mk-2206 and Prostatic-Neoplasms

mk-2206 has been researched along with Prostatic-Neoplasms* in 6 studies

Other Studies

6 other study(ies) available for mk-2206 and Prostatic-Neoplasms

ArticleYear
Adapted suspension tumor cells rewire metabolic pathways for anchorage-independent survival through AKT activation.
    Experimental cell research, 2022, 02-15, Volume: 411, Issue:2

    Metastatic spread of cancer cells is the main cause of cancer-related death. As cancer cells adapt themselves in a suspended state in the blood stream before penetration and regrowth at distal tissues, understanding their survival strategy in an anchorage-independent condition is important to develop appropriate therapeutics. We have previously generated adapted suspension cells (ASCs) from parental adherent cancer cells to study the characteristics of circulating tumor cells. In this study, we explored metabolic rewiring in MDA-MB-468 ASCs to adapt to suspension growth conditions through extracellular flux analyses and various metabolic assays. We also determined the relationship between AKT activation and metabolic rewiring in ASCs using the AKT inhibitor, MK2206. ASCs reprogramed metabolism to enhance glycolysis and basal oxygen consumption rate. RNA-sequencing analysis revealed the upregulation in the genes related to glycolysis, tricarboxylic acid cycle, and oxidative phosphorylation. The changes in the metabolic program led to a remarkable dependency of ASCs on carbohydrates as an energy source for proliferation as compared to parental adherent cells (ADs). AKT activation was observed in ASCs and those generated from pancreatic and other breast cancer cells, and AKT activation inhibition in ASCs decreased glycolysis and oxygen consumption. AKT activation is an important strategy for obtaining energy through the enhancement of glycolysis in ASCs. The regulation of AKT activity and/or glycolysis may provide a strong therapeutic strategy to prevent the metastatic spread of cancer cells.

    Topics: Adaptation, Physiological; Breast Neoplasms; Cell Adhesion; Cell Culture Techniques; Cell Line, Tumor; Cell Survival; Doxorubicin; Female; Gene Expression Regulation, Neoplastic; Glycolysis; Heterocyclic Compounds, 3-Ring; Humans; Male; Metabolic Networks and Pathways; Neoplastic Cells, Circulating; Oxidative Phosphorylation; Oxygen Consumption; Pancreatic Neoplasms; Prostatic Neoplasms; Proto-Oncogene Proteins c-akt

2022
The antiandrogenic effect of neferine, liensinine, and isoliensinine by inhibiting 5-α-reductase and androgen receptor expression via PI3K/AKT signaling pathway in prostate cancer.
    Die Pharmazie, 2021, 05-01, Volume: 76, Issue:5

    Topics: 5-alpha Reductase Inhibitors; Amino Acid Chloromethyl Ketones; Androgen Antagonists; Androgen Receptor Antagonists; Antineoplastic Agents; Apoptosis; Apoptosis Regulatory Proteins; Autophagy; Benzylisoquinolines; Biological Products; Cell Line, Tumor; Cell Movement; Chromones; Heterocyclic Compounds, 3-Ring; Humans; Isoquinolines; Male; Morpholines; Nelumbo; Phenols; Phosphatidylinositol 3-Kinases; Prostatic Neoplasms; Proto-Oncogene Proteins c-akt; Signal Transduction

2021
AZD5153 Inhibits Prostate Cancer Cell Growth in Vitro and in Vivo.
    Cellular physiology and biochemistry : international journal of experimental cellular physiology, biochemistry, and pharmacology, 2018, Volume: 50, Issue:2

    Bromodomain-containing protein 4 (BRD4) overexpression participates in prostate cancer progression by enhancing the transcriptional activity and expression of several key oncogenes. AZD5153 is a novel BRD4 inhibitor.. Prostate cancer cells were treated with AZD5153. Cell survival was tested by MTT assay and clonogenicity assay. Cell proliferation was tested by [H3] DNA incorporation assay. Cell apoptosis was tested by caspase-3/-9 activity assay, Histone DNA ELISA assay, Annexin V FACS assay and TUNEL staining assay. Cell cycle progression was tested by propidium iodide (PI) FACS assay. Signaling was tested by Western blotting assay. The nude mice PC-3 xenograft model was applied to test AZD5153's activity in vivo.. AZD5153 inhibited proliferation and survival of established and primary prostate cancer cells. AZD5153 induced apoptosis activation and cell cycle arrest in prostate cancer cells. AZD5153 was non-cytotoxic to the prostate epithelial cells. AZD5153 downregulated BRD4 targets (cyclin D1, Myc, Bcl-2, FOSL1 and CDK4) in PC-3 and primary prostate cancer cells. Further studies show that AKT could be the primary resistance factor of AZD5153. Pharmacological inhibition or genetic depletion of AKT induced BRD4 downregulation, sensitizing AZD5153-induced cytotoxicity in PC-3 cells. In vivo, AZD5153 oral administration inhibited PC-3 xenograft tumor growth in nude mice. Its anti-tumor activity was further enhanced with co-treatment of the AKT specific inhibitor MK-2206.. Together, our results indicate a promising therapeutic value of the novel BRD4 inhibitor AZD5153 against prostate cancer cells.

    Topics: Administration, Oral; Animals; Apoptosis; B-Cell Lymphoma 3 Protein; Cell Cycle Checkpoints; Cell Cycle Proteins; Cell Proliferation; Cyclin D1; Down-Regulation; Heterocyclic Compounds, 2-Ring; Heterocyclic Compounds, 3-Ring; Humans; Male; Mice; Mice, Nude; Nuclear Proteins; Piperazines; Prostatic Neoplasms; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-akt; Pyrazoles; Pyridazines; RNA Interference; RNA, Small Interfering; Transcription Factors; Tumor Cells, Cultured

2018
Metabolic biomarkers of response to the AKT inhibitor MK-2206 in pre-clinical models of human colorectal and prostate carcinoma.
    British journal of cancer, 2018, Volume: 119, Issue:9

    AKT is commonly overexpressed in tumours and plays an important role in the metabolic reprogramming of cancer. We have used magnetic resonance spectroscopy (MRS) to assess whether inhibition of AKT signalling would result in metabolic changes that could potentially be used as biomarkers to monitor response to AKT inhibition.. Cellular and metabolic effects of the allosteric AKT inhibitor MK-2206 were investigated in HT29 colon and PC3 prostate cancer cells and xenografts using flow cytometry, immunoblotting, immunohistology and MRS.. In vitro treatment with MK-2206 inhibited AKT signalling and resulted in time-dependent alterations in glucose, glutamine and phospholipid metabolism. In vivo, MK-2206 resulted in inhibition of AKT signalling and tumour growth compared with vehicle-treated controls. In vivo MRS analysis of HT29 subcutaneous xenografts showed similar metabolic changes to those seen in vitro including decreases in the tCho/water ratio, tumour bioenergetic metabolites and changes in glutamine and glutathione metabolism. Similar phosphocholine changes compared to in vitro were confirmed in the clinically relevant orthotopic PC3 model.. This MRS study suggests that choline metabolites detected in response to AKT inhibition are time and microenvironment-dependent, and may have potential as non-invasive biomarkers for monitoring response to AKT inhibitors in selected cancer types.

    Topics: Animals; Antineoplastic Agents; Biomarkers, Tumor; Cell Line, Tumor; Colorectal Neoplasms; Enzyme Inhibitors; Heterocyclic Compounds, 3-Ring; Heterografts; Humans; Magnetic Resonance Spectroscopy; Male; Prostatic Neoplasms; Proto-Oncogene Proteins c-akt

2018
An Atomic Force Microscope Study Revealed Two Mechanisms in the Effect of Anticancer Drugs on Rate-Dependent Young's Modulus of Human Prostate Cancer Cells.
    PloS one, 2015, Volume: 10, Issue:5

    Mechanical properties of cells have been recognized as a biomarker for cellular cytoskeletal organization. As chemical treatments lead to cell cytoskeletal rearrangements, thereby, modifications of cellular mechanical properties, investigating cellular mechanical property variations provides insightful knowledge to effects of chemical treatments on cancer cells. In this study, the effects of eight different anticancer drugs on the mechanical properties of human prostate cancer cell (PC-3) are investigated using a recently developed control-based nanoindentation measurement (CNM) protocol on atomic force microscope (AFM). The CNM protocol overcomes the limits of other existing methods to in-liquid nanoindentation measurement of live cells on AFM, particularly for measuring mechanical properties of live cells. The Young's modulus of PC-3 cells treated by the eight drugs was measured by varying force loading rates over three orders of magnitude, and compared to the values of the control. The results showed that the Young's modulus of the PC-3 cells increased substantially by the eight drugs tested, and became much more pronounced as the force load rate increased. Moreover, two distinct trends were clearly expressed, where under the treatment of Disulfiram, paclitaxel, and MK-2206, the exponent coefficient of the frequency- modulus function remained almost unchanged, while with Celebrex, BAY, Totamine, TPA, and Vaproic acid, the exponential rate was significantly increased.

    Topics: Amino Acids; Celecoxib; Cell Line, Tumor; Cytoskeleton; Disulfiram; Elastic Modulus; Fluorescent Antibody Technique; Heterocyclic Compounds, 3-Ring; Humans; Male; Microscopy, Atomic Force; Models, Theoretical; Paclitaxel; Prostatic Neoplasms

2015
Dual targeting of the Akt/mTOR signaling pathway inhibits castration-resistant prostate cancer in a genetically engineered mouse model.
    Cancer research, 2012, Sep-01, Volume: 72, Issue:17

    Although the prognosis for clinically localized prostate cancer is now favorable, there are still no curative treatments for castration-resistant prostate cancer (CRPC) and, therefore, it remains fatal. In this study, we investigate a new therapeutic approach for treatment of CRPC, which involves dual targeting of a major signaling pathway that is frequently deregulated in the disease. We found that dual targeting of the Akt and mTOR signaling pathways with their respective inhibitors, MK-2206 and ridaforolimus (MK-8669), is highly effective for inhibiting CRPC in preclinical studies in vivo using a refined genetically engineered mouse model of the disease. The efficacy of the combination treatment contrasts with their limited efficacy as single agents, since delivery of MK-2206 or MK-8669 individually had a modest impact in vivo on the overall tumor phenotype. In human prostate cancer cell lines, although not in the mouse model, the synergistic actions of MK-2206 and ridaforolimus (MK-8669) are due in part to limiting the mTORC2 feedback activation of Akt. Moreover, the effects of these drugs are mediated by inhibition of cellular proliferation via the retinoblastoma (Rb) pathway. Our findings suggest that dual targeting of the Akt and mTOR signaling pathways using MK-2206 and ridaforolimus (MK-8669) may be effective for treatment of CRPC, particularly for patients with deregulated Rb pathway activity.

    Topics: Animals; Antineoplastic Agents; Cell Line, Tumor; Cluster Analysis; Disease Models, Animal; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Heterocyclic Compounds, 3-Ring; Humans; Male; Mice; Mice, Transgenic; Orchiectomy; Phenotype; Prostatic Neoplasms; Proto-Oncogene Proteins c-akt; Retinoblastoma Protein; Signal Transduction; Sirolimus; TOR Serine-Threonine Kinases; Tumor Burden; Xenograft Model Antitumor Assays

2012