Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cell ; 165(3): 643-55, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27104980

RESUMEN

Oncogenic activation of RAS genes via point mutations occurs in 20%-30% of human cancers. The development of effective RAS inhibitors has been challenging, necessitating new approaches to inhibit this oncogenic protein. Functional studies have shown that the switch region of RAS interacts with a large number of effector proteins containing a common RAS-binding domain (RBD). Because RBD-mediated interactions are essential for RAS signaling, blocking RBD association with small molecules constitutes an attractive therapeutic approach. Here, we present evidence that rigosertib, a styryl-benzyl sulfone, acts as a RAS-mimetic and interacts with the RBDs of RAF kinases, resulting in their inability to bind to RAS, disruption of RAF activation, and inhibition of the RAS-RAF-MEK pathway. We also find that ribosertib binds to the RBDs of Ral-GDS and PI3Ks. These results suggest that targeting of RBDs across multiple signaling pathways by rigosertib may represent an effective strategy for inactivation of RAS signaling.


Asunto(s)
Glicina/análogos & derivados , Proteínas de Unión al ARN/química , Transducción de Señal/efectos de los fármacos , Sulfonas/farmacología , Secuencia de Aminoácidos , Animales , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Transformación Celular Neoplásica/efectos de los fármacos , Cristalografía por Rayos X , Dimerización , Glicina/administración & dosificación , Glicina/química , Glicina/farmacología , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Desnudos , Modelos Moleculares , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Neoplasias Pancreáticas/tratamiento farmacológico , Fosforilación , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas B-raf/química , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas de Unión al ARN/metabolismo , Alineación de Secuencia , Sulfonas/administración & dosificación , Sulfonas/química , Proteínas ras/metabolismo , Quinasa Tipo Polo 1
2.
Mol Cell ; 79(1): 180-190.e4, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32619468

RESUMEN

Rigosertib is a styryl benzyl sulfone that inhibits growth of tumor cells and acts as a RAS mimetic by binding to Ras binding domains of RAS effectors. A recent study attributed rigosertib's mechanism of action to microtubule binding. In that study, rigosertib was obtained from a commercial vendor. We compared the purity of clinical-grade and commercially sourced rigosertib and found that commercially sourced rigosertib contains approximately 5% ON01500, a potent inhibitor of tubulin polymerization. Clinical-grade rigosertib, which is free of this impurity, does not exhibit tubulin-binding activity. Cell lines expressing mutant ß-tubulin have also been reported to be resistant to rigosertib. However, our study showed that these cells failed to proliferate in the presence of rigosertib at concentrations that are lethal to wild-type cells. Rigosertib induced a senescence-like phenotype in the small percentage of surviving cells, which could be incorrectly scored as resistant using short-term cultures.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular , Glicina/análogos & derivados , Neoplasias Pulmonares/patología , Sulfonas/farmacología , Tubulina (Proteína)/metabolismo , Contaminación de Medicamentos , Resistencia a Antineoplásicos , Glicina/farmacología , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Mutación , Tubulina (Proteína)/química , Tubulina (Proteína)/genética , Células Tumorales Cultivadas
3.
Bioorg Med Chem ; 24(4): 521-44, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26762835

RESUMEN

Several families of protein kinases have been shown to play a critical role in the regulation of cell cycle progression, particularly progression through mitosis. These kinase families include the Aurora kinases, the Mps1 gene product and the Polo Like family of protein kinases (PLKs). The PLK family consists of five members and of these, the role of PLK1 in human cancer is well documented. PLK2 (SNK), which is highly homologous to PLK1, has been shown to play a critical role in centriole duplication and is also believed to play a regulatory role in the survival pathway by physically stabilizing the TSC1/2 complex in tumor cells under hypoxic conditions. As a part of our research program, we have developed a library of novel ATP mimetic chemotypes that are cytotoxic against a panel of cancer cell lines. We show that one of these chemotypes, the 6-arylsulfonyl pyridopyrimidinones, induces apoptosis of human tumor cell lines in nanomolar concentrations. The most potent of these compounds, 7ao, was found to be a highly specific inhibitor of PLK2 when profiled against a panel of 288 wild type, 55 mutant and 12 lipid kinases. Here, we describe the synthesis, structure activity relationship, in vitro kinase specificity and biological activity of the lead compound, 7ao.


Asunto(s)
Descubrimiento de Drogas , Indoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirimidinonas/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Indoles/síntesis química , Indoles/química , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Pirimidinonas/síntesis química , Pirimidinonas/química , Relación Estructura-Actividad
4.
Org Biomol Chem ; 11(12): 1964-77, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23386308

RESUMEN

A stereoselective and efficient method for free radical addition of benzyl thiol to aryl acetylene in the presence of Et3B-hexane has been developed for the synthesis of (Z) and (E)-styryl benzyl sulfides where base catalyzed hydrothiolations have failed. The scope of this reaction was successfully extended for the synthesis of (E)-ON 01910·Na, a phase III clinical stage anti-cancer agent and its inactive geometrical isomer (Z)-ON 01910·Na. It is interesting to note that all the E-isomers synthesized have shown better cytotoxicity profile on cancer cells compared to the Z-isomers.


Asunto(s)
Alquinos/farmacología , Antineoplásicos/farmacología , Glicina/análogos & derivados , Compuestos de Sulfhidrilo/química , Sulfonas/farmacología , Alquinos/síntesis química , Alquinos/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensayos Clínicos Fase III como Asunto , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Glicina/síntesis química , Glicina/química , Glicina/farmacología , Humanos , Células K562 , Estructura Molecular , Estereoisomerismo , Relación Estructura-Actividad , Sulfonas/síntesis química , Sulfonas/química
5.
Cancer Cell ; 7(3): 275-86, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15766665

RESUMEN

Elevated expression of polo-like kinase1 (Plk1) has been reported in many human tumors, and inhibition of Plk1 activity results in their mitotic arrest and apoptosis. Here we describe the profile of ON01910, a small molecule inhibitor of Plk1 activity, which induces mitotic arrest of tumor cells characterized by spindle abnormalities leading to their apoptosis. This compound was not ATP-competitive, but competed for the substrate binding site of the enzyme. In vivo, this compound did not exhibit hematotoxicity, liver damage, or neurotoxicity, and was a potent inhibitor of tumor growth in a variety of xenograft nude mouse models. ON01910 showed strong synergy with several chemotherapeutic agents, often inducing complete regression of tumors.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Ciclo Celular/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Huso Acromático/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/toxicidad , Apoptosis , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Ratones , Ratones Desnudos , Estructura Molecular , Neoplasias Experimentales/metabolismo , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/toxicidad , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/genética , Huso Acromático/metabolismo , Quinasa Tipo Polo 1
6.
Nat Commun ; 12(1): 4671, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34344863

RESUMEN

Triple negative breast cancer (TNBC) remains challenging because of heterogeneous responses to chemotherapy. Incomplete response is associated with a greater risk of metastatic progression. Therefore, treatments that target chemotherapy-resistant TNBC and enhance chemosensitivity would improve outcomes for these high-risk patients. Breast cancer stem cell-like cells (BCSCs) have been proposed to represent a chemotherapy-resistant subpopulation responsible for tumor initiation, progression and metastases. Targeting this population could lead to improved TNBC disease control. Here, we describe a novel multi-kinase inhibitor, 108600, that targets the TNBC BCSC population. 108600 treatment suppresses growth, colony and mammosphere forming capacity of BCSCs and induces G2M arrest and apoptosis of TNBC cells. In vivo, 108600 treatment of mice bearing triple negative tumors results in the induction of apoptosis and overcomes chemotherapy resistance. Finally, treatment with 108600 and chemotherapy suppresses growth of pre-established TNBC metastases, providing additional support for the clinical translation of this agent to clinical trials.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Nitrobencenos/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Tiazinas/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Quinasa de la Caseína II/antagonistas & inhibidores , Quinasa de la Caseína II/química , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Células Madre Neoplásicas/patología , Nitrobencenos/química , Nitrobencenos/farmacología , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/química , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/química , Tiazinas/química , Tiazinas/farmacología , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasas DyrK
7.
Bioorg Med Chem ; 18(6): 2317-2326, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20188579

RESUMEN

Novel (E)-alpha-benzylthio chalcones are reported with preliminary in vitro activity data indicating that several of them are potent inhibitors (comparable to imatinib, the reference compound) of BCR-ABL phosphorylation in leukemic K562 cells, known to express high levels of BCR-ABL. The ability of such compounds to significantly inhibit K562 cell proliferation suggests that this scaffold could be a promising lead for the development of anticancer agents that are able to block BCR-ABL phosphorylation in leukemic cells.


Asunto(s)
Chalconas/síntesis química , Chalconas/farmacología , Diseño de Fármacos , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Chalconas/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Células K562 , Estructura Molecular , Fosforilación , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Proteínas Tirosina Quinasas/metabolismo , Estereoisomerismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
8.
Oncotarget ; 10(20): 1932-1942, 2019 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-30956775

RESUMEN

Aberrant signaling triggered by oncogenic or hyperactive RAS proteins contributes to the malignant phenotypes in a significant percentage of myeloid malignancies. Of these, juvenile myelomonocytic leukemia (JMML), an aggressive childhood cancer, is largely driven by mutations in RAS genes and those that encode regulators of these proteins. The Mx1-cre kras+/G12D mouse model mirrors several key features of this disease and has been used extensively to determine the utility and mechanism of small molecule therapeutics in the context of RAS-driven myeloproliferative disorders. Treatment of disease-bearing KRASG12D mice with rigosertib (RGS), a small molecule RAS mimetic that is in phase II and III clinical trials for MDS and AML, decreased the severity of leukocytosis and splenomegaly and extended their survival. RGS also increased the frequency of HSCs and rebalanced the ratios of myeloid progenitors. Further analysis of KRASG12D HSPCs in vitro revealed that RGS suppressed hyperproliferation in response to GM-CSF and inhibited the phosphorylation of key RAS effectors. Together, these data suggest that RGS might be of clinical benefit in RAS-driven myeloid disorders.

9.
J Med Chem ; 51(1): 86-100, 2008 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-18088089

RESUMEN

Cell cycle progression is regulated by cyclins and cyclin-dependent kinases, which are formed at specific stages of the cell cycle and regulate the G1/S and G2/M phase transitions, employing a series of "checkpoints" governed by phosphorylation of their substrates. Tumor development is associated with the loss of these checkpoint controls, and this provides an approach for the development of therapeutic agents that can specifically target tumor cells. Here, we describe the synthesis and SAR of a novel group of cytotoxic molecules that selectively induce growth arrest of normal cells in the G1 phase while inducing a mitotic arrest of tumor cells resulting in selective killing of tumor cell populations with little or no effect on normal cell viability. The broad spectrum of antitumor activity in vitro and xenograft models, lack of in vivo toxicity, and drug resistance suggest potential for use of these agents in cancer therapy.


Asunto(s)
Antineoplásicos/síntesis química , Estirenos/síntesis química , Sulfonas/síntesis química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Diseño de Fármacos , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Ratones , Ratones Desnudos , Ratas , Estereoisomerismo , Relación Estructura-Actividad , Estirenos/química , Estirenos/farmacología , Sulfonas/química , Sulfonas/farmacología , Pruebas de Toxicidad
10.
Oncotarget ; 9(102): 37753-37765, 2018 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-30701029

RESUMEN

Overexpression and constitutive activation of CYCLIN D1 and Casein Kinase 2 are common features of many hematologic malignancies, including mantle cell lymphoma (MCL) and leukemias such as T-cell acute lymphoblastic leukemia (T-ALL). Although both CK2 and CDK4 inhibitors have shown promising results against these tumor types, none of these agents have achieved objective responses in the clinic as monotherapies. Because both proteins play key roles in these and other hematological malignancies, we have analyzed the therapeutic potential of ON108110, a novel dual specificity ATP-competitive inhibitor of protein kinase CK2 as well as CDK4/6 in MCL and T-ALL. We show that in cell growth inhibition assays, MCL and T-ALL cell lines exhibited increased sensitivity to ON108110 when compared to other tumor types. Treatment with ON108110 reduced the level of phosphorylated RB-family proteins. In addition, ON108110 treatment resulted in concentration dependent inhibition of PTEN phosphorylation and a concomitant decrease in PI3K-AKT signaling mediated by CK2. Accordingly, cells treated with ON108110 rapidly accumulated in the G0/G1 stage of the cell cycle as a function of increasing concentration followed by rapid onset of apoptosis. Together, these results indicate that dual inhibition of CK2 and CDK4/6 may be an efficient treatment of MCL and T-ALLs displaying upregulation of CK2/PI3K and CDK4 signaling pathways.

11.
Mol Cell Biol ; 22(2): 644-56, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11756559

RESUMEN

Mutations in CDK4 and its key kinase inhibitor p16(INK4a) have been implicated in the genesis and progression of familial human melanoma. The importance of the CDK4 locus in human cancer first became evident following the identification of a germ line CDK4-Arg24Cys (R24C) mutation, which abolishes the ability of CDK4 to bind to p16(INK4a). To determine the role of the Cdk4(R24C) germ line mutation in the genesis of other cancer types, we introduced the R24C mutation in the Cdk4 locus of mice by using Cre-loxP-mediated "knock-in" technology. Cdk4(R24C/R24C) mouse embryo fibroblasts (MEFs) displayed increased Cdk4 kinase activity resulting in hyperphosphorylation of all three members of the Rb family, pRb, p107, and p130. MEFs derived from Cdk4(R24C/R24C) mice displayed decreased doubling times, escape from replicative senescence, and escape sensitivity to contact-induced growth arrest. These MEFs also exhibited a high degree of susceptibility to oncogene-induced transformation, suggesting that the Cdk4(R24C) mutation can serve as a primary event in the progression towards a fully transformed phenotype. In agreement with the in vitro data, homozygous Cdk4(R24C/R24C) mice developed tumors of various etiology within 8 to 10 months of their life span. The majority of these tumors were found in the pancreas, pituitary, brain, mammary tissue, and skin. In addition, Cdk4(R24C/R24C) mice showed extraordinary susceptibility to carcinogens and developed papillomas within the first 8 to 10 weeks following cutaneous application of the carcinogens 9,10-di-methyl-1,2-benz[a]anthracene (DMBA) and 12-O-tetradecanoylphorbol-13-acetate (TPA). This report formally establishes that the activation of Cdk4 is sufficient to promote cancer in many tissues. The observation that a wide variety of tumors develop in mice harboring the Cdk4(R24C) mutation offers a genetic proof that Cdk4 activation may constitute a central event in the genesis of many types of cancers in addition to melanoma.


Asunto(s)
Senescencia Celular/genética , Quinasas Ciclina-Dependientes/genética , Mutación de Línea Germinal , Neoplasias Experimentales/genética , Proteínas Proto-Oncogénicas , Animales , Ciclo Celular/genética , División Celular/genética , Transformación Celular Neoplásica/genética , Cocarcinogénesis , Quinasa 4 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Genes ras , Humanos , Ratones , Ratones Mutantes , Neoplasias Experimentales/etiología , Neoplasias Experimentales/patología , Oncogenes , Fenotipo , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/genética
12.
Nucl Med Biol ; 34(4): 371-81, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17499726

RESUMEN

UNLABELLED: Disintegrins, which contain an Arg-Gly-Asp sequence in their binding domains are antagonists of integrins such as alphavbeta3. The purpose of this study was to compare a range of disintegrins with different integrin selectivities for their binding behavior in vitro to vascular endothelial cells bearing alphavbeta3 and to cultured tumor cells which express alphavbeta3. METHODS: Five disintegrins (bitistatin, kistrin, flavoridin, VLO4 and echistatin) and a cyclic pentapeptide, c[RGDyK], were radiolabeled with (99m)Tc and tested for binding to cells in vitro. RESULTS: (99m)Tc-Kistrin, flavoridin and VLO4 had the highest binding, (99m)Tc-echistatin had moderate binding, and (99m)Tc-bitistatin and (99m)Tc-c[RGDyK] had low binding to cells. The observed binding was attributed to alphavbeta3 to various extents: echistatin, bitistatin>kistrin>flavoridin>VLO4. Cancer cells internalized bound disintegrins after binding, but endothelial cells did not. After binding to endothelial cells, (99m)Tc-kistrin was not displaced by competing peptide or plasma proteins. CONCLUSIONS: These data suggest that radiolabeled kistrin, flavoridin and VLO4 may have advantages over labeled bitistatin and small cyclic peptides for targeting alphavbeta3 in vivo. Since receptor-bound radioligand is not internalized by endothelial cells, disintegrins may provide an advantage for targeting alphavbeta3 on vasculature because they bind strongly to surface receptors and are not readily displaced.


Asunto(s)
Desintegrinas/síntesis química , Integrina alfaVbeta3/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Compuestos de Organotecnecio/síntesis química , Radiofármacos , Unión Competitiva/efectos de los fármacos , Línea Celular Tumoral , Desintegrinas/farmacocinética , Células Endoteliales/metabolismo , Humanos , Marcaje Isotópico , Ligandos , Compuestos de Organotecnecio/farmacocinética , Ensayo de Unión Radioligante , Radiofármacos/farmacocinética , Receptores de Superficie Celular/metabolismo
13.
Eur J Cancer ; 50(11): 1982-92, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24835034

RESUMEN

Heat shock protein (Hsp) 90 is a key component of the super-chaperone complex that maintains functionally active conformation of various client proteins. Many of these client proteins regulate important nodal points in multiple signalling pathways that promote cancer cell growth and survival. Inhibitors of Hsp90, therefore, have the potential of functioning as anti-cancer agents with pleiotropic effects. Identification of novel Hsp90 inhibitors with more favourable pharmacological properties is a priority in cancer therapy. To achieve this goal, we screened a compound library using a biochemical assay based on refolding of denatured firefly luciferase. The assay revealed high sensitivity, reliability and reproducibility with a Z-factor of 0.81 ± 0.17. Six Hsp90 inhibitory compounds identified by this screening with IC50 values between 1.0 and 6 µM were further characterised for anti-proliferative activity by Cell Titer-Blue Cell Viability Assay using multiple tumour cell lines. Of particular interest was ONO4140 with lowest GI50 values in three different cancer cell lines viz; DU-145, BT-474 and K562 cell lines. This study also revealed that short-term exposure of tumour cells with ONO4140 is sufficient to inhibit the catalytic activity of Hsp90, evaluated through disruption of Hsp90-p23 association by immunoprecipitation. This short term exposure appears to initiate events like degradation of Hsp90 client proteins such as ErbB2/Her-2 and Akt with concomitant inhibition of survival signalling leading to the apoptotic death of tumour cells as seen by western blotting and Caspase Glow-3,7 assay. The study also reveals that apoptosis following Hsp90 inhibition with ONO4140 occurs via Caspase9-Caspase3 intrinsic apoptotic pathway, a process that is likely triggered by inactivation of Akt. In conclusion, we have identified a novel class of synthetic compounds which show potent Hsp90 inhibitory action in preclinical studies. The discovery of this novel class of synthetic Hsp90 inhibitors with simple chemical backbone allows us to conduct further structural modifications to improve their potency and pharmacokinetic properties for use in cancer therapy.


Asunto(s)
Chalconas/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Sulfonas/farmacología , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Proteínas HSP90 de Choque Térmico/química , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Células K562 , Luciferasas de Luciérnaga/química , Luciferasas de Luciérnaga/metabolismo , Conejos , Transducción de Señal
14.
J Med Chem ; 57(3): 578-99, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24417566

RESUMEN

The success of imatinib, a BCR-ABL inhibitor for the treatment of chronic myelogenous leukemia, has created a great impetus for the development of additional kinase inhibitors as therapeutic agents. However, the complexity of cancer has led to recent interest in polypharmacological approaches for developing multikinase inhibitors with low toxicity profiles. With this goal in mind, we analyzed more than 150 novel cyano pyridopyrimidine compounds and identified structure-activity relationship trends that can be exploited in the design of potent kinase inhibitors. One compound, 8-cyclopentyl-2-[4-(4-methyl-piperazin-1-yl)-phenylamino]-7-oxo-7,8-dihydro-pyrido[2,3-d]pyrimidine-6-carbonitrile (7x), was found to be the most active, inducing apoptosis of tumor cells at a concentration of approximately 30-100 nM. In vitro kinase profiling revealed that 7x is a multikinase inhibitor with potent inhibitory activity against the CDK4/CYCLIN D1 and ARK5 kinases. Here, we report the synthesis, structure-activity relationship, kinase inhibitory profile, in vitro cytotoxicity, and in vivo tumor regression studies by this lead compound.


Asunto(s)
Antineoplásicos/síntesis química , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Piridinas/síntesis química , Pirimidinas/síntesis química , Proteínas Represoras/antagonistas & inhibidores , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Simulación del Acoplamiento Molecular , Trasplante de Neoplasias , Proteínas Quinasas , Piridinas/química , Piridinas/farmacología , Pirimidinas/química , Pirimidinas/farmacología , Relación Estructura-Actividad
15.
PLoS One ; 8(3): e58355, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23505494

RESUMEN

Development of radio-protective agents that are non-toxic is critical in light of ever increasing threats associated with proliferation of nuclear materials, terrorism and occupational risks associated with medical and space exploration. In this communication, we describe the discovery, characterization and mechanism of action of ON01210.Na, which effectively protects mouse and human bone marrow cells from radiation-induced damage both in vitro and in vivo. Our results show that treatment of normal fibroblasts with ON01210.Na before and after exposure to ionizing radiation provides dose dependent protection against radiation-induced damage. Treatment of mice with ON01210.Na prior to radiation exposure was found to result in a more rapid recovery of their hematopoietic system. The mechanistic studies described here show that ON01210.Na manifests its protective effects through the up-regulation of PI3-Kinase/AKT pathways in cells exposed to radiation. These results suggest that ON 01210.Na is a safe and effective radioprotectant and could be a novel agent for use in radiobiological disasters.


Asunto(s)
Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Protectores contra Radiación/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Sulfonamidas/farmacología , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/efectos de la radiación , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de la radiación , Humanos , Ratones , Bibliotecas de Moléculas Pequeñas
16.
J Pharm Biomed Anal ; 75: 138-44, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23261806

RESUMEN

ON 013100, (E)-2,4,6-trimethoxystyryl-3-hydroxy-4-methoxybenzyl sulfone, is a potent kinase inhibitor whose phosphate form is in Phase I clinical trials in lymphoma and acute lymphoid leukemia. The objectives were to: (a) investigate the possible presence of the glucuronide metabolite of the drug in two representative colon cancer cell lines, a drug resistant (colo-205) and a drug sensitive (colo-320); (b) quantify the glucuronide metabolite and the unchanged drug in the cells after treatment with ON 013100. The glucuronide was synthesized and a selective LC/MS/MS method was developed and validated for the characterization and quantification of the metabolite. The glucuronide metabolite (570.6 Da) was found in the drug-resistant cells upon a 1h incubation with ON 013100 (20 µg/ml). After treatment with the drug, the concentration of the metabolite gradually decreased from 0.84 µg/ml at 0 h through 0.21 µg/ml at 6h to below detection limit of 8.0 ng/ml at 9 h. No glucuronide metabolite was detected in the drug-sensitive cells. The concentrations of intact ON 013100 in the drug-resistant cells gradually decreased from 0.41 µg/ml (0 h) to 0.06 µg/ml (9 h). The corresponding concentrations of the intact drug in the drug-sensitive cells were from 2.88 µg/ml to 0.94 µg/ml.


Asunto(s)
Antineoplásicos/metabolismo , Compuestos de Bencilo/metabolismo , Neoplasias del Colon/metabolismo , Glucurónidos/análisis , Inhibidores de Proteínas Quinasas/metabolismo , Estirenos/metabolismo , Antineoplásicos/análisis , Antineoplásicos/farmacología , Compuestos de Bencilo/análisis , Compuestos de Bencilo/farmacología , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Neoplasias del Colon/tratamiento farmacológico , Resistencia a Antineoplásicos , Glucurónidos/química , Glucurónidos/metabolismo , Humanos , Cinética , Límite de Detección , Fase I de la Desintoxicación Metabólica , Peso Molecular , Inhibidores de Proteínas Quinasas/análisis , Inhibidores de Proteínas Quinasas/farmacología , Espectrometría de Masa por Ionización de Electrospray , Estereoisomerismo , Estirenos/análisis , Estirenos/farmacología , Sulfonas , Espectrometría de Masas en Tándem
17.
J Med Chem ; 56(13): 5562-86, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23750455

RESUMEN

A series of novel (E)-N-aryl-2-arylethenesulfonamides (6) were synthesized and evaluated for their anticancer activity. Some of the compounds in this series showed potent cytotoxicity against a wide spectrum of cancer cell-lines (IC50 values ranging from 5 to 10 nM) including all drug resistant cell-lines. Nude mice xenograft assays with compound (E)-N-(3-amino-4-methoxyphenyl)-2-(2',4',6'-trimethoxyphenyl)ethenesulfonamide (6t) showed dramatic reduction in tumor size, indicating their in vivo potential as anticancer agents. A preliminary drug development study with compound 6t is predicted to have increased blood-brain barrier permeability relative to many clinically used antimitotic agents. Mechanistic studies indicate that 6t and some other analogues disrupted microtubule formation, formation of mitotic spindles, and arrest of cells in mitotic phase. Compound 6t inhibited purified tubulin polymerization in vitro and in vivo and circumvented drug resistance mediated by P-glycoprotein. Compound 6t specifically competed with colchicine binding to tubulin and with similar avidity as podophylltoxin, indicating its binding site on tubulin.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Microtúbulos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Sulfonamidas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Administración Oral , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Disponibilidad Biológica , Barrera Hematoencefálica/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Células HCT116 , Humanos , Células K562 , Células MCF-7 , Ratones , Ratones Desnudos , Microtúbulos/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Polimerizacion/efectos de los fármacos , Sulfonamidas/síntesis química , Sulfonamidas/farmacocinética , Tubulina (Proteína)/metabolismo , Carga Tumoral/efectos de los fármacos
18.
J Med Chem ; 55(11): 5174-87, 2012 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-22587519

RESUMEN

Tubulin, the major structural component of microtubules, is a target for the development of anticancer agents. A series of (Z)-1-aryl-3-arylamino-2-propen-1-one (10) were synthesized and evaluated for antiproliferative activity in cell-based assay. The most active compound (Z)-1-(2-bromo-3,4,5-trimethoxyphenyl)-3-(3-hydroxy-4-methoxyphenylamino)prop-2-en-1-one (10ae) was tested in 20 tumor cell lines including multidrug resistant phenotype and was found to induce apoptosis in all these cell lines with similar GI(50) values. Flow cytometry studies showed that 10ae arrested the cells in G2/M phase of cell cycle. In addition to G2/M block, these compounds caused microtubule stabilization like paclitaxel and induced apoptosis via activation of the caspase family. The observations made in this investigation demonstrate that (Z)-1-Aryl-3-arylamino-2-propen-1-one (10) represents a new class of microtubule-stabilizing agents.


Asunto(s)
Alquenos/síntesis química , Aminofenoles/síntesis química , Antineoplásicos/síntesis química , Moduladores de Tubulina/síntesis química , Tubulina (Proteína)/metabolismo , Alquenos/química , Alquenos/farmacología , Aminofenoles/química , Aminofenoles/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Fase G2/efectos de los fármacos , Humanos , Microtúbulos/efectos de los fármacos , Microtúbulos/ultraestructura , Polimerizacion , Huso Acromático/efectos de los fármacos , Huso Acromático/ultraestructura , Estereoisomerismo , Relación Estructura-Actividad , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacología
19.
J Med Chem ; 54(18): 6254-76, 2011 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-21812421

RESUMEN

Cyclin D proteins are elevated in many cancer cells, and targeted deletion of cyclin D1 gene in the mammary tissues protects mice from breast cancer. Accordingly, there is an increasing awareness of this novel nonenzymatic target for cancer therapeutics. We have developed novel, nonalkylating styrylbenzylsulfones that induce cell death in wide variety of cancer cells without affecting the proliferation and survival of normal cells. The development of derivatized styrylbenzylsulfones followed logically from a tumor cell cytotoxicity screen performed in our laboratory that did not have an a priori target profile. Modifications of some of the precursor molecules led to lead optimization with regard to tumor cell cytotoxicity. In this report we describe the synthesis and structure-activity relationships of novel, nonalkylating (E)-styrylbenzylsulfones and the development of the novel anticancer agent sodium (E)-2-{2-methoxy-5-[(2',4',6'-trimethoxystyrylsulfonyl)methyl]phenylamino}acetate (ON 01910.Na), which is in phase III trials for myelodysplastic syndromes (MDS) associated with aberrant expression of cyclin D proteins.


Asunto(s)
Antineoplásicos/síntesis química , Glicina/análogos & derivados , Inhibidores de Proteínas Quinasas/síntesis química , Sulfonas/síntesis química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis , Disponibilidad Biológica , Línea Celular Tumoral , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Glicina/síntesis química , Glicina/química , Glicina/farmacología , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Estereoisomerismo , Relación Estructura-Actividad , Sulfonas/química , Sulfonas/farmacología , Trasplante Heterólogo
20.
Genes Cancer ; 1(4): 331-45, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20717479

RESUMEN

Here we report the discovery of ON044580, an α-benzoyl styryl benzyl sulfide that possesses potent inhibitory activity against two unrelated kinases, JAK2 and BCR-ABL, and exhibits cytotoxicity to human tumor cells derived from chronic myelogenous leukemia (CML) and myelodysplasia (MDS) patients or cells harboring a mutant JAK2 kinase. This novel spectrum of activity is explained by the non-ATP-competitive inhibition of JAK2 and BCR-ABL kinases. ON044580 inhibits mutant JAK2 kinase and the proliferation of JAK2(V617F)-positive leukemic cells and blocks the IL-3-mediated phosphorylation of JAK2 and STAT5. Interestingly, this compound also directly inhibits the kinase activity of both wild-type and imatinib-resistant (T315I) forms of the BCR-ABL kinase. Finally, ON044580 effectively induces apoptosis of imatinib-resistant CML patient cells. The apparently unrelated JAK2 and BCR-ABL kinases share a common substrate, STAT5, and such substrate competitive inhibitors represent an alternative therapeutic strategy for development of new inhibitors. The novel mechanism of kinase inhibition exhibited by ON044580 renders it effective against mutant forms of kinases such as the BCR-ABL(T315I) and JAK2(V617F). Importantly, ON044580 selectively reduces the number of aneuploid cells in primary bone marrow samples from monosomy 7 MDS patients, suggesting another regulatory cascade amenable to this agent in these aberrant cells. Data presented suggest that this compound could have multiple therapeutic applications including monosomy 7 MDS, imatinib-resistant CML, and myeloproliferative neoplasms that develop resistance to ATP-competitive agents.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA