Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Trends Cancer ; 7(6): 511-524, 2021 06.
Article in English | MEDLINE | ID: mdl-33358571

ABSTRACT

Despite the dramatic advances in cancer research over the decades, effective therapeutic strategies are still urgently needed. Increasing evidence indicates that connective tissue growth factor (CTGF), a multifunctional signaling modulator, promotes cancer initiation, progression, and metastasis by regulating cell proliferation, migration, invasion, drug resistance, and epithelial-mesenchymal transition (EMT). CTGF is also involved in the tumor microenvironment in most of the nodes, including angiogenesis, inflammation, and cancer-associated fibroblast (CAF) activation. In this review, we comprehensively discuss the expression of CTGF and its regulation, oncogenic role, clinical relevance, targeting strategies, and therapeutic agents. Herein, we propose that CTGF is a promising cancer therapeutic target that could potentially improve the clinical outcomes of cancer patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Connective Tissue Growth Factor/antagonists & inhibitors , Neoplasms/drug therapy , Oncogene Proteins/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Clinical Trials as Topic , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Curcumin/pharmacology , Curcumin/therapeutic use , Disease Progression , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Extracellular Matrix/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Mice , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Prognosis , Signal Transduction/drug effects , Survival Rate , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
2.
J Pharmacol Exp Ther ; 373(2): 279-289, 2020 05.
Article in English | MEDLINE | ID: mdl-32102917

ABSTRACT

Cholangiocarcinoma (CCA) is a malignant tumor that arises from the epithelial cells of the bile duct and is notorious for its poor prognosis. The clinical outcome remains disappointing, and thus more effective therapeutic options are urgently required. Cordycepin, a traditional Chinese medicine, provides multiple pharmacological strategies in antitumors, but its mechanisms have not been fully elucidated. In this study, we reported that cordycepin inhibited the viability and proliferation capacity of CCA cells in a time- and dose-dependent manner determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) and colony formation assay. Flow cytometry and Hoechst dye showed that cordycepin induced cancer cell apoptosis via extracellular signal-regulated kinase (ERK) 1/2 deactivation. Moreover, cordycepin significantly reduced the angiogenetic capabilities of CCA in vitro as examined by tube formation assay. We also discovered that cordycepin inhibited DEK expression by using Western blot assay. DEK serves as an oncogenic protein that is overexpressed in various gastrointestinal tumors. DEK silencing inhibited CCA cell viability and angiogenesis but not apoptosis induction determined by Western blot and flow cytometry. Furthermore, cordycepin significantly inhibited tumor growth and angiogenic capacities in a xenograft model by downregulating the expression of DEK, phosphorylated ERK1/2 CD31 and von Willebrand factor (vWF). Taken together, we demonstrated that cordycepin inhibited CCA cell proliferation and angiogenesis with a DEK interaction via downregulation in ERK signaling. These data indicate that cordycepin may serve as a novel agent for CCA clinical treatment and prognosis improvement. SIGNIFICANCE STATEMENT: Cordycepin provides multiple strategies in antitumors, but its mechanisms are not fully elucidated, especially on cholangiocarcinoma (CCA). We reported that cordycepin inhibited the viability of CCA cells, induced apoptosis via extracellular signal-regulated kinase 1/2 deactivation and DEK inhibition, and reduced the angiogenetic capabilities of CCA both in vivo and in vitro.


Subject(s)
Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Deoxyadenosines/pharmacology , Extracellular Signal-Regulated MAP Kinases/physiology , MAP Kinase Signaling System/drug effects , Neovascularization, Pathologic/prevention & control , Oncogene Proteins/antagonists & inhibitors , Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors , Animals , Bile Duct Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cholangiocarcinoma/pathology , Humans , Male , Mice , Xenograft Model Antitumor Assays
3.
Biochem Biophys Res Commun ; 508(1): 102-108, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30471851

ABSTRACT

The NSD family (NSD1, NSD2/MMSET/WHSC1, and NSD3/WHSC1L1) are histone lysine methyltransferases (HMTases) essential for chromatin regulation. The NSDs are oncoproteins, drivers of a number of tumors and are considered important drug-targets but the lack of potent and selective inhibitors hampers further therapeutic development and limits exploration of their biology. In particular, MMSET/NSD2 selective inhibition is being pursued for therapeutic interventions against multiple myeloma (MM) cases, especially in multiple myeloma t(4;14)(p16.3;q32) translocation that is associated with a significantly worse prognosis than other MM subgroups. Multiple myeloma is the second most common hematological malignancy, after non-Hodgkin lymphoma and remains an incurable malignancy. Here we report the discovery of LEM-14, an NSD2 specific inhibitor with an in vitro IC50 of 132 µM and that is inactive against the closely related NSD1 and NSD3. LEM-14-1189, a LEM-14 derivative, differentially inhibits the NSDs with in vitro IC50 of 418 µM (NSD1), IC50 of 111 µM (NSD2) and IC50 of 60 µM (NSD3). We propose LEM-14 and derivative LEM-14-1189 as tools for studying the biology of the NSDs and constitute meaningful steps toward potent NSDs therapeutic inhibitors.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Oncogene Proteins/antagonists & inhibitors , Repressor Proteins/antagonists & inhibitors , Catalytic Domain , Drug Design , Drug Discovery , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Humans , Kinetics , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Protein Conformation , Repressor Proteins/chemistry , Repressor Proteins/genetics , User-Computer Interface
4.
Fungal Genet Biol ; 116: 33-41, 2018 07.
Article in English | MEDLINE | ID: mdl-29684553

ABSTRACT

Many human cancer cells contain more than two centrosomes, yet these cancer cells can form pseudo-bipolar spindles through the mechanism, called centrosome clustering, and survive, instead of committing lethal multipolar mitoses. Kinesin-14/HSET, a minus end-directed motor, plays a crucial role in centrosome clustering. Accordingly, HSET is deemed to be a promising chemotherapeutic target to selectively kill cancer cells. Recently, three HSET inhibitors (AZ82, CW069 and SR31527) have been reported, but their specificity and efficacy have not been evaluated rigorously. This downside partly stems from the lack of robust systems for the assessment of these drugs. Yeasts and filamentous fungi provide not only powerful models for basic and applied biology but also versatile tools for drug discovery and evaluation. Here we show that these three inhibitors on their own are cytotoxic to fission yeast, suggesting that they have off-targets in vivo except for kinesin-14. Nonetheless, intriguingly, AZ82 can neutralize otherwise toxic overproduced HSET; this includes a substantial reduction in the percentage of HSET-driven abnormal mitotic cells and partial suppression of its lethality. SR31527 also displays modest neutralizing activity, while we do not detect such activity in CW069. As an experimental proof-of-principle study, we have treated HSET-overproducing fission yeast cells with extracts prepared from various plant species and found activities that rescue HSET-driven lethality in those from Chamaecyparis pisifera and Toxicodendron trichocarpum. This methodology of protein overproduction in fission yeast, therefore, provides a convenient, functional assay system by which to screen for not only selective human kinesin-14 inhibitors but also those against other molecules of interest.


Subject(s)
Kinesins/antagonists & inhibitors , Kinesins/biosynthesis , Oncogene Proteins/antagonists & inhibitors , Schizosaccharomyces/genetics , Alanine/analogs & derivatives , Alanine/pharmacology , Drug Evaluation, Preclinical/methods , Humans , Kinesins/genetics , Kinesins/metabolism , Plant Extracts/pharmacology , Pyridines/pharmacology , Schizosaccharomyces/drug effects , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism
5.
Cancer Biol Ther ; 16(6): 821-33, 2015.
Article in English | MEDLINE | ID: mdl-25897893

ABSTRACT

Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase that plays a significant role in mitotic progression and cellular responses to DNA damage. While traditionally viewed as a tumor suppressor, inhibition of PP2A has recently come to attention as a novel therapeutic means of driving senescent cancer cells into mitosis and promoting cell death via mitotic catastrophe. These findings have been corroborated in numerous studies utilizing naturally produced compounds that selectively inhibit PP2A. To overcome the known human toxicities associated with these compounds, a water-soluble small molecule inhibitor, LB100, was recently developed to competitively inhibit the PP2A protein. This review summarizes the pre-clinical studies to date that have demonstrated the anti-cancer activity of LB100 via its chemo- and radio-sensitizing properties. These studies demonstrate the tremendous therapeutic potential of LB100 in a variety of cancer types. The results of an ongoing phase 1 trial are eagerly anticipated.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Enzyme Inhibitors/pharmacology , Piperazines/pharmacology , Protein Phosphatase 2/antagonists & inhibitors , Radiation-Sensitizing Agents/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , Humans , Mitosis/drug effects , Mitosis/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Tumor Protein, Translationally-Controlled 1 , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Wnt Signaling Pathway/drug effects
6.
Clin Ther ; 35(9): 1271-81, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24054704

ABSTRACT

BACKGROUND: Once a promising drug target is identified, the steps to actually discover and optimize a drug are diverse and challenging. OBJECTIVE: The goal of this study was to provide a road map to navigate drug discovery. METHODS: Review general steps for drug discovery and provide illustrating references. RESULTS: A number of approaches are available to enhance and accelerate target identification and validation. Consideration of a variety of potential mechanisms of action of potential drugs can guide discovery efforts. The hit to lead stage may involve techniques such as high-throughput screening, fragment-based screening, and structure-based design, with informatics playing an ever-increasing role. Biologically relevant screening models are discussed, including cell lines, 3-dimensional culture, and in vivo screening. The process of enabling human studies for an investigational drug is also discussed. CONCLUSIONS: Drug discovery is a complex process that has significantly evolved in recent years.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Drugs, Investigational/pharmacology , Molecular Targeted Therapy , Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Cell Line, Tumor , Computational Biology , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Drugs, Investigational/therapeutic use , High-Throughput Screening Assays , Humans , Molecular Docking Simulation , Neoplasms/genetics , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Reproducibility of Results , United States , United States Food and Drug Administration
7.
Bioorg Med Chem ; 16(21): 9420-4, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18842418

ABSTRACT

The aberrant hedgehog (Hh)/GLI signaling pathway causes the formation and progression of a variety of tumors. By screening tropical plant extracts by using our screening system, Zizyphus cambodiana was found to include Hh/GLI signaling inhibitors. Bioassay-guided fractionation of this plant extract led to the isolation of three active pentacyclic triterpenes, colubrinic acid (1), betulinic acid (2) and alphitolic acid (3), as potent inhibitors. The inhibition of GLI-related protein expression with 1 or 2 was observed in HaCaT cells with exogenous GLI1, or human pancreatic cancer cells (PANC1), which express Hh/GLI components aberrantly. The expressions of GLI-related proteins PTCH and BCL2 were clearly inhibited by 1 or 2. We also examined the cytotoxicity of these active compounds against PANC1, human prostate cancer cells (DU145) and mouse embryo fibroblast cells (C3H10T1/2). The cytotoxicity against cancer cells (PANC1 and DU145) by 1 or 2 would be caused by inhibition of the expression of the anti-apoptosis protein BCL2. These pentacyclic triterpene inhibitors showed an important relationship between Hh/GLI signaling inhibition, the decrease of BCL2, and cytotoxicity against cancer cells.


Subject(s)
Oncogene Proteins/antagonists & inhibitors , Plant Extracts/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Trans-Activators/antagonists & inhibitors , Transcription, Genetic/drug effects , Ziziphus/chemistry , Animals , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Fibroblasts/drug effects , Humans , Male , Maytansine/analogs & derivatives , Maytansine/chemistry , Maytansine/pharmacology , Mice , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Patched Receptors , Patched-1 Receptor , Pentacyclic Triterpenes , Phenols/chemistry , Phenols/pharmacology , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/chemistry , Phenylethyl Alcohol/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Trans-Activators/metabolism , Triterpenes/chemistry , Triterpenes/pharmacology , Zinc Finger Protein GLI1 , Zinc Fingers , Betulinic Acid
8.
Chembiochem ; 9(7): 1082-92, 2008 May 05.
Article in English | MEDLINE | ID: mdl-18357592

ABSTRACT

The aberrant hedgehog (Hh)/GLI signaling pathway causes the formation and progression of a variety of tumors. To search for Hh/GLI inhibitors, we screened for naturally occurring inhibitors of the transcriptional activator GLI1 by using a cell-based assay. We identified zerumbone (1), zerumbone epoxide (2), staurosporinone (9), 6-hydroxystaurosporinone (10), arcyriaflavin C (11) and 5,6-dihydroxyarcyriaflavin A (12) as inhibitors of GLI-mediated transcription. In addition, we isolated physalins F (17) and B (18) from Physalis minima, which are also potent inhibitors. These compounds also inhibited GLI2-mediated transactivation. Semiquantitative RT-PCR and Western blotting analysis further revealed that 1, 9, 17, and 18 decreased Hh-related component expressions. We also show that inhibitors of GLI-mediated transactivation reduce the level of the antiapoptosis Bcl2 expression. Finally, these identified compounds were cytotoxic to PANC1 pancreatic cancer cells, which express Hh/GLI components. These results strongly suggest that the cytotoxicity of the compounds to PANC1 cells correlates with their inhibition of GLI-mediated transcription.


Subject(s)
Biological Products/chemistry , Biological Products/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Oncogene Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Trans-Activators/antagonists & inhibitors , Transcription, Genetic/drug effects , Animals , Cell Line, Tumor , Drug Evaluation, Preclinical , Gene Expression Regulation, Neoplastic/drug effects , Hedgehog Proteins/metabolism , Humans , Inhibitory Concentration 50 , Oncogene Proteins/metabolism , Plant Extracts/chemistry , Plant Extracts/pharmacology , Signal Transduction/drug effects , Trans-Activators/metabolism , Transcriptional Activation/drug effects , Zinc Finger Protein GLI1
9.
Hematol Oncol Clin North Am ; 15(3): 547-57, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11525296

ABSTRACT

Given the poor results with currently available therapies, it is imperative that new treatments be developed for patients with advanced prostate cancer. The next generation of therapies will include many novel biologic agents targeted at molecular defects in the cancer cell. Investigating the efficacy and safety of these compounds and evaluating their utility in combination with traditional therapies such as chemotherapy or radiotherapy are major goals of prostate cancer clinical research for the next decade.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Prostatic Neoplasms/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/classification , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Clinical Trials as Topic , Drug Design , Drug Screening Assays, Antitumor , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Growth Substances/physiology , Humans , Male , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/physiology , Neovascularization, Pathologic/drug therapy , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/physiology , Phytotherapy , Plant Preparations/therapeutic use , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/physiology , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Signal Transduction/drug effects , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL