA. We also found that dicumarol reduced the cccDNA level in HBV infected cells, but did not affect HBV adsorption/entry. This is a novel assay system for screening inhibitors targeting nuclear cccDNA and is useful for finding new antiviral substances for HBV.


Asunto(s)
Antivirales/farmacología , Núcleo Celular/metabolismo , ADN Viral/metabolismo , Dicumarol/farmacología , Virus de la Hepatitis B/metabolismo , Plásmidos/metabolismo , Núcleo Celular/genética , Núcleo Celular/virología , ADN Viral/genética , Evaluación Preclínica de Medicamentos , Vectores Genéticos , Células HEK293 , Células Hep G2 , Virus de la Hepatitis B/genética , Humanos , Lentivirus , Plásmidos/genética , ARN Viral/genética , ARN Viral/metabolismo
4.
Int Immunol ; 29(3): 109-120, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28338936

RESUMEN

Hepatitis B virus (HBV) is a virus whose replication cycle cannot be completely reproduced using cultured cell lines. Here, we report an engineered cell line capable of supporting the complete HBV life cycle. We generated HepG2 cells over-expressing the HBV entry receptor human NTCP (sodium taurocholate cotransporting polypeptide), and defective in RIG-I (retinoic acid-inducible gene-I)-like receptor signaling, by knocking down the IPS-1 (IFNß-promoter stimulator-1) adaptor molecule. The resultant NtG20.i7 cells were susceptible to HBV, and its replication was detectable at 14 days post-infection and persisted for at least 35 days with a gradual increase of HBV core expression. The cells produced infectious HBV in the culture supernatant, and the addition of preS1 peptide myr47-WT, which blocks HBV entry, impaired the persistence of the infection. These findings suggest that the persistence of the infection was maintained by continuous release of infectious HBV virions and their re-infection. This system is useful for expanding our basic understanding of the HBV replication cycle and for screening of anti-HBV chemicals.


Asunto(s)
Virus de la Hepatitis B/crecimiento & desarrollo , Hepatocitos/citología , Hepatocitos/virología , Replicación Viral , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteína 58 DEAD Box/genética , Células Hep G2 , Virus de la Hepatitis B/genética , Humanos , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Receptores Inmunológicos , Transducción de Señal/genética , Simportadores/genética
5.
Blood ; 129(10): 1308-1319, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28082445

RESUMEN

p53-related protein kinase (TP53RK, also known as PRPK) is an upstream kinase that phosphorylates (serine residue Ser15) and mediates p53 activity. Here we show that TP53RK confers poor prognosis in multiple myeloma (MM) patients, and, conversely, that TP53RK knockdown inhibits p53 phosphorylation and triggers MM cell apoptosis, associated with downregulation of c-Myc and E2F-1-mediated upregulation of pro-apoptotic Bim. We further demonstrate that TP53RK downregulation also triggers growth inhibition in p53-deficient and p53-mutant MM cell lines and identify novel downstream targets of TP53RK including ribonucleotide reductase-1, telomerase reverse transcriptase, and cyclin-dependent kinase inhibitor 2C. Our previous studies showed that immunomodulatory drugs (IMiDs) downregulate p21 and trigger apoptosis in wild-type-p53 MM.1S cells, Importantly, we demonstrate by pull-down, nuclear magnetic resonance spectroscopy, differential scanning fluorimetry, and isothermal titration calorimetry that IMiDs bind and inhibit TP53RK, with biologic sequelae similar to TP53RK knockdown. Our studies therefore demonstrate that either genetic or pharmacological inhibition of TP53RK triggers MM cell apoptosis via both p53-Myc axis-dependent and axis-independent pathways, validating TP53RK as a novel therapeutic target in patients with poor-prognosis MM.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mieloma Múltiple/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/inmunología , Apoptosis/fisiología , Western Blotting , Técnicas de Silenciamiento del Gen , Humanos , Factores Inmunológicos/farmacología , Mieloma Múltiple/mortalidad , Mieloma Múltiple/patología , Pronóstico , Transducción de Señal/efectos de los fármacos
6.
Invest New Drugs ; 29(4): 534-43, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20084424

RESUMEN

Deregulation of cell-cycle control is a hallmark of cancer. Thus, cyclin-dependent kinases (Cdks) are an attractive target for the development of anti-cancer drugs. Here, we report the biological characterization of a highly potent pan-Cdk inhibitor with a macrocycle-quinoxalinone structure. Compound M inhibited Cdk1, 2, 4, 5, 6, and 9 with equal potency in the nM range and was selective against kinases other than Cdks. This compound inhibited multiple events in the cell cycle in vitro, including retinoblastoma protein (pRb) phosphorylation, E2F-dependent transcription, DNA replication (determined by bromodeoxyuridine incorporation), and mitosis completion (assayed by flow cytometry) in the 10 nM range. Moreover, this compound induced cell death, as determined by induction of the subG1 fraction, activated caspase-3, and anexin V. In vivo, Compound M showed anti-tumor efficacy at a tolerated dose. In a nude rat xenograft tumor model, an 8-h constant infusion of Compound M inhibited pRb phosphorylation and induced apoptosis in tumor cells at ~ 30 nM, which led to the inhibition of tumor growth. Immunosuppression was the only liability observed at this dose, but immune function returned to normal after 10 days. Suppression of pRb phosphorylation in tumor cells was clearly correlated with tumor cell growth inhibition and cell death in vitro and in vivo. In vivo, Compound M inhibited pRb phosphorylation in both tumor and gut crypt cells. Rb phosphorylation may be a suitable pharmacodynamic biomarker in both tumors and normal tissues for monitoring target engagement and predicting the efficacy of Compound M.


Asunto(s)
Antineoplásicos/farmacología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Compuestos Macrocíclicos/farmacología , Quinoxalinas/farmacología , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/química , Bromodesoxiuridina/metabolismo , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quinasas Ciclina-Dependientes/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Células HCT116 , Humanos , Recuento de Leucocitos , Compuestos Macrocíclicos/efectos adversos , Compuestos Macrocíclicos/química , Quinoxalinas/efectos adversos , Quinoxalinas/química , Ratas , Ratas Desnudas , Especificidad por Sustrato/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Invest New Drugs ; 29(5): 921-31, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20524038

RESUMEN

Immunosuppression is one of the common side effects of many anti-tumor agents targeting proliferating cells. We previously reported the development of a new class of pan-cyclin-dependent kinase (Cdk) inhibitor compounds that induce immunosuppression in rodents. Here, we demonstrated that a pan-Cdk inhibitor, Compound 1 very rapidly reduced white blood cells in mice, only 8 h after administration. Compound 1 induced death of peripheral blood cells or purified resting (non-stimulated) lymphocytes ex vivo. Cell death was induced very rapidly, after 4 h of incubation, suggesting that acute immunosuppression observed in rodents might be, at least in part, due to direct cytotoxic effects of Compound 1 on resting lymphocytes. While cell cycle-related Cdks were not activated, the carboxyl terminal domain (CTD) of the largest subunit of RNA polymerase II was phosphorylated, indicating activation of Cdk7 or Cdk9, which phosphorylates this domain, in resting lymphocytes. Indeed, the pan-Cdk inhibitor suppressed CTD phosphorylation in resting cells at the dose required for cell death induction. Inhibition of Cdk7 or Cdk9 by Compound 1 was also confirmed by suppression of nuclear factor-kappa B (NF-κB)-dependent transcription activity in the human cancer cell line U2OS. Interestingly, a Cdk4/6 inhibitor with selectivity against Cdk7 and Cdk9 did not induce cell death in resting lymphocytes. These results suggest that CTD phosphorylation possibly by Cdk7 or Cdk9 might be important for survival of resting lymphocytes and that Cdk inhibitors without inhibitory activity on these kinases might be an attractive agent for cancer chemotherapy.


Asunto(s)
Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Linfocitos/citología , Linfocitos/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Animales , Muerte Celular/efectos de los fármacos , Quinasas Ciclina-Dependientes/metabolismo , Células HEK293 , Humanos , Terapia de Inmunosupresión , Concentración 50 Inhibidora , Leucocitos/citología , Leucocitos/efectos de los fármacos , Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Estructura Terciaria de Proteína , ARN Polimerasa II/química , ARN Polimerasa II/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transcripción Genética/efectos de los fármacos
8.
Acta Crystallogr D Biol Crystallogr ; 66(Pt 5): 577-83, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20445233

RESUMEN

Protein kinase C (PKC) plays an essential role in a wide range of cellular functions. Although crystal structures of the PKC-theta, PKC-iota and PKC-betaII kinase domains have previously been determined in complexes with small-molecule inhibitors, no structure of a PKC-substrate complex has been determined. In the previously determined PKC-iota complex, residues 533-551 in the C-terminal tail were disordered. In the present study, crystal structures of the PKC-iota kinase domain in its ATP-bound and apo forms were determined at 2.1 and 2.0 A resolution, respectively. In the ATP complex, the electron density of all of the C-terminal tail residues was well defined. In the structure, the side chain of Phe543 protrudes into the ATP-binding pocket to make van der Waals interactions with the adenine moiety of ATP; this is also observed in other AGC kinase structures such as binary and ternary substrate complexes of PKA and AKT. In addition to this interaction, the newly defined residues around the turn motif make multiple hydrogen bonds to glycine-rich-loop residues. These interactions reduce the flexibility of the glycine-rich loop, which is organized for ATP binding, and the resulting structure promotes an ATP conformation that is suitable for the subsequent phosphoryl transfer. In the case of the apo form, the structure and interaction mode of the C-terminal tail of PKC-iota are essentially identical to those of the ATP complex. These results indicate that the protein structure is pre-organized before substrate binding to PKC-iota, which is different from the case of the prototypical AGC-branch kinase PKA.


Asunto(s)
Adenosina Trifosfato/metabolismo , Isoenzimas/química , Proteína Quinasa C/química , Adenosina Trifosfato/química , Animales , Línea Celular , Cristalografía por Rayos X , Expresión Génica , Humanos , Isoenzimas/genética , Isoenzimas/aislamiento & purificación , Isoenzimas/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica , Proteína Quinasa C/genética , Proteína Quinasa C/aislamiento & purificación , Proteína Quinasa C/metabolismo
9.
Cell Cycle ; 9(8): 1590-600, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20372067

RESUMEN

Abnormalities in the p16INK4a/ cyclin-dependent kinase (Cdk)4, 6/ Retinoblastoma (Rb) pathway frequently occur in various human cancers. Thus, Cdk4/6 is an attractive target for cancer therapy. Here we report the biological characterization of a 2-aminothiazole-derived Cdk4/6 selective inhibitor, named Compound A in vitro and in vivo. Compound A potently inhibits Cdk4 and Cdk6 with high selectivity (more than 57-fold) against other Cdks and 45 serine/threonine and tyrosine kinases. Compound A inhibits Rb protein (pRb) phosphorylation at Ser780, inhibits E2F-dependent transcription, and induces cell-cycle arrest at G1 in the T98G human glioma cell line. Among 82 human cells derived from various tissues, cell lines derived from hematological cancers (leukemia/lymphoma) tended to be more sensitive to Compound A in cell proliferation assay. Rb-negative cells tended to be insensitive to Compound A, as we had expected. In a nude rat xenograft model, Compound A inhibited pRb phosphorylation and bromodeoxyuridine (BrdU) incorporation in Eol-1 xenograft tumor at plasma concentration of 510 nM. Interestingly Compound A only moderately inhibited those pharmacodynamic and cell cycle parameters of normal crypt cells in small intestine even at 5 times higher plasma concentration. In F344 rats, Compound A did not cause immunosuppression even at 17 times higher plasma conc. These results suggest that Cdk4/6 selective inhibitors only moderately affects on the cell cycle of normal proliferating tissues and has a safer profile than pan-Cdk inhibitor in vivo.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Tiazoles/farmacología , Animales , Línea Celular Tumoral , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Factores de Transcripción E2F/antagonistas & inhibidores , Factores de Transcripción E2F/metabolismo , Fase G1 , Humanos , Masculino , Fosforilación , Inhibidores de Proteínas Quinasas/química , Pirimidinas/química , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Desnudas , Proteína de Retinoblastoma/antagonistas & inhibidores , Proteína de Retinoblastoma/metabolismo , Tiazoles/química , Trasplante Heterólogo
10.
Cancer Biol Ther ; 9(7): 514-22, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20107315

RESUMEN

MK-1775 is a potent and selective small molecule Wee1 inhibitor. Previously we have shown that it abrogated DNA damaged checkpoints induced by gemcitabine, carboplatin, and cisplatin and enhanced the anti-tumor efficacy of these agents selectively in p53-deficient tumor cells. MK-1775 is currently in Phase I clinical trial in combination with these anti-cancer drugs. In this study, the effects of MK-1775 on 5-fluorouracil (5-FU) and other DNA-damaging agents with different modes of action were determined. MK-1775 enhanced the cytotoxic effects of 5-FU in p53-deficient human colon cancer cells. MK-1775 inhibited CDC2 Y15 phosphorylation in cells, abrogated DNA damaged checkpoints induced by 5-FU treatment, and caused premature entry of mitosis determined by induction of Histone H3 phosphorylation. Enhancement by MK-1775 was specific for p53-deficient cells since this compound did not sensitize p53-wild type human colon cancer cells to 5-FU in vitro. In vivo, MK-1775 potentiated the anti-tumor efficacy of 5-FU or its prodrug, capecitabine, at tolerable doses. These enhancements were well correlated with inhibition of CDC2 phosphorylation and induction of Histone H3 phosphorylation in tumors. In addition, MK-1775 also potentiated the cytotoxic effects of pemetrexed, doxorubicin, camptothecin, and mitomycin C in vitro. These studies support the rationale for testing the combination of MK-1775 with various DNA-damaging agents in cancer patients.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Daño del ADN/efectos de los fármacos , Fluorouracilo/farmacología , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Pirimidinas/farmacología , Animales , Antibióticos Antineoplásicos/farmacología , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Camptotecina/farmacología , Capecitabina , Caspasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Doxorrubicina/farmacología , Citometría de Flujo , Fluorouracilo/análogos & derivados , Glutamatos/farmacología , Guanina/análogos & derivados , Guanina/farmacología , Histonas/metabolismo , Humanos , Técnicas para Inmunoenzimas , Mitomicina/farmacología , Proteínas Nucleares/metabolismo , Pemetrexed , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Pirimidinonas , Ratas , Ratas Endogámicas F344 , Ratas Desnudas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Mol Cancer Ther ; 9(1): 157-66, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20053775

RESUMEN

Aurora-A kinase is a one of the key regulators during mitosis progression. Aurora-A kinase is a potential target for anticancer therapies because overexpression of Aurora-A, which is frequently observed in some human cancers, results in aberrant mitosis leading to chromosomal instability and possibly tumorigenesis. MK-5108 is a novel small molecule with potent inhibitory activity against Aurora-A kinase. Although most of the Aurora-kinase inhibitors target both Aurora-A and Aurora-B, MK-5108 specifically inhibited Aurora-A kinase in a panel of protein kinase assays. Inhibition of Aurora-A by MK-5108 in cultured cells induced cell cycle arrest at the G(2)-M phase in flow cytometry analysis. The effect was confirmed by the accumulation of cells with expression of phosphorylated Histone H3 and inhibition of Aurora-A autophosphorylation by immunostaining assays. MK-5108 also induced phosphorylated Histone H3 in skin and xenograft tumor tissues in a nude rat xenograft model. MK-5108 inhibited growth of human tumor cell lines in culture and in different xenograft models. Furthermore, the combination of MK-5108 and docetaxel showed enhanced antitumor activities compared with control and docetaxel alone-treated animals without exacerbating the adverse effects of docetaxel. MK-5108 is currently tested in clinical trials and offers a new therapeutic approach to combat human cancers as a single agent or in combination with existing taxane therapies.


Asunto(s)
Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Ácidos Ciclohexanocarboxílicos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Taxoides/farmacología , Tiazoles/farmacología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Aurora Quinasa A , Aurora Quinasa B , Aurora Quinasas , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ácidos Ciclohexanocarboxílicos/administración & dosificación , Ácidos Ciclohexanocarboxílicos/química , Docetaxel , Humanos , Concentración 50 Inhibidora , Ratones , Mitosis/efectos de los fármacos , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Ratas , Taxoides/toxicidad , Tiazoles/administración & dosificación , Tiazoles/química , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Biol Chem ; 285(7): 4587-94, 2010 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-19864428

RESUMEN

p70 ribosomal S6 kinase (p70S6K) is a downstream effector of the mTOR signaling pathway involved in cell proliferation, cell growth, cell-cycle progression, and glucose homeostasis. Multiple phosphorylation events within the catalytic, autoinhibitory, and hydrophobic motif domains contribute to the regulation of p70S6K. We report the crystal structures of the kinase domain of p70S6K1 bound to staurosporine in both the unphosphorylated state and in the 3'-phosphoinositide-dependent kinase-1-phosphorylated state in which Thr-252 of the activation loop is phosphorylated. Unphosphorylated p70S6K1 exists in two crystal forms, one in which the p70S6K1 kinase domain exists as a monomer and the other as a domain-swapped dimer. The crystal structure of the partially activated kinase domain that is phosphorylated within the activation loop reveals conformational ordering of the activation loop that is consistent with a role in activation. The structures offer insights into the structural basis of the 3'-phosphoinositide-dependent kinase-1-induced activation of p70S6K and provide a platform for the rational structure-guided design of specific p70S6K inhibitors.


Asunto(s)
Proteínas Quinasas S6 Ribosómicas 70-kDa/química , Cromatografía en Gel , Cristalografía por Rayos X , Humanos , Fosforilación , Reacción en Cadena de la Polimerasa , Unión Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Estaurosporina/metabolismo , Ultracentrifugación
13.
Mol Cancer Ther ; 8(11): 2992-3000, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19887545

RESUMEN

Wee1 is a tyrosine kinase that phosphorylates and inactivates CDC2 and is involved in G(2) checkpoint signaling. Because p53 is a key regulator in the G(1) checkpoint, p53-deficient tumors rely only on the G(2) checkpoint after DNA damage. Hence, such tumors are selectively sensitized to DNA-damaging agents by Wee1 inhibition. Here, we report the discovery of a potent and selective small-molecule inhibitor of Wee1 kinase, MK-1775. This compound inhibits phosphorylation of CDC2 at Tyr15 (CDC2Y15), a direct substrate of Wee1 kinase in cells. MK-1775 abrogates G(2) DNA damage checkpoint, leading to apoptosis in combination with DNA-damaging chemotherapeutic agents such as gemcitabine, carboplatin, and cisplatin selectively in p53-deficient cells. In vivo, MK-1775 potentiates tumor growth inhibition by these agents, and cotreatment does not significantly increase toxicity. The enhancement of antitumor effect by MK-1775 was well correlated with inhibition of CDC2Y15 phosphorylation in tumor tissue and skin hair follicles. Our data indicate that Wee1 inhibition provides a new approach for treatment of multiple human malignancies.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Daño del ADN , Neoplasias/tratamiento farmacológico , Proteínas Nucleares/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Pirimidinas/farmacología , Proteína p53 Supresora de Tumor/deficiencia , Animales , Apoptosis/efectos de los fármacos , Proteína Quinasa CDC2 , Línea Celular Tumoral , Ciclina B/metabolismo , Quinasas Ciclina-Dependientes , Sinergismo Farmacológico , Citometría de Flujo , Células HeLa , Humanos , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/patología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Pirimidinonas , Ratas , Ratas Endogámicas F344 , Ratas Desnudas , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Bioorg Med Chem Lett ; 19(16): 4673-8, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19589677
15.
J Med Chem ; 52(14): 4091-4, 2009 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-19537798

RESUMEN

Our efforts to optimize prototype opioid receptor-like 1 (ORL1) antagonist 1 led to the discovery of 4-{3-[(2R)-2,3-dihydroxypropyl]-2-oxo-2,3-dihydro-1H-benzimidazol-1-yl}-1-[(1S,3S,4R)-spiro[bicyclo[2.2.1]heptane-2,1'-cyclopropan]-3-ylmethyl]piperidine 10. 10 showed potent ORL1 antagonistic activity, excellent selectivity over other opioid receptors, and in vivo efficacy after oral dosing. Currently clinical trials of 10 are underway.


Asunto(s)
Bencimidazoles/administración & dosificación , Bencimidazoles/farmacología , Antagonistas de Narcóticos , Piperidinas/administración & dosificación , Piperidinas/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Administración Oral , Animales , Bencimidazoles/metabolismo , Bencimidazoles/farmacocinética , Células CHO , Cricetinae , Cricetulus , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Concentración 50 Inhibidora , Ratones , Piperidinas/metabolismo , Piperidinas/farmacocinética , Ratas , Receptores Opioides/metabolismo , Relación Estructura-Actividad , Receptor de Nociceptina
16.
Bioorg Med Chem Lett ; 18(11): 3278-81, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18468891

RESUMEN

Structure-activity studies on benzimidazole lead 1 obtained from library screening led to the discovery of potent and selective ORL1 antagonist 28, 5-chloro-2-[(1-ethyl-1-methylpropyl)thio]-6-[4-(2-hydroxyethyl)piperazin-1-yl]-1H-benzimidazole, which is structurally distinct from conventional non-peptide antagonists known to date.


Asunto(s)
Bencimidazoles/síntesis química , Bencimidazoles/farmacología , Antagonistas de Narcóticos , Bencimidazoles/química , Técnicas Químicas Combinatorias , Humanos , Estructura Molecular , Receptores Opioides , Relación Estructura-Actividad , Receptor de Nociceptina
17.
Bioorg Med Chem Lett ; 16(14): 3751-4, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16682184

RESUMEN

5-Pyrimidinyl-2-aminothiazole 1 was identified as an inhibitor of cyclin-dependent kinases (CDKs) by a screening of the Merck sample repository. The introduction of a methyl group at the C-5 or C-6 position on the pyrimidine ring, directed toward the gate keeper residue of CDK4 (Phe93), led to significant enhancement of selectivity for CDK4 over other CDKs. Compound 3 exhibited more than 300-fold selectivity for CDK4 over CDK1, 2, 5, 7, and 9. Subsequent improvements in aqueous solubility afforded compound 4, which is available for further in vivo studies and this compound inhibited pRb phosphorylation and BrdU incorporation in tumor models.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Pirimidinas/farmacología , Tiazoles/farmacología , Bromodesoxiuridina/antagonistas & inhibidores , Bromodesoxiuridina/metabolismo , Proteína Quinasa CDC2/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Humanos , Fosforilación , Proteína de Retinoblastoma/antagonistas & inhibidores , Proteína de Retinoblastoma/metabolismo , Células Tumorales Cultivadas , Quinasa Activadora de Quinasas Ciclina-Dependientes
18.
J Med Chem ; 49(3): 847-9, 2006 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-16451050

RESUMEN

A focused library approach identifying novel leads to develop a potent ORL1 antagonist is described. Beginning from a compound identified by random screening, an exploratory library that exhibited a diverse display of pharmacophores was designed. After evaluating ORL1 antagonistic activity, a highly focused library was designed based on 3D-pharmacophore similarity to known actives. A novel D-proline amide class was identified in this library and was found to possess potent ORL1 antagonistic activity.


Asunto(s)
Antagonistas de Narcóticos , Piperidinas/síntesis química , Receptores Opioides/química , Compuestos de Espiro/síntesis química , Animales , Unión Competitiva , Células CHO , Técnicas Químicas Combinatorias , Cricetinae , Cricetulus , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Estructura Molecular , Piperidinas/química , Prolina/análogos & derivados , Prolina/síntesis química , Prolina/química , Prolina/farmacología , Relación Estructura-Actividad Cuantitativa , Ensayo de Unión Radioligante , Receptores Opioides/agonistas , Compuestos de Espiro/química , Compuestos de Espiro/farmacología , Estereoisomerismo , Receptor de Nociceptina
19.
Curr Top Med Chem ; 5(2): 167-79, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15853645

RESUMEN

Loss of normal cell cycle regulation is the hallmark of human cancers, and alteration of the components involved in cell cycle regulation occurs in most human tumors. This suggests that Cyclin dependent kinases (CDKs) are an attractive target for the development of pharmacological agents for the treatment of cancer. Recently, CDK family members that are not directly involved in cell cycle regulation have been identified. This includes CDK7, CDK8, and CDK9, which participate in transcription regulation, and CDK5, which plays a role in neuronal and secretory functions. Given the involvement of CDKs in multiple cellular processes, development of selective small molecule inhibitors for specific CDKs is expected to help clarify whether improved specificity of cell cycle CDK inhibitors will enhance their therapeutic potential in cancer treatment. Selective inhibitors are also needed as tools to explore the biology of diseases in which CDKs may participate and to help develop therapeutics to treat them. Intensive screening and drug design based on CDK/inhibitor co-crystal structure and SAR studies have led to the identification of a large variety of chemical inhibitors of CDKs. Although they are competitive with ATP at the catalytic site, their kinase selectivity varies greatly, and inhibitors selective for certain CDKs have begun to be identified. There are currently two categories of selective CDK inhibitors: those that are selective for CDK2 and CDK1 and those that are selective for CDK4/6. These two types of inhibitors have different effects on tumor cells and are expected to be useful in the treatment of cancer.


Asunto(s)
Antineoplásicos/farmacología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Animales , Antineoplásicos/química , Quinasas CDC2-CDC28/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Quinasa 6 Dependiente de la Ciclina , Inhibidores Enzimáticos/química , Humanos , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...