Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Sci Rep ; 12(1): 16104, 2022 09 27.
Article in English | MEDLINE | ID: mdl-36167880

ABSTRACT

Radio-resistance resulting from radiotherapy-induced fibrosis is a major clinical obstacle in breast cancer treatment because it typically leads to cancer recurrence, treatment failure, and patient death. Transforming growth factor-ß (TGF-ß) is a key signal messenger in fibrosis, which plays an important role in radiation-induced fibrosis and cancer stem cell (CSC) development, may be mediated through the generation of oxidative stress. This study was conducted to confirm the efficacy of vactosertib, a TGF-ß/ALK5 inhibitor, as a potent inhibitor in radiation-induced oxidative stress generation, fibrosis and CSC development. We used a 4T1-Luc allograft BALB/c syngeneic mouse model and 4T1-Luc and MDA-MB-231 cells for histological analysis, qRT-PCR, western blotting, ROS analysis, mammosphere formation analysis, monolayer fluorescence imaging analysis. Radiotherapy induces TGF-ß signaling, oxidative stress markers (4-HNE, NOX2, NOX4, PRDX1, NRF2, HO-1, NQO-1), fibrosis markers (PAI-1, α-SMA, FIBRONECTIN, COL1A1), and CSC properties. However, combination therapy with vactosertib not only inhibits these radiation-induced markers and properties by blocking TGF-ß signaling, but also enhances the anticancer effect of radiation by reducing the volume of breast cancer. Therefore, these data suggest that vactosertib can effectively reduce radiation fibrosis and resistance in breast cancer treatment by inhibiting radiation-induced TGF-ß signaling and oxidative stress, fibrosis, and CSC.


Subject(s)
Fibrocystic Breast Disease , Transforming Growth Factor beta , Aniline Compounds , Animals , Female , Fibronectins , Fibrosis , Humans , Mice , NF-E2-Related Factor 2/metabolism , Neoplasm Recurrence, Local , Oxidative Stress , Plasminogen Activator Inhibitor 1 , Reactive Oxygen Species , Transforming Growth Factor beta/metabolism , Transforming Growth Factors , Triazoles
2.
Ther Innov Regul Sci ; 53(2): 233-242, 2019 03.
Article in English | MEDLINE | ID: mdl-29888622

ABSTRACT

Globalization of the pharmaceutical industry has continued over the past few decades, and various regulatory authorities have put considerable effort into harmonizing and standardizing drug regulations. However, the regulatory practices of each regulatory authority, in addition to local differences in ethnic, social, and cultural backgrounds, create discrepancies in risk/benefit assessments, regulatory decisions, and drug label information in various countries. This study examines discrepancies in the label information for direct oral anticoagulants approved in the US, Europe, Korea, and Japan and reviews the causes of those discrepancies, focusing on regulatory practices. Although the label information for each direct oral anticoagulant in all 4 regions was supported by the same global, pivotal clinical data, it differed depending on regulatory authorities' judgments about the risk/benefit balance, which were based on their own requirements, regulations, perspectives on making regulatory decisions, and regulatory approval experiences, in addition to their review of the scientific data. In particular, the Korean Ministry of Food and Drug Safety and Japanese Pharmaceuticals and Medical Devices Agency have taken a comparatively conservative stance, with more emphasis on safety than on efficacy compared with regulatory authorities in western countries, because of the double threshold in their regulatory practice. Our findings suggest that drug label information in various regions will not be equal as long as differences in regulatory practice and non-regulatory factors exist among regulatory authorities. Also, those differences should be considered in order to streamline global drug discovery, development, and approval.


Subject(s)
Anticoagulants , Drug Labeling , Administration, Oral , Drug and Narcotic Control , Europe , Japan , Republic of Korea , United States
3.
Cell Physiol Biochem ; 38(2): 571-88, 2016.
Article in English | MEDLINE | ID: mdl-26845171

ABSTRACT

BACKGROUND/AIMS: Hypoxia is an environmental factor that aggravates liver fibrosis. HIF1α activates hepatic stellate cells (HSCs) and increases transforming growth factor-ß (TGF-ß) signaling and the epithelial mesenchymal transition (EMT), accelerating the progression of fibrosis. We evaluated the anti-fibrotic therapeutic potential of a small-molecule inhibitor of TGF-ß type I receptor kinase, EW-7197, on HIF1α-derived TGF-ß signaling in cholestatic liver fibrosis. METHODS: We used a bile duct ligation (BDL)-operated rat model to characterize the role of HIF1α-derived TGF-ß signaling in liver fibrosis. Cellular assays were performed in LX-2 cells (human immortalized HSCs). The anti-fibrotic effects of EW-7197 in liver tissues and HSCs were investigated via biochemical assays, immunohistochemistry (IHC), immunofluorescence (IF), chromatin immunoprecipitation (ChIP) assays, real-time PCR, and western blotting. RESULTS: In our BDL rat model, orally administered EW-7197 inhibited fibrosis and attenuated HIF1α-induced activation of HSCs and EMT in vivo. In addition, EW-7197 inhibited HIF1α-derived HSC activation and expression of EMT markers in LX-2 cells in vitro. CONCLUSION: This study suggests that EW-7197 exhibits potential as a treatment for liver fibrosis because it inhibits HIF1α-induced TGF-ß signaling.


Subject(s)
Aniline Compounds/therapeutic use , Epithelial-Mesenchymal Transition/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Cirrhosis/drug therapy , Liver/drug effects , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Triazoles/therapeutic use , Animals , Cell Line , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Oxidative Stress/drug effects , Protein Serine-Threonine Kinases/metabolism , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism
4.
Cancer Sci ; 107(2): 140-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26583567

ABSTRACT

Recent strategies for treating CML patients have focused on investigating new combinations of tyrosine kinase inhibitors (TKIs) as well as identifying novel translational research agents that can eradicate CML leukemia-initiating cells (CML-LICs). However, little is known about the therapeutic benefits such CML-LIC targeting therapies might bring to CML patients. In this study, we investigated the therapeutic potential of EW-7197, an orally bioavailable transforming growth factor-ß signaling inhibitor which has recently been approved as an Investigational New Drug (NIH, USA), to suppress CML-LICs in vivo. Compared to TKI treatment alone, administration of TKI plus EW-7197 to CML-affected mice significantly delayed disease relapse and prolonged survival. Notably, combined treatment with EW-7197 plus TKI was effective in eliminating CML-LICs even if they expressed the TKI-resistant T315I mutant BCR-ABL1 oncogene. Collectively, these results indicate that EW-7197 may be a promising candidate for a new therapeutic that can greatly benefit CML patients by working in combination with TKIs to eradicate CML-LICs.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Triazoles/pharmacology , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Humans , Imidazoles/administration & dosage , Mice , Mice, Inbred C57BL , Protein Kinase Inhibitors/administration & dosage , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridazines/administration & dosage , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Transfection , Transforming Growth Factor beta/antagonists & inhibitors
5.
Bioorg Med Chem Lett ; 25(22): 5228-31, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26483198

ABSTRACT

To further optimize a clinical candidate 5 (EW-7197), a series of 5-(3-, 4-, or 5-fluoro-substituted-6-methylpyridin-2-yl)-4-([1,2,4]triazolo[1,5-a]pyridin-6-yl)imidazoles 19a-l have been synthesized and evaluated for their TGF-ß type I receptor kinase (ALK5) and p38α MAP kinase inhibitory activity in an enzyme assay. The 5-(5-fluoro-substituted-6-methylpyridin-2-yl)-4-([1,2,4]triazolo[1,5-a]pyridin-6-yl)imidazoles 19h-l displayed the similar level of potency to that of 5 against both ALK5 (IC50=7.68-13.70 nM) and p38α MAP kinase (IC50=1240-3370 nM). Among them, 19j inhibited ALK5 with IC50 value of 7.68 nM in a kinase assay and displayed 82% inhibition at 100 nM in a luciferase reporter assay.


Subject(s)
Imidazoles/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/chemical synthesis , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Triazoles/chemical synthesis , Cell Line , Enzyme Assays , Genes, Reporter , Humans , Imidazoles/pharmacology , Luciferases/genetics , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Receptor, Transforming Growth Factor-beta Type I , Triazoles/pharmacology
6.
Oncotarget ; 6(35): 37526-43, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26462028

ABSTRACT

Distant relapse after chemotherapy is an important clinical issue for treating breast cancer patients and results from the development of cancer stem-like cells (CSCs) during chemotherapy. Here we report that blocking epithelial-to-mesenchymal transition (EMT) suppresses paclitaxel-induced CSCs properties by using a MDA-MB-231-xenografted mice model (in vivo), and breast cancer cell lines (in vitro). Paclitaxel, one of the cytotoxic taxane-drugs such as docetaxel, increases mesenchymal markers (Vimentin and Fibronectin) and decreases an epithelial marker (Zo-1). Blocking TGF-ß signaling with the TGF-ß type I receptor kinase (ALK5) inhibitor, EW-7197, suppresses paclitaxel-induced EMT and CSC properties such as mammosphere-forming efficiency (MSFE), aldehyde dehydrogenase (ALDH) activity, CD44+/CD24- ratio, and pluripotency regulators (Oct4, Nanog, Klf4, Myc, and Sox2). The combinatorial treatment of EW-7197 improves the therapeutic effect of paclitaxel by decreasing the lung metastasis and increasing the survival time in vivo. We confirmed that Snail is increased by paclitaxel-induced intracellular reactive oxygen species (ROS) and EW-7197 suppresses the paclitaxel-induced Snail and EMT by attenuating paclitaxel-induced intracellular ROS. Knock-down of SNAI1 suppresses paclitaxel-induced EMT and CSC properties. These data together suggest that blocking the Snail-induced EMT with the ALK5 inhibitor attenuates metastasis after paclitaxel-therapy and that this combinatorial approach could prove useful in treating breast cancer.


Subject(s)
Breast Neoplasms/prevention & control , Epithelial-Mesenchymal Transition/drug effects , Lung Neoplasms/prevention & control , Neoplastic Stem Cells/drug effects , Paclitaxel/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement , Cell Proliferation , Combined Modality Therapy , Drug Resistance, Neoplasm , Female , Humans , Kruppel-Like Factor 4 , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/genetics , Tumor Cells, Cultured , Wound Healing , Xenograft Model Antitumor Assays
7.
Cell Mol Life Sci ; 72(10): 2023-39, 2015 May.
Article in English | MEDLINE | ID: mdl-25487606

ABSTRACT

Fibrosis is an inherent response to chronic damage upon immense apoptosis or necrosis. Transforming growth factor-beta1 (TGF-ß1) signaling plays a key role in the fibrotic response to chronic liver injury. To develop anti-fibrotic therapeutics, we synthesized a novel small-molecule inhibitor of the TGF-ß type I receptor kinase (ALK5), EW-7197, and evaluated its therapeutic potential in carbon tetrachloride (CCl4) mouse, bile duct ligation (BDL) rat, bleomycin (BLM) mouse, and unilateral ureteral obstruction (UUO) mouse models. Western blot, immunofluorescence, siRNA, and ChIP analysis were carried out to characterize EW-7197 as a TGF-ß/Smad signaling inhibitor in LX-2, Hepa1c1c7, NRK52E, and MRC5 cells. In vivo anti-fibrotic activities of EW-7197 were examined by microarray, immunohistochemistry, western blotting, and a survival study in the animal models. EW-7197 decreased the expression of collagen, α-smooth muscle actin (α-SMA), fibronectin, 4-hydroxy-2, 3-nonenal, and integrins in the livers of CCl4 mice and BDL rats, in the lungs of BLM mice, and in the kidneys of UUO mice. Furthermore, EW-7197 extended the lifespan of CCl4 mice, BDL rats, and BLM mice. EW-7197 blocked the TGF-ß1-stimulated production of reactive oxygen species (ROS), collagen, and α-SMA in LX-2 cells and hepatic stellate cells (HSCs) isolated from mice. Moreover, EW-7197 attenuated TGF-ß- and ROS-induced HSCs activation to myofibroblasts as well as extracellular matrix accumulation. The mechanism of EW-7197 appeared to be blockade of both TGF-ß1/Smad2/3 and ROS signaling to exert an anti-fibrotic activity. This study shows that EW-7197 has a strong potential as an anti-fibrosis therapeutic agent via inhibition of TGF-ß-/Smad2/3 and ROS signaling.


Subject(s)
Aniline Compounds/pharmacology , Fibrosis/prevention & control , Kidney/metabolism , Liver/metabolism , Lung/metabolism , Signal Transduction/drug effects , Triazoles/pharmacology , Animals , Bleomycin , Blotting, Western , Carbon Tetrachloride , Cell Line , Chromatin Immunoprecipitation , DNA Primers/genetics , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Kidney/drug effects , Liver/drug effects , Lung/drug effects , Mice , Microarray Analysis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , Rats , Reactive Oxygen Species/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Smad Proteins/metabolism
8.
Cancer Lett ; 351(1): 72-80, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-24887560

ABSTRACT

TGF-ß signaling plays an important role in breast cancer progression and metastasis. Epithelial-mesenchymal transition (EMT) is an important step in the progression of solid tumors to metastatic disease. We previously reported that IN-1130, a novel transforming growth factor-ß type I receptor kinase (ALK5) inhibitor, suppressed renal fibrosis in obstructive nephropathy (Moon et al., 2006). Here, we show that IN-1130 suppressed EMT and the lung metastasis of mammary tumors in mouse models. Treating human and mouse cell lines with IN-1130 inhibited TGF-ß-mediated transcriptional activation, the phosphorylation and nuclear translocation of Smad2, and TGF-ß-induced-EMT, which induces morphological changes in epithelial cells. Additionally, we demonstrated that IN-1130 blocked TGF-ß-induced 4T1 mammary cancer cell migration and invasion. The TGF-ß-mediated increase in matrix metalloproteinase (MMP)-2 and MMP-9 expression was restored by IN-1130 co-treatment with TGF-ß in human epithelial cells and in 4T1 cells. Furthermore, we found that lung metastasis from primary breast cancer was inhibited by IN-1130 in both 4T1-xenografted BALB/c mice and MMTV/c-Neu transgenic mice without any change in primary tumor volume. IN-1130 prolonged the life span of tumor-bearing mice. In summary, this study indicated that IN-1130 has therapeutic potential for preventing breast cancer metastasis to the lung.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Imidazoles/pharmacology , Lung Neoplasms/prevention & control , Mammary Neoplasms, Experimental/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Quinoxalines/pharmacology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Movement , Cell Nucleus/metabolism , Drug Screening Assays, Antitumor , Female , Humans , Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Transplantation , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/physiology , Tumor Burden/drug effects
9.
Mol Cancer Ther ; 13(7): 1704-16, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24817629

ABSTRACT

Advanced tumors produce an excessive amount of transforming growth factor ß (TGFß), which promotes tumor progression at late stages of malignancy. The purpose of this study was to develop anti-TGFß therapeutics for cancer. We synthesized a novel small-molecule TGFß receptor I kinase (activin receptor-like kinase 5) inhibitor termed N-[[4-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-5-(6-methylpyridin-2-yl)-1H-imidazol-2-yl]methyl]-2-fluoroaniline (EW-7197), and we investigated its potential antimetastatic efficacy in mouse mammary tumor virus (MMTV)/c-Neu mice and 4T1 orthotopic-grafted mice. EW-7197 inhibited Smad/TGFß signaling, cell migration, invasion, and lung metastasis in MMTV/c-Neu mice and 4T1 orthotopic-grafted mice. EW-7197 also inhibited the epithelial-to-mesenchymal transition (EMT) in both TGFß-treated breast cancer cells and 4T1 orthotopic-grafted mice. Furthermore, EW-7197 enhanced cytotoxic T lymphocyte activity in 4T1 orthotopic-grafted mice and increased the survival time of 4T1-Luc and 4T1 breast tumor-bearing mice. In summary, EW-7197 showed potent in vivo antimetastatic activity, indicating its potential for use as an anticancer therapy.


Subject(s)
Aniline Compounds/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Triazoles/pharmacology , Animals , Breast Neoplasms/enzymology , Cell Line, Tumor , Female , Humans , Lung Neoplasms/enzymology , Mice , Mice, Inbred BALB C , Random Allocation , Receptor, Transforming Growth Factor-beta Type I , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
10.
J Med Chem ; 57(10): 4213-38, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24786585

ABSTRACT

A series of 2-substituted-4-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-5-(6-methylpyridin-2-yl)imidazoles was synthesized and evaluated to optimize a prototype inhibitor of TGF-ß type I receptor kinase (ALK5), 6. Combination of replacement of a quinoxalin-6-yl moiety of 6 with a [1,2,4]triazolo[1,5-a]pyridin-6-yl moiety, insertion of a methyleneamino linker, and a o-F substituent in the phenyl ring markedly increased ALK5 inhibitory activity, kinase selectivity, and oral bioavailability. The 12b (EW-7197) inhibited ALK5 with IC50 value of 0.013 µM in a kinase assay and with IC50 values of 0.0165 and 0.0121 µM in HaCaT (3TP-luc) stable cells and 4T1 (3TP-luc) stable cells, respectively, in a luciferase assay. Selectivity profiling of 12b using a panel of 320 protein kinases revealed that it is a highly selective ALK5/ALK4 inhibitor. Pharmacokinetic study with 12b·HCl in rats showed an oral bioavailability of 51% with high systemic exposure (AUC) of 1426 ng × h/mL and maximum plasma concentration (Cmax) of 1620 ng/mL. Rational optimization of 6 has led to the identification of a highly potent, selective, and orally bioavailable ALK5 inhibitor 12b.


Subject(s)
Aniline Compounds/chemical synthesis , Antifibrinolytic Agents/chemical synthesis , Antineoplastic Agents/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Triazoles/chemical synthesis , Administration, Oral , Aniline Compounds/pharmacokinetics , Aniline Compounds/pharmacology , Aniline Compounds/toxicity , Animals , Antifibrinolytic Agents/pharmacokinetics , Antifibrinolytic Agents/pharmacology , Antifibrinolytic Agents/toxicity , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Biological Availability , Drug Discovery , HEK293 Cells , Humans , Immunotherapy , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/toxicity , Rats , Receptor, Transforming Growth Factor-beta Type I , Structure-Activity Relationship , Triazoles/pharmacokinetics , Triazoles/pharmacology , Triazoles/toxicity
11.
Bioorg Med Chem ; 22(9): 2724-32, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24704197

ABSTRACT

A series of 4-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-5(3)-(6-methylpyridin-2-yl)imidazoles and -pyrazoles 14a-c, 15a-c, 16a, 16b, 19a-d, 21a, and 21b has been synthesized and evaluated for their ALK5 inhibitory activity in an enzyme assay and in a cell-based luciferase reporter assay. Among them, the pyrazole derivative 21b inhibited ALK5 phosphorylation with an IC50 value of 0.018 µM and showed 95% inhibition at 0.03 µM in a luciferase reporter assay using HaCaT cells permanently transfected with p3TP-luc reporter construct. The 21b showed a high selectivity index of 284 against p38α MAP kinase. The binding pose of 21b generated by docking analysis reveals that it fits well into the ATP binding cavity of ALK5 by forming several hydrogen bond interactions.


Subject(s)
Imidazoles/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazoles/chemistry , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Binding Sites , Catalytic Domain , Cell Line , Humans , Imidazoles/chemical synthesis , Imidazoles/metabolism , Molecular Docking Simulation , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Serine-Threonine Kinases/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Structure-Activity Relationship
12.
Biomol Ther (Seoul) ; 21(5): 323-31, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-24244818

ABSTRACT

TGF-ß pathway is being extensively evaluated as a potential therapeutic target. The transforming growth factor-ß (TGF-ß) signaling pathway has the dual role in both tumor suppression and tumor promotion. To design cancer therapeutics successfully, it is important to understand TGF-ß related functional contexts. This review discusses the molecular mechanism of the TGF-ß pathway and describes the different ways of tumor suppression and promotion by TGF-ß. In the last part of the review, the data on targeting TGF-ß pathway for cancer treatment is assessed. The TGF-ß inhibitors in pre-clinical studies, and Phase I and II clinical trials are updated.

13.
Cancer Lett ; 333(1): 18-31, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23174107

ABSTRACT

High aldehyde dehydrogenase (ALDH) activity has been recognized as a marker of cancer stem cells (CSCs) in breast cancer. In this study, we examined whether inhibition of ALDH activity suppresses stem-like cell properties in a 4T1 syngeneic mouse model of breast cancer. We found that ALDH-positive 4T1 cells showed stem cell-like properties in vitro and in vivo. Blockade of ALDH activity reduced the growth of CSCs in breast cancer cell lines. Treatment of mice with the ALDH inhibitor diethylaminobenzaldehyde (DEAB) significantly suppressed 4T1 cell metastasis to the lung. Recent evidence suggests that ALDH affects the response of stem cells to hypoxia; therefore, we examined a possible link between ALDH and hypoxia signaling in breast cancer. Hypoxia-inducible factor-2α (HIF-2α) was highly dysregulated in ALDH-positive 4T1 cells. We observed that ALDH was highly correlated with the HIF-2α expression in breast cancer cell lines and tissues. DEAB treatment of breast cancer cells reduced the expression of HIF-2α in vitro. In addition, reduction of HIF-2α expression suppressed in vitro self-renewal ability and in vivo tumor initiation in ALDH-positive 4T1 cells. Therefore, our findings may provide the evidence necessary for exploring a new strategy in the treatment of breast cancer.


Subject(s)
Aldehyde Dehydrogenase/physiology , Basic Helix-Loop-Helix Transcription Factors/physiology , Breast Neoplasms/pathology , Neoplastic Stem Cells/physiology , Aldehyde Dehydrogenase/antagonists & inhibitors , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/analysis , Cell Line, Tumor , Enzyme Activation , Female , Humans , Mice , Mice, Inbred BALB C , Octamer Transcription Factor-3/physiology
14.
Eur J Med Chem ; 57: 74-84, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23047226

ABSTRACT

A series of 2-benzylamino-4(5)-(6-methylpyridin-2-yl)-5(4)-([1,2,4]triazolo[1,5-a]pyridin-6-yl)thiazoles 12a-ab, 13a, 13b, and 18a-d has been synthesized and evaluated for their ALK5 inhibitory activity in an enzyme assay and in a cell-based luciferase reporter assay. The N-(3-fluorobenzyl)-4-(6-methylpyridin-2-yl)-5-([1,2,4]triazolo[1,5-a]pyridin-6-yl)thiazol-2-amine (12b) inhibited ALK5 phosphorylation with an IC(50) value of 7.01 nM and showed 61% inhibition at 30 nM in a luciferase reporter assay using HaCaT cells permanently transfected with p3TP-luc reporter construct.


Subject(s)
Keratinocytes/drug effects , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/chemistry , Receptors, Transforming Growth Factor beta/chemistry , Thiazoles/chemical synthesis , Cell Line, Transformed , Enzyme Assays , Genes, Reporter , Humans , Keratinocytes/enzymology , Luciferases , Molecular Docking Simulation , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Structure-Activity Relationship , Thiazoles/pharmacology , Transfection
15.
Environ Health Toxicol ; 27: e2012010, 2012.
Article in English | MEDLINE | ID: mdl-22639737

ABSTRACT

OBJECTIVES: In recent years, a number of structurally diverse Histone deacetylase (HDAC) inhibitors have been identified and these HDAC inhibitors induce growth arrest, differentiation and/or apoptosis of cancer cells in vitro and in vivo. This study aimed at investigating the anti-tumor activity of newly synthesized HDAC inhibitor, 3-(4-dimethylamino phenyl)-N-hydroxy-2-propenamide (IN-2001) using human breast cancer cells. METHODS: We have synthesized a new HDAC inhibitor, IN-2001, and cell proliferation inhibition assay with this chemical in estrogen receptor-positive human breast cancer MCF-7 cells. Cell cycle analysis on MCF-7 cells treated with IN-2001 was carried out by flow cytometry and gene expression was measured by RT-PCR. RESULTS: In MCF-7 cells IN-2001 showed remarkable anti-proliferative effects in a dose- and time-dependent manner. In MCF-7 cells, IN-2001 showed a more potent growth inhibitory effect than that of suberoylanilide hydroxamic acid. These growth inhibitory effects were related to the cell cycle arrest and induction of apoptosis. IN-2001 showed accumulation of cells at G(2)/M phase and of the sub-G(1) population in a time-dependent manner, representing apoptotic cells. IN-2001-mediated cell cycle arrest was associated with HDAC inhibitor-mediated induction of CDK inhibitor expression. In MCF-7 cells, IN-2001 significantly increased p21(WAF1) expression. CONCLUSIONS: In summary, cyclin-dependent kinase (CDK) induced growth inhibition, possibly through modulation of cell cycle and apoptosis regulatory proteins, such as CDK inhibitors, and cyclins. Taken together, these results provide an insight into the utility of HDAC inhibitors as a novel chemotherapeutic regime for hormone-sensitive and insensitive breast cancer.

16.
Mol Cells ; 33(3): 309-16, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22434383

ABSTRACT

To identify specific biomarkers generated upon exposure of L5178Y mouse lymphoma cells to carcinogens, 2-DE and MALDI-TOF MS analysis were conducted using the cellular proteome of L5178Y cells that had been treated with the known carcinogens, 1,2-dibromoethane and O-nitrotoluene and the noncarcinogens, emodin and D-mannitol. Eight protein spots that showed a greater than 1.5-fold increase or decrease in intensity following carcinogen treatment compared with treatment with noncarcinogens were selected. Of the identified proteins, we focused on the candidate biomarker ERM-binding phosphoprotein 50 (EBP50), the expression of which was specifically increased in response to treatment with the carcinogens. The expression level of EBP50 was determined by western analysis using polyclonal rabbit anti-EBP50 antibody. Further, the expression level of EBP50 was increased in cells treated with seven additional carcinogens, verifying that EBP50 could serve as a specific biomarker for carcinogens.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinogens/pharmacology , Phosphoproteins/metabolism , Proteome/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Ethylene Dibromide/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Lymphoma , Mice , Phosphoproteins/genetics , Sodium-Hydrogen Exchangers/genetics , Toluene/analogs & derivatives , Toluene/pharmacology
17.
Mol Cells ; 33(2): 203-10, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22358511

ABSTRACT

1,2-Dibromoethane and glycidol are well known genotoxic carcinogens, which have been widely used in industry. To identify a specific biomarker for these carcinogens in cells, the cellular proteome of L5178Y mouse lymphoma cells treated with these compounds was analyzed by 2-dimensional gel electrophoresis (2-DE) and MALDI-TOF mass spectrometry (MS). Of 50 protein spots showing a greater than 1.5-fold increase or decrease in intensity compared to control cells on a 2-D gel, we focused on the candidate biomarker moesin. Western analysis using monoclonal rabbit anti-moesin confirmed the identity of the protein and its increased level of expression upon exposure to the carcinogenic compounds. Moesin expression also increased in cells treated with six additional genotoxic carcinogens, verifying that moesin could serve as a biomarker to monitor phenotypic change upon exposure to genotoxic carcinogens in L5178Y mouse lymphoma cells.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinogens/toxicity , DNA Damage , Epoxy Compounds/toxicity , Ethylene Dibromide/toxicity , Microfilament Proteins/metabolism , Propanols/toxicity , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Mice , Microfilament Proteins/genetics , Proteome , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Up-Regulation
18.
Biomol Ther (Seoul) ; 20(1): 81-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-24116279

ABSTRACT

Histone deacetylases (HDACs) are enzymes involved in the remodelling of chromatin, and have a key role in the epigenetic regulation of gene expression. Histone deacetylase (HDAC) inhibitors are emerging as an exciting new class of potential anti-cancer agents. In recent years, a number of structurally diverse HDAC inhibitors have been identified and these HDAC inhibitors induce growth arrest, differentiation and/or apoptosis of cancer cells in vitro and in vivo. However, the underlying molecular mechanisms remain unclear. This study aimed at investigating the anti-tumor activity of various HDAC inhibitors, IN-2001, using T47D human breast cancer cells. Moreover, the possible mechanism by which HDAC inhibitors exhibit anti-tumor activity was also explored. In estrogen receptor positive T47D cells, IN-2001, HDAC inhibitor showed anti-proliferative effects in dose-and time-dependent manner. In T47D human breast cancer cells showed anti-tumor activity of IN-2001 and the growth inhibitory effects of IN-2001 were related to the cell cycle arrest and induction of apoptosis. Flow cytometry studies revealed that IN-2001 showed accumulation of cells at G2/M phase. At the same time, IN-2001 treatment time-dependently increased sub-G1 population, representing apoptotic cells. IN-2001-mediated cell cycle arrest was associated with induction of cdk inhibitor expression. In T47D cells, IN-2001 as well as other HDAC inhibitors treatment significantly increased p21(WAF1) and p27(KIP1) expression. In addition, thymidylate synthase, an essential enzyme for DNA replication and repair, was down-regulated by IN-2001 and other HDAC inhibitors in the T47D human breast cancer cells. In summary, IN-2001 with a higher potency than other HDAC inhibitors induced growth inhibition, cell cycle arrest, and eventual apoptosis in human breast cancer possibly through modulation of cell cycle and apoptosis regulatory proteins, such as cdk inhibitors, cyclins, and thymidylate synthase.

19.
Biomol Ther (Seoul) ; 20(3): 313-9, 2012 May.
Article in English | MEDLINE | ID: mdl-24130929

ABSTRACT

In recent years, inhibition of HDACs has emerged as a potential strategy to reverse aberrant epigenetic changes associated with cancer, and several classes of HDAC inhibitors have been found to have potent and specific anticancer activities in preclinical studies. But their precise mechanism of action has not been elucidated. In this study, a novel synthetic inhibitor of HDAC, 3-(4-dimethylamino phenyl)-N-hydroxy-2-propenamide [IN-2001] was examined for its antitumor activity and the underlying molecular mechanisms of any such activity on human breast cancer cell lines. IN-2001 effectively inhibited cellular HDAC activity (IC50 = 0.585 nM) in MDA-MB-231 human breast cancer cells. IN-2001 caused a significant dose-dependent inhibition of cell proliferation in estrogen receptor (ER) negative MDA-MB-231 human breast cancer cells. Cell cycle analysis revealed that the gowth inhibitory effects of IN-2001 might be attributed to cell cycle arrest at G0/G1 and/or G2/Mphase and subsequent apoptosis in human breast cancer cells. These events are accompanied by modulating several cell cycle and apoptosis regulatory genes such as CDK inhibitors p21(WAF1) and p27(KIP1) cyclin D1, and other tumor suppressor genes such as cyclin D2. Collectively, IN-2001 inhibited cell proliferation and induced apoptosis in human breast cancer cells and these findings may provide new therapeutic approaches, combination of antiestrogen together with a HDAC inhibitor, in the hormonal therapy-resistant ER-negative breast cancers. In summary, our data suggest that this histone deacetylase inhibitor, IN-2001, is a novel promising therapeutic agent with potent antitumor effects against human breast cancers.

20.
Bioorg Med Chem Lett ; 21(20): 6049-53, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21911290

ABSTRACT

A series of 1-substituted-3-(6-methylpyridin-2-yl)-4-([1,2,4]triazolo[1,5-a]pyridin-6-yl)pyrazoles 14a-ae, 16a, 16b, and 21a-c has been prepared and evaluated for their ALK5 inhibitory activity in an enzyme assay and in a cell-based luciferase reporter assay. The 4-([1,2,4]triazolo[1,5-a]pyridin-6-yl)-N-(4-methoxyphenyl)-3-(6-methylpyridin-2-yl)-1H-pyrazole-1-carbothioamide (14n) inhibited ALK5 phosphorylation with IC(50) value of 0.57 nM and showed 94% inhibition at 100 nM in a luciferase reporter assay using HaCaT cells permanently transfected with p3TP-luc reporter construct.


Subject(s)
Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazoles/chemistry , Pyrazoles/pharmacology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Animals , Cell Line , Humans , Insecta/cytology , Luciferases/analysis , Protein Kinase Inhibitors/chemical synthesis , Protein Serine-Threonine Kinases/metabolism , Pyrazoles/chemical synthesis , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...