Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Bioorg Med Chem Lett ; 27(24): 5473-5480, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29150397

RESUMEN

In this work, we describe the use of the rule of 3 fragment-based strategies from biochemical screening data of 1100 in-house, small, low molecular weight fragments. The sequential combination of in silico fragment hopping and fragment linking based on S160/Y161/A162 hinge residues hydrogen bonding interactions leads to the identification of novel 1H-benzo[d]imidazol-2-yl)-1H-indazol class of Phosphoinositide-Dependent Kinase-1 (PDK1) inhibitors. Consequent SAR and follow-up screening data led to the discovery of two potent PDK1 inhibitors: compound 32 and 35, with an IC50 of 80 nM and 94 nM, respectively. Further biological evaluation showed that, at the low nanomolar concentration, the drug had potent ability to inhibit phosphorylation of AKT and p70S6, and selectively kill the cancer cells with mutations in both PTEN and PI3K. The microarray data showed that DUSP6, DUSP4, and FOSL1 were down-regulated in the sensitive cell lines with the compound treatment. The in vivo test showed that 35 can significantly inhibit tumor growth without influencing body weight growth. Our results suggest that these compounds, especially 35, merit further pre-clinical evaluation.


Asunto(s)
Diseño de Fármacos , Indazoles/química , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Sitios de Unión , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Fosfatasa 6 de Especificidad Dual/genética , Fosfatasa 6 de Especificidad Dual/metabolismo , Fosfatasas de Especificidad Dual/genética , Fosfatasas de Especificidad Dual/metabolismo , Humanos , Imidazoles/química , Indazoles/síntesis química , Indazoles/farmacología , Concentración 50 Inhibidora , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Simulación del Acoplamiento Molecular , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Relación Estructura-Actividad
2.
Leukemia ; 38(1): 82-95, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38007585

RESUMEN

We identified activin A receptor type I (ACVR1), a member of the TGF-ß superfamily, as a factor favoring acute myeloid leukemia (AML) growth and a new potential therapeutic target. ACVR1 is overexpressed in FLT3-mutated AML and inhibition of ACVR1 expression sensitized AML cells to FLT3 inhibitors. We developed a novel ACVR1 inhibitor, TP-0184, which selectively caused growth arrest in FLT3-mutated AML cell lines. Molecular docking and in vitro kinase assays revealed that TP-0184 binds to both ACVR1 and FLT3 with high affinity and inhibits FLT3/ACVR1 downstream signaling. Treatment with TP-0184 or in combination with BCL2 inhibitor, venetoclax dramatically inhibited leukemia growth in FLT3-mutated AML cell lines and patient-derived xenograft models in a dose-dependent manner. These findings suggest that ACVR1 is a novel biomarker and plays a role in AML resistance to FLT3 inhibitors and that FLT3/ACVR1 dual inhibitor TP-0184 is a novel potential therapeutic tool for AML with FLT3 mutations.


Asunto(s)
Leucemia Mieloide Aguda , Humanos , Simulación del Acoplamiento Molecular , Mutación , Línea Celular Tumoral , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/uso terapéutico , Apoptosis , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/uso terapéutico
3.
Cancer Immunol Res ; 11(9): 1222-1236, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37378662

RESUMEN

The receptor tyrosine kinase AXL is a member of the TYRO3, AXL, and proto-oncogene tyrosine-protein kinase MER family and plays pleiotropic roles in cancer progression. AXL is expressed in immunosuppressive cells, which contributes to decreased efficacy of immunotherapy. Therefore, we hypothesized that AXL inhibition could serve as a strategy to overcome resistance to chimeric antigen receptor T (CAR T)-cell therapy. To test this, we determined the impact of AXL inhibition on CD19-targeted CAR T (CART19)-cell functions. Our results demonstrate that T cells and CAR T cells express high levels of AXL. Specifically, higher levels of AXL on activated Th2 CAR T cells and M2-polarized macrophages were observed. AXL inhibition with small molecules or via genetic disruption in T cells demonstrated selective inhibition of Th2 CAR T cells, reduction of Th2 cytokines, reversal of CAR T-cell inhibition, and promotion of CAR T-cell effector functions. AXL inhibition is a novel strategy to enhance CAR T-cell functions through two independent, but complementary, mechanisms: targeting Th2 cells and reversing myeloid-induced CAR T-cell inhibition through selective targeting of M2-polarized macrophages.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Tirosina Quinasa del Receptor Axl , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras/genética
4.
Exp Cell Res ; 317(3): 307-18, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21075102

RESUMEN

HGF signaling induces epithelial cells to disassemble cadherin-based adhesion and increase cell motility and invasion, a process termed epithelial-mesenchymal transition (EMT). EMT plays a major role in cancer metastasis, allowing individual cells to detach from the primary tumor, invade local tissue, and colonize distant tissues with new tumors. While invasion of vascular and lymphatic networks is the predominant route of metastasis, nerves also can act as networks for dissemination of cancer cell to distant sites in a process termed perineual invasion (PNI). Signaling between nerves and invasive cancer cells remains poorly understood, as does cellular decision making that selects the specific route of invasion. Here we examine how HGF signaling contributes to PNI using reductionist culture model systems. We find that TGFß, produced by PC12 cells, enhances scattering in response to HGF stimulation, increasing both cell-cell junction disassembly and cell migration. Further, gradients of TGFß induce migratory mesenchymal cells to undergo chemotaxis towards the source of TGFß. Interestingly, VEGF suppresses TGFß-induced enhancement of scattering. These results have broad implications for how combinatorial growth factor signaling contributes to cancer metastasis, suggesting that VEGF and TGFß might modulate HGF signaling to influence route selection during cancer progression.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Factor de Crecimiento de Hepatocito/farmacología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Animales , Movimiento Celular , Metástasis de la Neoplasia , Células PC12 , Ratas , Transducción de Señal , Factor de Crecimiento Transformador beta/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología
5.
Mol Cancer Ther ; 21(1): 38-47, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34675118

RESUMEN

Pancreatic cancer is the third leading cause of cancer-related deaths in the United States with a 5-year survival less than 5%. Resistance to standard therapy and limited response to immune checkpoint blockade due to the immunosuppressive and stroma-rich microenvironment remain major challenges in the treatment of pancreatic cancer. A key cellular program involved in therapy resistance is epithelial plasticity, which is also associated with invasion, metastasis, and evasion of immune surveillance. The receptor tyrosine kinase AXL is a key driver of tumor cell epithelial plasticity. High expression and activity of AXL is associated with poor prognosis, metastasis, and therapy resistance in multiple types of cancer including pancreatic. Here, we show that an AXL inhibitor (TP-0903), has antitumor and therapy sensitizing effects in preclinical models of pancreatic ductal adenocarcinoma (PDA). We demonstrate that TP-0903 as a single agent or in combination with gemcitabine and/or anti-programmed cell death protein 1 (PD1) antibody has anti-metastatic and anti-tumor effects in PDA tumor bearing mice, leading to increased survival. In addition, gene expression analysis of tumors demonstrated upregulation of pro-inflammatory and immune activation genes in tumors from TP-0903-treated animals compared with the vehicle, indicating pharmacologic inhibition of AXL activation leads to an immunostimulatory microenvironment. This effect was augmented when TP-0903 was combined with gemcitabine and anti-PD1 antibody. These results provide clear rationale for evaluating TP-0903 in the treatment of pancreatic cancer.


Asunto(s)
Inmunoterapia/métodos , Neoplasias Pancreáticas/tratamiento farmacológico , Proteínas Proto-Oncogénicas/uso terapéutico , Pirimidinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/uso terapéutico , Sulfonamidas/uso terapéutico , Animales , Línea Celular Tumoral , Humanos , Ratones , Metástasis de la Neoplasia , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas/farmacología , Pirimidinas/farmacología , Proteínas Tirosina Quinasas Receptoras/farmacología , Sulfonamidas/farmacología , Análisis de Supervivencia , Microambiente Tumoral , Tirosina Quinasa del Receptor Axl
6.
Leukemia ; 36(3): 746-759, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34741118

RESUMEN

Myelofibrosis (MF) is the deadliest form of myeloproliferative neoplasm (MPN). The JAK inhibitor Ruxolitinib can reduce constitutional symptoms but it does not substantially improve bone marrow fibrosis. Pim1 expression is significantly elevated in MPN/MF hematopoietic progenitors. Here, we show that genetic ablation of Pim1 blocked the development of myelofibrosis induced by Jak2V617F and MPLW515L. Pharmacologic inhibition of Pim1 with a second-generation Pim kinase inhibitor TP-3654 significantly reduced leukocytosis and splenomegaly, and attenuated bone marrow fibrosis in Jak2V617F and MPLW515L mouse models of MF. Combined treatment of TP-3654 and Ruxolitinib resulted in greater reduction of spleen size, normalization of blood leukocyte counts and abrogation of bone marrow fibrosis in murine models of MF. TP-3654 treatment also preferentially inhibited Jak2V617F mutant hematopoietic progenitors in mice. Mechanistically, we show that TP-3654 treatment significantly inhibits mTORC1, MYC and TGF-ß signaling in Jak2V617F mutant hematopoietic cells and diminishes the expression of fibrotic markers in the bone marrow. Collectively, our results suggest that Pim1 plays an important role in the pathogenesis of MF, and inhibition of Pim1 with TP-3654 might be useful for treatment of MF.


Asunto(s)
Mielofibrosis Primaria/tratamiento farmacológico , Mielofibrosis Primaria/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-pim-1/genética , Animales , Línea Celular , Modelos Animales de Enfermedad , Eliminación de Gen , Humanos , Janus Quinasa 2/genética , Ratones , Ratones Noqueados , Inhibidores de Proteínas Quinasas/uso terapéutico
7.
Nat Commun ; 12(1): 5337, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34504101

RESUMEN

TNK1 is a non-receptor tyrosine kinase with poorly understood biological function and regulation. Here, we identify TNK1 dependencies in primary human cancers. We also discover a MARK-mediated phosphorylation on TNK1 at S502 that promotes an interaction between TNK1 and 14-3-3, which sequesters TNK1 and inhibits its kinase activity. Conversely, the release of TNK1 from 14-3-3 allows TNK1 to cluster in ubiquitin-rich puncta and become active. Active TNK1 induces growth factor-independent proliferation of lymphoid cells in cell culture and mouse models. One unusual feature of TNK1 is a ubiquitin-association domain (UBA) on its C-terminus. Here, we characterize the TNK1 UBA, which has high affinity for poly-ubiquitin. Point mutations that disrupt ubiquitin binding inhibit TNK1 activity. These data suggest a mechanism in which TNK1 toggles between 14-3-3-bound (inactive) and ubiquitin-bound (active) states. Finally, we identify a TNK1 inhibitor, TP-5801, which shows nanomolar potency against TNK1-transformed cells and suppresses tumor growth in vivo.


Asunto(s)
Proteínas 14-3-3/genética , Proteínas Fetales/genética , Linfocitos/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Proteínas Tirosina Quinasas/genética , Ubiquitina/genética , Proteínas 14-3-3/metabolismo , Células A549 , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Proteínas Fetales/antagonistas & inhibidores , Proteínas Fetales/metabolismo , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Linfocitos/efectos de los fármacos , Linfocitos/patología , Ratones , Fosfolipasa C gamma/genética , Fosfolipasa C gamma/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/mortalidad , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Pirimidinas/farmacología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Análisis de Supervivencia , Carga Tumoral/efectos de los fármacos , Ubiquitina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
SLAS Discov ; 24(1): 77-85, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30204534

RESUMEN

ETS transcription factors from the ERG and ETV1/4/5 subfamilies are overexpressed in the majority of prostate cancer patients and contribute to disease progression. Here, we have developed two in vitro assays for the interaction of ETS transcription factors with DNA that are amenable to high-throughput screening. Using ETS1 as a model, we applied these assays to screen 110 compounds derived from a high-throughput virtual screen. We found that the use of lower-affinity DNA binding sequences, similar to those that ERG and ETV1 bind to in prostate cells, allowed for higher inhibition from many of these test compounds. Further pilot experiments demonstrated that the in vitro assays are robust for ERG, ETV1, and ETV5, three of the ETS transcription factors that are overexpressed in prostate cancer.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Proteínas Proto-Oncogénicas c-ets/genética , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Humanos , Masculino , Próstata/metabolismo , Neoplasias de la Próstata/genética , Regulador Transcripcional ERG/genética
9.
Curr Oncol Rep ; 10(2): 122-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18377825

RESUMEN

Tubulin is a very important target for cancer-fighting therapies; therefore, the cancer research community continues to adopt new ways of developing the therapeutic potential of tubulin and tubulin-associated proteins. Two families of tubulin-associated kinases, Aurora and Polo-like, have received significant attention regarding how they contribute to tumorigenesis and can be targeted with selective small molecule inhibitors. Aurora and Polo-like kinases play essential roles in centrosome separation, chromosome alignment and segregation, and cytokinesis. Inhibition of any of these kinases results in abnormal mitotic events (which vary depending on the particular family member) and eventually leads to apoptosis. Because of the biological consequences of inhibiting these kinases, several Aurora or Polo-like selective inhibitors have advanced to various stages of preclinical and clinical development; the most advanced are currently in phase 2 clinical trials.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Neoplasias/enzimología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Tubulina (Proteína)/fisiología , Animales , Antineoplásicos/farmacología , Apoptosis , Aurora Quinasas , Cromosomas/ultraestructura , Ensayos Clínicos como Asunto , Citocinesis , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/farmacología , Humanos , Neoplasias/metabolismo , Quinasa Tipo Polo 1
10.
Oncotarget ; 9(98): 37173-37184, 2018 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-30647852

RESUMEN

Earlier we have shown the expression of a constitutively active receptor tyrosine kinase Axl in CLL B-cells from previously untreated CLL patients, and that Axl inhibitor TP-0903 induces robust leukemic B-cell death. To explore whether Axl is an effective target in relapsed/refractory CLL patients, we analyzed CLL B-cells obtained from CLL patients on ibrutinib therapy. Ibrutinib-exposed CLL B-cells were treated with increasing doses (0.01- 0.50µM) of a new formulation of high-affinity Axl inhibitor, TP-0903 (tartrate salt), for 24 hours and LD50 doses were determined. Sensitivity of CLL B-cells was compared with known prognostic factors and effect of TP-0903 was also evaluated on Axl signaling pathway in CLL B-cells from this cohort. We detected sustained overexpression of Axl in CLL B-cells from CLL patients on ibrutinib treatment, suggests targeting Axl could be a promising strategy to overcome drug resistance and killing of CLL B-cells in these patients. We found that CLL B-cells from sixty-nine percent of relapsed CLL patients actively on ibrutinib therapy were found to be highly sensitive to TP-0903 with induction of apoptosis at nanomolar doses (≤0.50 µM). TP-0903 treatment effectively inhibited Axl phosphorylation and reduced expression levels of anti-apoptotic proteins (Mcl-1, XIAP) in ibrutinib exposed CLL B-cells. In total, our in vitro preclinical studies showing that TP-0903 is very effective at inducing apoptosis in CLL B-cells obtained from ibrutinib-exposed patients supports further testing of this drug in relapsed/refractory CLL.

11.
Mol Cancer Ther ; 5(7): 1764-73, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16891462

RESUMEN

Aurora A and Aurora B are potential targets for anticancer drug development due to their roles in tumorigenesis and disease progression. To identify small-molecule inhibitors of the Aurora kinases, we undertook a structure-based design approach that used three-dimensional structural models of the Aurora A kinase and molecular docking simulations of chemical entities. Based on these computational methods, a new generation of inhibitors derived from quinazoline and pyrimidine-based tricyclic scaffolds were synthesized and evaluated for Aurora A kinase inhibitory activity, which led to the identification of 4-(6,7-dimethoxy-9H-1,3,9-triaza-fluoren-4-yl)-piperazine-1-carbothioic acid [4-(pyrimidin-2-ylsulfamoyl)-phenyl]-amide. The lead compound showed selectivity for the Aurora kinases when it was evaluated against a panel of diverse kinases. Additionally, the compound was evaluated in cell-based assays, showing a dose-dependent decrease in phospho-histone H3 levels and an arrest of the cell cycle in the G(2)-M fraction. Although biological effects were observed only at relatively high concentrations, this chemical series provides an excellent starting point for drug optimization and further development.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Plomo/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Sulfonamidas/farmacología , Tionas/farmacología , Antineoplásicos/química , Aurora Quinasa B , Aurora Quinasas , Ensayos de Selección de Medicamentos Antitumorales , Histonas/metabolismo , Humanos , Plomo/química , Conformación Proteica , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/química , Relación Estructura-Actividad , Sulfonamidas/química , Tionas/química
12.
Mol Cancer Ther ; 5(10): 2450-8, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17041088

RESUMEN

To address the increased need to understand the similarities and differences in targeting Aurora A or Aurora B for the treatment of cancer, we systematically evaluated the relative importance of Aurora A and/or Aurora B as molecular targets using antisense oligonucleotides. It was found that perturbations in Aurora A and Aurora B signaling result in growth arrest and apoptosis preferentially in cancer cells. The biological fingerprints of Aurora A and Aurora B inhibition were compared and contrasted in efforts to identify the superior therapeutic target. Due to the different biological responses, we conclude that each Aurora kinase should be treated as autonomous drug targets, which can be targeted independently or in combination. We observed no advantages to targeting both kinases simultaneously and feel that an Aurora A-targeted therapy may have some beneficial consequences over an Aurora B-targeted therapy, such as mitotic arrest and the rapid induction of apoptosis.


Asunto(s)
Neoplasias Pancreáticas/patología , Proteínas Serina-Treonina Quinasas/biosíntesis , Apoptosis , Aurora Quinasa B , Aurora Quinasas , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Activación Enzimática , Perfilación de la Expresión Génica , Humanos , Oligonucleótidos Antisentido/farmacología , Neoplasias Pancreáticas/enzimología , Poliploidía , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Ensayo de Tumor de Célula Madre
13.
Semin Oncol ; 33(4): 436-48, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16890798

RESUMEN

Unrestrained cell division in cancer cells is dependent upon mitosis and its related processes. Therefore, a proven effective strategy in cancer treatment has been to interfere with the function of the mitotic spindle. Despite the success of these anti-mitotic agents, tubulin itself remains the only spindle-associated protein targeted by clinically approved agents. However, in recent years major advances have been achieved in targeting proteins that associate with tubulin and the mitotic spindle. Mitotic kinases such as the Aurora and Polo families are receiving significant attention due to their vital roles in assuring proper centrosome separation and chromosome segregation. Indeed, potent and selective inhibitors of these kinases have entered clinical trials. Similarly, the kinesins, particularly kinesin spindle protein (KSP), have emerged as potential therapeutic targets and inhibitors of KSP are currently under evaluation in the clinic. Although inhibitors have not been reported, mitotic checkpoint kinases (Mad2) and separase are additional potential targets for therapeutic intervention. Continued investigation of mechanisms regulating mitotic events will likely reveal additional proteins and pathways that could be potentially targeted and thereby provide more effective therapeutic options for cancer patients.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas de Ciclo Celular/antagonistas & inhibidores , Mitosis/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Antineoplásicos/farmacología , Aurora Quinasas , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Endopeptidasas , Inestabilidad Genómica , Humanos , Cinesinas/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/efectos de los fármacos , Securina , Separasa , Tubulina (Proteína)/efectos de los fármacos , Quinasa Tipo Polo 1
15.
Mol Cancer Ther ; 2(6): 589-95, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12813139

RESUMEN

Aurora-2 kinase has been shown to contribute to oncogenic transformation and is frequently overexpressed and amplified in many human tumor types. Aurora-2 belongs to a small family of mitotic serine/threonine kinases that regulate centrosome maturation, chromosome segregation, and cytokinesis. The mechanism behind the transforming activity of aurora-2 is not fully understood; however, the role of aurora-2 in regulating the centrosome cycle is likely responsible for its ability to transform cells. Aurora-2 overexpression has been correlated with centrosome amplification, which can be a driving cause of genomic instability in tumor cells. In addition, recent work has demonstrated that aurora-2 plays an active function in promoting entry into mitosis by regulating local translation of centrosomal stored mRNA, such as cyclin B1. These recent findings implicate aurora-2 as an important regulator of both genomic integrity and cell cycle progression in cancer cells and suggest that aurora-2 is an attractive target for anticancer drug development.


Asunto(s)
Neoplasias/enzimología , Proteínas Serina-Treonina Quinasas/fisiología , Antineoplásicos/farmacología , Aurora Quinasas , Centrosoma/ultraestructura , Ciclina B/metabolismo , Ciclina B1 , Regulación Neoplásica de la Expresión Génica , Mitosis , Modelos Biológicos , Biosíntesis de Proteínas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/metabolismo
16.
Clin Cancer Res ; 21(9): 2115-26, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25673699

RESUMEN

PURPOSE: B-cell chronic lymphocytic leukemia (CLL) is an incurable disease despite aggressive therapeutic approaches. We previously found that Axl receptor tyrosine kinase (RTK) plays a critical role in CLL B-cell survival. Here, we explored the possibility of using a high-affinity Axl inhibitor as a single agent or in combination with Bruton's tyrosine kinase (BTK) inhibitors for future clinical trial to treat patients with CLL. EXPERIMENTAL DESIGN: Expression/activation status of other members of the TAM (e.g., Tyro3, Axl, and MER) family of RTKs in CLL B cells was evaluated. Cells were treated with a high-affinity orally bioavailable Axl inhibitor TP-0903 with or without the presence of CLL bone marrow stromal cells (BMSCs). Inhibitory effects of TP-0903 on the Axl signaling pathway were also evaluated in CLL B cells. Finally, cells were exposed to TP-0903 in combination with BTK inhibitors to determine any synergistic/additive effects of the combination. RESULTS: CLL B cells overexpress Tyro3, but not MER. Of interest, Tyro3 remains as constitutively phosphorylated and forms a complex with Axl in CLL B cells. TP-0903 induces massive apoptosis in CLL B cells with LD50 values of nanomolar ranges. Importantly, CLL BMSCs could not protect the leukemic B cells from TP-0903-induced apoptosis. A marked reduction of the antiapoptotic proteins Mcl-1, Bcl-2, and XIAP and upregulation of the proapoptotic protein BIM in CLL B cells was detected as a result of Axl inhibition. Finally, combination of TP-0903 with BTK inhibitors augments CLL B-cell apoptosis. CONCLUSIONS: Administration of TP-0903 either as a single agent or in combination with BTK inhibitors may be effective in treating patients with CLL.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Leucemia Linfocítica Crónica de Células B/patología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Pirimidinas/farmacología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Sulfonamidas/farmacología , Agammaglobulinemia Tirosina Quinasa , Linfocitos B , Western Blotting , Células Cultivadas , Técnicas de Cocultivo , Sinergismo Farmacológico , Citometría de Flujo , Humanos , Inmunoprecipitación , Inhibidores de Proteínas Quinasas/farmacología , Transfección , Tirosina Quinasa del Receptor Axl
17.
Future Med Chem ; 6(10): 1167-78, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25078136

RESUMEN

Pyruvate kinase converts phosphoenolpyruvate to pyruvate, catalyzing the rate-limiting step of glycolysis. The M1 isoenzyme of pyruvate kinase (PKM1) is found in adult tissues; whereas, PKM2 is a splicesome variant found in embryonic and cancer cells. PKM2 expression in malignant cells is a result of the tumor microenvironment and is responsible for maintaining a glycolytic phenotype. PKM2 has other nonmetabolic functions in malignant cells, including transcriptional coactivation and protein kinase activity. PKM2 activators have antitumor properties by inducing tetramerization of two PKM2 dimers causing PKM2 to function like PKM1. Restoring PKM2 to PKM1-like levels of activity causes reversal of the Warburg effect in cancer cells. PKM2 activators have therapeutic potential in the treatment of cancer and other metabolic diseases.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Proteínas Portadoras/agonistas , Proteínas de la Membrana/agonistas , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Hormonas Tiroideas/agonistas , Animales , Antineoplásicos/uso terapéutico , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Descubrimiento de Drogas , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Terapia Molecular Dirigida , Neoplasias/enzimología , Neoplasias/genética , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Hormonas Tiroideas/genética , Hormonas Tiroideas/metabolismo , Proteínas de Unión a Hormona Tiroide
18.
Biomed Res Int ; 2014: 273180, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25313354

RESUMEN

BACKGROUND: Nek2 is a serine/threonine kinase localized to the centrosome. It promotes cell cycle progression from G2 to M by inducing centrosome separation. Recent studies have shown that high Nek2 expression is correlated with drug resistance in multiple myeloma patients. MATERIALS AND METHODS: To investigate the role of Nek2 in bortezomib resistance, we ectopically overexpressed Nek2 in several cancer cell lines, including multiple myeloma lines. Small-molecule inhibitors of Nek2 were discovered using an in-house library of compounds. We tested the inhibitors on proteasome and cell cycle activity in several cell lines. RESULTS: Proteasome activity was elevated in Nek2-overexpressing cell lines. The Nek2 inhibitors inhibited proteasome activity in these cancer cell lines. Treatment with these inhibitors resulted in inhibition of proteasome-mediated degradation of several cell cycle regulators in HeLa cells, leaving them arrested in G2/M. Combining these Nek2 inhibitors with bortezomib increased the efficacy of bortezomib in decreasing proteasome activity in vitro. Treatment with these novel Nek2 inhibitors successfully mitigated drug resistance in bortezomib-resistant multiple myeloma. CONCLUSION: Nek2 plays a central role in proteasome-mediated cell cycle regulation and in conferring resistance to bortezomib in cancer cells. Taken together, our results introduce Nek2 as a therapeutic target in bortezomib-resistant multiple myeloma.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Ácidos Borónicos/farmacología , Bortezomib , Proteína Quinasa CDC2/metabolismo , Línea Celular Tumoral , Ciclina B/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Mitosis/efectos de los fármacos , Quinasas Relacionadas con NIMA , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteolisis/efectos de los fármacos , Pirazinas/farmacología , Bibliotecas de Moléculas Pequeñas/química
19.
Neoplasia ; 16(5): 403-12, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24953177

RESUMEN

The proto-oncogene proviral integration site for moloney murine leukemia virus (PIM) kinases (PIM-1, PIM-2, and PIM-3) are serine/threonine kinases that are involved in a number of signaling pathways important to cancer cells. PIM kinases act in downstream effector functions as inhibitors of apoptosis and as positive regulators of G1-S phase progression through the cell cycle. PIM kinases are upregulated in multiple cancer indications, including lymphoma, leukemia, multiple myeloma, and prostate, gastric, and head and neck cancers. Overexpression of one or more PIM family members in patient tumors frequently correlates with poor prognosis. The aim of this investigation was to evaluate PIM expression in low- and high-grade urothelial carcinoma and to assess the role PIM function in disease progression and their potential to serve as molecular targets for therapy. One hundred thirty-seven cases of urothelial carcinoma were included in this study of surgical biopsy and resection specimens. High levels of expression of all three PIM family members were observed in both noninvasive and invasive urothelial carcinomas. The second-generation PIM inhibitor, TP-3654, displays submicromolar activity in pharmacodynamic biomarker modulation, cell proliferation studies, and colony formation assays using the UM-UC-3 bladder cancer cell line. TP-3654 displays favorable human ether-à-go-go-related gene and cytochrome P450 inhibition profiles compared with the first-generation PIM inhibitor, SGI-1776, and exhibits oral bioavailability. In vivo xenograft studies using a bladder cancer cell line show that PIM kinase inhibition can reduce tumor growth, suggesting that PIM kinase inhibitors may be active in human urothelial carcinomas.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Células Transicionales/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Neoplasias de la Vejiga Urinaria/enzimología , Animales , Western Blotting , Femenino , Humanos , Imidazoles/farmacología , Masculino , Ratones , Ratones Desnudos , Reacción en Cadena de la Polimerasa Multiplex , Oligopéptidos/farmacología , Proto-Oncogenes Mas , Piridazinas/farmacología , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción Genética , Péptido Intestinal Vasoactivo/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
J Med Chem ; 56(23): 9496-508, 2013 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-24237195

RESUMEN

Lysine specific demethylase 1 (LSD1) plays an important role in regulating histone lysine methylation at residues K4 and K9 on histone H3 and is an attractive therapeutic target in multiple malignancies. Here we report a structure-based virtual screen of a compound library containing ∼2 million small molecular entities. Computational docking and scoring followed by biochemical screening led to the identification of a novel N'-(1-phenylethylidene)-benzohydrazide series of LSD1 inhibitors with hits showing biochemical IC50s in the 200-400 nM range. Hit-to-lead optimization and structure-activity relationship studies aided in the discovery of compound 12, with a Ki of 31 nM. Compound 12 is reversible and specific for LSD1 as compared to the monoamine oxidases shows minimal inhibition of CYPs and hERG and inhibits proliferation and survival in several cancer cell lines, including breast and colorectal cancer. Compound 12 may be used to probe LSD1's biological role in these cancers.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Histona Demetilasas/antagonistas & inhibidores , Hidrazinas/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Cinética , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA