1
|
Tian G, Chen Z, Shi K, Wang X, Xie L, Yang F. The evolution of small-molecule Akt inhibitors from hit to clinical candidate. Eur J Med Chem 2024; 279:116906. [PMID: 39353238 DOI: 10.1016/j.ejmech.2024.116906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024]
Abstract
Akt, a key regulator of cell survival, proliferation, and metabolism, has become a prominent target for treatment of cancer and inflammatory diseases. The journey of small-molecule Akt inhibitors from discovery to the clinic has faced numerous challenges, with a significant emphasis on optimization throughout the development process. Early discovery efforts identified various classes of inhibitors, including ATP-competitive and allosteric modulators. However, during preclinical and clinical development, several issues arose, including poor specificity, limited bioavailability, and toxicity. Optimization efforts have been central to overcoming these hurdles. Researchers focused on enhancing the selectivity of inhibitors to target Akt isoforms more precisely, reducing off-target effects, and improving pharmacokinetic properties to ensure better bioavailability and distribution. Structural modifications and the design of prodrugs have played a crucial role in refining the efficacy and safety profile of these inhibitors. Additionally, efforts have been made to optimize the therapeutic window, balancing effective dosing with minimal adverse effects. The review highlights how these optimization strategies have been key in advancing small-molecule Akt inhibitors toward clinical success and underscores the importance of continued refinement in their development.
Collapse
Affiliation(s)
- Gengren Tian
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhuo Chen
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Keqing Shi
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xinwai Wang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Lijuan Xie
- Department of Vascularsurgery, China-Japan Union Hospital of Jilin University, Changchun, China.
| | - Fuwei Yang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
2
|
Altıntop MD, Ertorun İ, Akalın Çiftçi G, Özdemir A. Design, synthesis and biological evaluation of a new series of imidazothiazole-hydrazone hybrids as dual EGFR and Akt inhibitors for NSCLC therapy. Eur J Med Chem 2024; 276:116698. [PMID: 39047611 DOI: 10.1016/j.ejmech.2024.116698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/24/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
In search of small molecules for targeted therapy of non-small cell lung carcinoma (NSCLC), an efficient four-step synthetic route was followed for the synthesis of new imidazothiazole-hydrazone hybrids, which were assessed for their cytotoxic effects on human lung adenocarcinoma (A549) and human lung fibroblast (CCD-19Lu) cells. Among them, compounds 4, 6, 13, 16, 17 and 21 exhibited selective cytotoxic activity against A549 cell line. In vitro mechanistic studies were performed to assess their effects on apoptosis, caspase-3, cell cycle, EGFR and Akt in A549 cells. Compounds 6, 16, 17 and 21 promoted apoptotic cell death more than erlotinib. According to the in vitro data, it is quite clear that compound 6 promotes apoptosis through caspase-3 activation and arrests the cell cycle at the G0/G1 phase in A549 cells. Compounds 16 and 17 arrested the cell cycle at the S phase, whereas compounds 4, 13 and 21 caused the cell cycle arrest at the G2/M phase. The most effective EGFR inhibitor in this series was found as compound 13, followed by compounds 17 and 16. Furthermore, Akt inhibitory effects of compounds 16 and 17 in A549 cells were close to that of GSK690693. In particular, it can be concluded that the cytotoxic and apoptotic effects of compounds 16 and 17 are associated with their inhibitory effects on both EGFR and Akt. Molecular docking studies suggest that compounds 16 and 17 interact with crucial amino acid residues in the binding sites of human EGFR (PDB ID: 1M17) and Akt2 (PDB ID: 3D0E). Based on the in silico data, both compounds are predicted to possess favorable oral bioavailability and drug-likeness. Further studies are required to benefit from these compounds as anticancer agents for targeted therapy of NSCLC.
Collapse
Affiliation(s)
- Mehlika Dilek Altıntop
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey
| | - İpek Ertorun
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey
| | - Gülşen Akalın Çiftçi
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey
| | - Ahmet Özdemir
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey.
| |
Collapse
|
3
|
Nussinov R, Jang H. The value of protein allostery in rational anticancer drug design: an update. Expert Opin Drug Discov 2024; 19:1071-1085. [PMID: 39068599 PMCID: PMC11390313 DOI: 10.1080/17460441.2024.2384467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION Allosteric drugs are advantageous. However, they still face hurdles, including identification of allosteric sites that will effectively alter the active site. Current strategies largely focus on identifying pockets away from the active sites into which the allosteric ligand will dock and do not account for exactly how the active site is altered. Favorable allosteric inhibitors dock into sites that are nearby the active sites and follow nature, mimicking diverse allosteric regulation strategies. AREAS COVERED The following article underscores the immense significance of allostery in drug design, describes current allosteric strategies, and especially offers a direction going forward. The article concludes with the authors' expert perspectives on the subject. EXPERT OPINION To select a productive venue in allosteric inhibitor development, we should learn from nature. Currently, useful strategies follow this route. Consider, for example, the mechanisms exploited in relieving autoinhibition and in harnessing allosteric degraders. Mimicking compensatory, or rescue mutations may also fall into such a thesis, as can molecular glues that capture features of scaffolding proteins. Capturing nature and creatively tailoring its mimicry can continue to innovate allosteric drug discovery.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD, USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD, USA
| |
Collapse
|
4
|
Etikyala U, Reddyrajula R, Vani T, Kuchana V, Dalimba U, Manga V. An in silico approach to identify novel and potential Akt1 (protein kinase B-alpha) inhibitors as anticancer drugs. Mol Divers 2024:10.1007/s11030-024-10887-9. [PMID: 38796797 DOI: 10.1007/s11030-024-10887-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 04/27/2024] [Indexed: 05/29/2024]
Abstract
Akt1 (protein kinase B) has become a major focus of attention due to its significant functionality in a variety of cellular processes and the inhibition of Akt1 could lead to a decrease in tumour growth effectively in cancer cells. In the present work, we discovered a set of novel Akt1 inhibitors by using multiple computational techniques, i.e. pharmacophore-based virtual screening, molecular docking, binding free energy calculations, and ADME properties. A five-point pharmacophore hypothesis was implemented and validated with AADRR38. The obtained R2 and Q2 values are in the acceptable region with the values of 0.90 and 0.64, respectively. The generated pharmacophore model was employed for virtual screening to find out the potential Akt1 inhibitors. Further, the selected hits were subjected to molecular docking, binding free energy analysis, and refined using ADME properties. Also, we designed a series of 6-methoxybenzo[b]oxazole analogues by comprising the structural characteristics of the hits acquired from the database. Molecules D1-D10 were found to have strong binding interactions and higher binding free energy values. In addition, Molecular dynamic simulation was performed to understand the conformational changes of protein-ligand complex.
Collapse
Affiliation(s)
- Umadevi Etikyala
- Medicinal Chemistry Laboratory, Department of Chemistry, Osmania University, Hyderabad, 500076, India
| | - Rajkumar Reddyrajula
- Central Research Facility, National Institute of Technology Karnataka, Surathkal, Mangalore, 575025, India
| | - T Vani
- Medicinal Chemistry Laboratory, Department of Chemistry, Osmania University, Hyderabad, 500076, India
| | - Vinutha Kuchana
- Medicinal Chemistry Laboratory, Department of Chemistry, Osmania University, Hyderabad, 500076, India
| | - Udayakumar Dalimba
- Organic Chemistry Laboratory, Department of Chemistry, National Institute of Technology Karnataka, Surathkal, Mangalore, 575025, India
| | - Vijjulatha Manga
- Medicinal Chemistry Laboratory, Department of Chemistry, Osmania University, Hyderabad, 500076, India.
| |
Collapse
|
5
|
Pervanidis KA, D'Angelo GD, Weisner J, Brandherm S, Rauh D. Akt Inhibitor Advancements: From Capivasertib Approval to Covalent-Allosteric Promises. J Med Chem 2024; 67:6052-6063. [PMID: 38592948 DOI: 10.1021/acs.jmedchem.4c00075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Akt kinase is vital in cell growth, survival, metabolism, and migration. Dysregulation of Akt signaling is implicated in cancer and metabolic disorders. In the context of cancer, overactive Akt promotes cell survival and proliferation. This has spurred extensive research into developing Akt inhibitors as potential therapeutic agents to disrupt aberrant Akt signaling. Akt inhibitors are classified into three main types: ATP-competitive, allosteric, and covalent-allosteric inhibitors (CAAIs). ATP-competitive inhibitors compete with ATP for binding to Akt, allosteric inhibitors interact with the Pleckstrin homology (PH) domain, and covalent-allosteric inhibitors form covalent bonds, making them more potent and selective. Notably, capivasertib (AZD5363), a potent ATP-competitive Akt inhibitor, received FDA approval in November 2023 for use in combination with the estrogen receptor degrader fulvestrant to treat breast cancer. Challenges remain, including improving selectivity, identifying biomarkers to tailor treatments, and enhancing therapeutic efficacy while minimizing adverse effects. Particularly covalent-allosteric inhibitors hold promise for future more effective and personalized treatments.
Collapse
Affiliation(s)
- Kosmas Alexandros Pervanidis
- Department of Chemistry and Chemical Biology, TU Dortmund University and Drug Discovery Hub Dortmund (DDHD), Zentrum für Integrierte Wirkstoffforschung (ZIW), Otto-Hahn-Strasse 4a, 44227 Dortmund, Germany
| | - Giovanni Danilo D'Angelo
- Department of Chemistry and Chemical Biology, TU Dortmund University and Drug Discovery Hub Dortmund (DDHD), Zentrum für Integrierte Wirkstoffforschung (ZIW), Otto-Hahn-Strasse 4a, 44227 Dortmund, Germany
| | - Jörn Weisner
- Department of Chemistry and Chemical Biology, TU Dortmund University and Drug Discovery Hub Dortmund (DDHD), Zentrum für Integrierte Wirkstoffforschung (ZIW), Otto-Hahn-Strasse 4a, 44227 Dortmund, Germany
- KyDo Therapeutics, Otto-Hahn-Strasse 15, 44227 Dortmund, Germany
| | - Sven Brandherm
- Department of Chemistry and Chemical Biology, TU Dortmund University and Drug Discovery Hub Dortmund (DDHD), Zentrum für Integrierte Wirkstoffforschung (ZIW), Otto-Hahn-Strasse 4a, 44227 Dortmund, Germany
- KyDo Therapeutics, Otto-Hahn-Strasse 15, 44227 Dortmund, Germany
| | - Daniel Rauh
- Department of Chemistry and Chemical Biology, TU Dortmund University and Drug Discovery Hub Dortmund (DDHD), Zentrum für Integrierte Wirkstoffforschung (ZIW), Otto-Hahn-Strasse 4a, 44227 Dortmund, Germany
| |
Collapse
|
6
|
Lin H, Ai D, Liu Q, Wang X, Chen Q, Hong Z, Tao Y, Gao J, Wang L. Natural isoflavone glabridin targets PI3Kγ as an adjuvant to increase the sensitivity of MDA-MB-231 to tamoxifen and DU145 to paclitaxel. J Steroid Biochem Mol Biol 2024; 236:106426. [PMID: 37984749 DOI: 10.1016/j.jsbmb.2023.106426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/21/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023]
Abstract
Glabridin is a natural isoflavone with estrogen receptor agonism and significant anti-tumor activity. Additionally, glabridin has a regulation effect on PI3K/AKT/mTOR pathway, but its exact target remains unclear. In this study, we evaluated the antitumor activity of glabridin against breast cancer and prostate cancer cells, and further clarified its targeting to PI3K. We found that glabridin could significantly inhibit the cell viability of human breast cancer and prostate cancer cell lines. It induced caspase activation cascade and cell apoptosis through decreasing the mitochondrial transmembrane potential and increasing the intracellular reactive oxygen species (ROS). Moreover, glabridin could attenuate epithelial-mesenchymal transition (EMT) progression by inhibiting cell migration. PharmMapper calculation showed that PI3Kγ might be the most potential target protein because of the highest Normal Fit score (0.9735) and z'-score (0.9797). Molecular docking and bio-layer interferometry (BLI) analysis further demonstrated the PI3Kγ targeting of glabridin. In vivo experiments showed that glabridin can effectively inhibit the tumor growth of breast cancer xenograft model, and does not show obvious hepatorenal toxicity. Moreover, glabridin could effectively promote the anti-proliferation and pro-apoptotic effects of tamoxifen on MDA-MB-231 cell and taxol on DU145 cell. Elucidating the targeting of glabridin to PI3K may lay a theoretical foundation for the structural derivatization of glabridin, which is expected to greatly promote the application and development of glabridin in the field of cancer therapy.
Collapse
Affiliation(s)
- Hongyan Lin
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Dongxuan Ai
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Qingqing Liu
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Xinling Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Qingqing Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Zhongbin Hong
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Yuheng Tao
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Jian Gao
- School of Medicine, Anhui University of Science and Technology, Huainan 232002, Anhui, China.
| | - Liqun Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| |
Collapse
|
7
|
Hino T, Nakahara F, Miyauchi M, Ito Y, Masamoto Y, Morita K, Kagoya Y, Kojima H, Kurokawa M. AKT2 inhibition accelerates the acquisition of phagocytic ability in induced pluripotent stem cell-derived neutrophils. Exp Hematol 2024; 130:104137. [PMID: 38103826 DOI: 10.1016/j.exphem.2023.104137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 11/01/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Neutrophils are key components of the immune system that inhibit bacterial infections. Systemic bacterial infections can cause lethal conditions, especially in patients with neutropenia associated with chemotherapy or other systemic illnesses; hence, early detection of the symptoms and prompt management are crucial in such cases. Previously, we established expandable engineered neutrophil-primed progenitors (NeuPs-XL) using human-induced pluripotent stem cells (iPSCs), which can produce neutrophil-like cells at a clinically suitable scale within 4 days of inducing myeloid differentiation. In this study, using small-molecule compound-based screening, we detected that MK-2206, a selective pan-AKT inhibitor, can accelerate this differentiation process, promote phagocytic ability in neutrophils, and enhance cytokine and chemokine expression in response to lipopolysaccharides. The inhibition of AKT2 has been identified as the key mechanism underlying this acceleration. These results can make a substantial contribution to the development of strategies for the prompt production of clinically applicable iPSC-derived neutrophils, which can potentially lead to the management of severe infections associated with life-threatening neutropenia and the effective treatment of related health conditions in the future.
Collapse
Affiliation(s)
- Toshiya Hino
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Fumio Nakahara
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masashi Miyauchi
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yusuke Ito
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yosuke Masamoto
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ken Morita
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuki Kagoya
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hirotatsu Kojima
- Drug Discovery Initiative, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Mineo Kurokawa
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Therapy and Transplantation Medicine, The University of Tokyo Hospital, Tokyo, Japan.
| |
Collapse
|
8
|
Kosonocky CW, Feller AL, Wilke CO, Ellington AD. Using alternative SMILES representations to identify novel functional analogues in chemical similarity vector searches. PATTERNS (NEW YORK, N.Y.) 2023; 4:100865. [PMID: 38106612 PMCID: PMC10724362 DOI: 10.1016/j.patter.2023.100865] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/09/2023] [Accepted: 10/06/2023] [Indexed: 12/19/2023]
Abstract
Chemical similarity searches are a widely used family of in silico methods for identifying pharmaceutical leads. These methods historically relied on structure-based comparisons to compute similarity. Here, we use a chemical language model to create a vector-based chemical search. We extend previous implementations by creating a prompt engineering strategy that utilizes two different chemical string representation algorithms: one for the query and the other for the database. We explore this method by reviewing search results from nine queries with diverse targets. We find that the method identifies molecules with similar patent-derived functionality to the query, as determined by our validated LLM-assisted patent summarization pipeline. Further, many of these functionally similar molecules have different structures and scaffolds from the query, making them unlikely to be found with traditional chemical similarity searches. This method may serve as a new tool for the discovery of novel molecular structural classes that achieve target functionality.
Collapse
Affiliation(s)
- Clayton W. Kosonocky
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78705, USA
| | - Aaron L. Feller
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78705, USA
| | - Claus O. Wilke
- Department of Integrative Biology, University of Texas at Austin, Austin, TX 78705, USA
| | - Andrew D. Ellington
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78705, USA
- Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX 78705, USA
| |
Collapse
|
9
|
Gaikwad SS, Nimal SK, Pol R, Markad D, Jadhao AR, Jadhav U, Kate AN, Gacche RN, Patil LR, Chikate RC. Targeting AKT2 in MDA-MB-231 Cells by Pyrazole Hybrids: Structural, Biological and Molecular Docking Studies. Chem Biodivers 2023; 20:e202300799. [PMID: 37702285 DOI: 10.1002/cbdv.202300799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 09/14/2023]
Abstract
Pyrazolic hybrids appended with naphthalene, p-chlorobenzene, o-phenol and toluene have been synthesized using Claisen Schmidt condensation reaction of 1-benzyl-3,5-dimethyl-1H-pyrazole-4-carbaldehyde. All compounds were characterized by various spectroscopic techniques. Compound (E)-3-(1-benzyl-3,5-dimethyl-1H-pyrazol-4-yl)-1-(4-chlorophenyl)prop-2-en-1-one crystallizes in monoclinic crystal system with C2/c space group. These synthesized compounds were tested for cytotoxic activity and among these compounds 4b and 5a shows prominent cytotoxic activity against triple-negative breast cancer (TNBC) cells MDA-MB-231 with IC50 values 47.72 μM and 24.25 μM, respectively. Distinguishing morphological changes were noticed in MDA-MB-231 cells treated with pyrazole hybrids contributing to apoptosis action. To get more insight into cytotoxic activity, in silico molecular docking of these compounds were performed and the results suggested that (E)-3-(1-benzyl-3,5-dimethyl-1H-pyrazol-4-yl)-1-(p-tolyl)prop-2-en-1-one and 1-(1'-benzyl-5-(4-chlorophenyl)-3',5'-dimethyl-3,4-dihydro-1'H,2H-[3,4'-bipyrazol]-2-yl)ethan-1-one binds to the prominent domain of Akt2 indicating their potential ability as Akt2 inhibitor. Moreover, from in silico ADME studies clearly demonstrated that these compounds may be regarded as a drug candidate for sub-lingual absorption based on log p values (2.157-4.924). These compounds also show promising antitubercular activity. The overall results suggest that pyrazolic hybrids with substitution at less sterically hindered positions have appealing potent cytotoxic activity and antituberculosis activity due to which they may act as multidrug candidate.
Collapse
Affiliation(s)
- Sanjay S Gaikwad
- Department of Chemistry, MES, Abasaheb Garware College, Pune, Maharashtra, India-, 411004
| | - Snehal K Nimal
- Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra, India-, 411007
| | - Rushikesh Pol
- Department of Microbiology, Savitribai Phule Pune University, Pune, Maharashtra, India-, 411007
| | - Datta Markad
- Department of Chemistry, University of Liverpool, Liverpool, L69 7ZD, United Kingdom
| | - Amardeep R Jadhao
- Department of Chemistry, Late Pushpadevi Patil Arts and Science College, Risod, Dist., Washim, Maharashtra, India-, 444506
| | - Umesh Jadhav
- Department of Microbiology, Savitribai Phule Pune University, Pune, Maharashtra, India-, 411007
| | - Anup N Kate
- Department of Chemistry, MES, Abasaheb Garware College, Pune, Maharashtra, India-, 411004
| | - Rajesh N Gacche
- Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra, India-, 411007
| | - Limbraj R Patil
- Department of Chemistry, Maharaja Jivajirao Shinde Arts, Science, Commerce, College, Shrigonda, Maharashtra, India-, 413701
| | - Rajeev C Chikate
- Department of Chemistry, MES, Abasaheb Garware College, Pune, Maharashtra, India-, 411004
| |
Collapse
|
10
|
Peytam F, Emamgholipour Z, Mousavi A, Moradi M, Foroumadi R, Firoozpour L, Divsalar F, Safavi M, Foroumadi A. Imidazopyridine-based kinase inhibitors as potential anticancer agents: A review. Bioorg Chem 2023; 140:106831. [PMID: 37683538 DOI: 10.1016/j.bioorg.2023.106831] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/16/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023]
Abstract
Considering the fundamental role of protein kinases in the mechanism of protein phosphorylation in critical cellular processes, their dysregulation, especially in cancers, has underscored their therapeutic relevance. Imidazopyridines represent versatile scaffolds found in abundant bioactive compounds. Given their structural features, imidazopyridines have possessed pivotal potency to interact with different protein kinases, inspiring researchers to carry out numerous structural variations. In this comprehensive review, we encompass an extensive survey of the design and biological evaluations of imidazopyridine-based small molecules as potential agents targeting diverse kinases for anticancer applications. We describe the structural elements critical to inhibitory potency, elucidating their key structure-activity relationships (SAR) and mode of actions, where available. We classify these compounds into two groups: Serine/threonine and Tyrosine inhibitors. By highlighting the promising role of imidazopyridines in kinase inhibition, we aim to facilitate the design and development of more effective, targeted compounds for cancer treatment.
Collapse
Affiliation(s)
- Fariba Peytam
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Emamgholipour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Mousavi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahfam Moradi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Roham Foroumadi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Loghman Firoozpour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Divsalar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Maliheh Safavi
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran, Iran
| | - Alireza Foroumadi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran; Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Chapdelaine AG, Sun G. Challenges and Opportunities in Developing Targeted Therapies for Triple Negative Breast Cancer. Biomolecules 2023; 13:1207. [PMID: 37627272 PMCID: PMC10452226 DOI: 10.3390/biom13081207] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/26/2023] [Accepted: 07/29/2023] [Indexed: 08/27/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a heterogeneous group of breast cancers characterized by their lack of estrogen receptors, progesterone receptors, and the HER2 receptor. They are more aggressive than other breast cancer subtypes, with a higher mean tumor size, higher tumor grade, the worst five-year overall survival, and the highest rates of recurrence and metastasis. Developing targeted therapies for TNBC has been a major challenge due to its heterogeneity, and its treatment still largely relies on surgery, radiation therapy, and chemotherapy. In this review article, we review the efforts in developing targeted therapies for TNBC, discuss insights gained from these efforts, and highlight potential opportunities going forward. Accumulating evidence supports TNBCs as multi-driver cancers, in which multiple oncogenic drivers promote cell proliferation and survival. In such multi-driver cancers, targeted therapies would require drug combinations that simultaneously block multiple oncogenic drivers. A strategy designed to generate mechanism-based combination targeted therapies for TNBC is discussed.
Collapse
Affiliation(s)
| | - Gongqin Sun
- Department of Cell and Molecular Biology, University of Rhode Island, Kingston, RI 02881, USA;
| |
Collapse
|
12
|
Kciuk M, Marciniak B, Celik I, Zerroug E, Dubey A, Sundaraj R, Mujwar S, Bukowski K, Mojzych M, Kontek R. Pyrazolo[4,3- e]tetrazolo[1,5- b][1,2,4]triazine Sulfonamides as an Important Scaffold for Anticancer Drug Discovery-In Vitro and In Silico Evaluation. Int J Mol Sci 2023; 24:10959. [PMID: 37446136 DOI: 10.3390/ijms241310959] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Pyrazolo[4,3-e]tetrazolo[1,5-b][1,2,4]triazine sulfonamides (MM-compounds) are a relatively new class of heterocyclic compounds that exhibit a wide variety of biological actions, including anticancer properties. Here, we used caspase enzyme activity assays, flow cytometry analysis of propidium iodide (PI)-stained cells, and a DNA laddering assay to investigate the mechanisms of cell death triggered by the MM-compounds (MM134, -6, -7, and -9). Due to inconsistent results in caspase activity assays, we have performed a bromodeoxyuridine (BrdU) incorporation assay, colony formation assay, and gene expression profiling. The compounds' cytotoxic and pro-oxidative properties were also assessed. Additionally, computational studies were performed to demonstrate the potential of the scaffold for future drug discovery endeavors. MM-compounds exhibited strong micromolar (0.06-0.35 µM) anti-proliferative and pro-oxidative activity in two cancer cell lines (BxPC-3 and PC-3). Activation of caspase 3/7 was observed following a 24-h treatment of BxPC-3 cells with IC50 concentrations of MM134, -6, and -9 compounds. However, no DNA fragmentation characteristics for apoptosis were observed in the flow cytometry and DNA laddering analysis. Gene expression data indicated up-regulation of BCL10, GADD45A, RIPK2, TNF, TNFRSF10B, and TNFRSF1A (TNF-R1) following treatment of cells with the MM134 compound. Moreover, in silico studies indicated AKT2 kinase as the primary target of compounds. MM-compounds exhibit strong cytotoxic activity with pro-oxidative, pro-apoptotic, and possibly pro-necroptotic properties that could be employed for further drug discovery approaches.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland
| | - Beata Marciniak
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland
| | - Ismail Celik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Kayseri 38280, Turkey
| | - Enfale Zerroug
- Group of Computational and Pharmaceutical Chemistry, LMCE Laboratory, University of Biskra, BP 145, Biskra 07000, Algeria
| | - Amit Dubey
- Computational Chemistry and Drug Discovery Division, Quanta Calculus, Greater Noida 274203, Uttar Prades, India
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India
| | - Rajamanikandan Sundaraj
- Centre for Drug Discovery, Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
| | - Somdutt Mujwar
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - Karol Bukowski
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland
| | - Mariusz Mojzych
- Department of Chemistry, Siedlce University of Natural Sciences and Humanities, 3 Maja 54, 08-110 Siedlce, Poland
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland
| |
Collapse
|
13
|
Occhiuzzi MA, Lico G, Ioele G, De Luca M, Garofalo A, Grande F. Recent advances in PI3K/PKB/mTOR inhibitors as new anticancer agents. Eur J Med Chem 2023; 246:114971. [PMID: 36462440 DOI: 10.1016/j.ejmech.2022.114971] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022]
Abstract
The biochemical role of the PI3K/PKB/mTOR signalling pathway in cell-cycle regulation is now well known. During the onset and development of different forms of cancer it becomes overactive reducing apoptosis and allowing cell proliferation. Therefore, this pathway has become an important target for the treatment of various forms of malignant tumors, including breast cancer and follicular lymphoma. Recently, several more or less selective inhibitors targeting these proteins have been identified. In general, drugs that act on multiple targets within the entire pathway are more efficient than single targeting inhibitors. Multiple inhibitors exhibit high potency and limited drug resistance, resulting in promising anticancer agents. In this context, the present survey focuses on small molecule drugs capable of modulating the PI3K/PKB/mTOR signalling pathway, thus representing drugs or drug candidates to be used in the pharmacological treatment of different forms of cancer.
Collapse
Affiliation(s)
| | - Gernando Lico
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Giuseppina Ioele
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Michele De Luca
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Antonio Garofalo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Fedora Grande
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| |
Collapse
|
14
|
Sain A, Kandasamy T, Naskar D. In silico approach to target PI3K/Akt/mTOR axis by selected Olea europaea phenols in PIK3CA mutant colorectal cancer. J Biomol Struct Dyn 2022; 40:10962-10977. [PMID: 34296655 DOI: 10.1080/07391102.2021.1953603] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Worldwide disease burden of colorectal cancer (CRC) increasing alarmingly, but a suitable therapeutic strategy is not available yet. Abnormal activation of the PI3K/Akt/mTOR signalling because of mutation in the PIK3CA gene is a driving force behind CRC development. Therefore, this study aimed to comprehensively characterise the potential of phenolic compounds from Olea europaea against the PI3K/Akt/mTOR axis by using in silico methodologies. Molecular docking was utilised to study key interactions between phenolic compounds of O. europaea and target proteins PI3K, Akt, mTOR with reference to known inhibitor of target. Drug likeness and ADME/T properties of selected phenols were explored by online tools. Dynamic properties and binding free energy of target-ligand interactions were studied by molecular dynamic simulation and MM-PBSA method respectively. Molecular docking revealed apigenin, luteolin, pinoresinol, oleuropein, and oleuropein aglycone as the top five phenolic compounds which showed comparable/better binding affinity than the known inhibitor of the respective target protein. Drug likeness and ADME/T properties were employed to select the top three phenols namely, apigenin, luteolin, and pinoresinol which shown to bind stably to the catalytic cleft of target proteins as confirmed by molecular dynamics simulations. Therefore, Apigenin, luteolin, and pinoresinol have the potential to be used as the non-toxic alternative to synthetic chemical inhibitors generally used in CRC treatment as they can target PI3K/Akt/mTOR axis. Particularly, pinoresinol showed great potential as dual PI3K/mTOR inhibitor. However, this study needs to be complemented with future in vitro and in vivo studies to provide an alternative way of CRC treatment. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Arindam Sain
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Nadia, West Bengal, India
| | - Thirukumaran Kandasamy
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Debdut Naskar
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Nadia, West Bengal, India
| |
Collapse
|
15
|
Yu X, Xu J, Cahuzac KM, Xie L, Shen Y, Chen X, Liu J, Parsons RE, Jin J. Novel Allosteric Inhibitor-Derived AKT Proteolysis Targeting Chimeras (PROTACs) Enable Potent and Selective AKT Degradation in KRAS/BRAF Mutant Cells. J Med Chem 2022; 65:14237-14260. [PMID: 36197750 PMCID: PMC9613624 DOI: 10.1021/acs.jmedchem.2c01454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AKT is an important target for cancer therapeutics. Significant advancements have been made in developing ATP-competitive and allosteric AKT inhibitors. Recently, several AKT proteolysis targeting chimeras (PROTACs) derived from ATP-competitive AKT inhibitors have been reported, including MS21. While MS21 potently degraded AKT and inhibited the growth in tumor cells harboring PI3K/PTEN pathway mutation, it was largely ineffective in degrading AKT in KRAS/BRAF mutated cells as a single agent. To overcome the AKT degradation resistance in KRAS/BRAF mutated cells, we developed novel AKT PROTACs derived from an AKT allosteric inhibitor, including degrader 62 (MS15). 62 displayed potent and selective AKT degradation activity and potent antiproliferative activity in KRAS/BRAF mutated cancer cells, in addition to PI3K/PTEN mutated cancer cells. Furthermore, 62 was bioavailable in mice through intraperitoneal administration. Overall, 62 is a valuable chemical tool to degrade AKT in cells harboring KRAS/BRAF mutation and expands the tool box for pharmacologically modulating AKT.
Collapse
Affiliation(s)
- Xufen Yu
- Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Jia Xu
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Kaitlyn M. Cahuzac
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Ling Xie
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, United States
| | - Yudao Shen
- Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Xian Chen
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, United States
| | - Jing Liu
- Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Ramon E. Parsons
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
16
|
Toson B, Fortes IS, Roesler R, Andrade SF. Targeting Akt/PKB in pediatric tumors: A review from preclinical to clinical trials. Pharmacol Res 2022; 183:106403. [PMID: 35987481 DOI: 10.1016/j.phrs.2022.106403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/01/2022] [Accepted: 08/15/2022] [Indexed: 11/25/2022]
Abstract
The serine/threonine kinase Akt is a major player in the phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway, and its modulation impacts multiple cellular processes such as growth, proliferation, and survival. Several abnormalities in this pathway have been documented over the years, and these alterations were shown to have great implications in tumorigenesis and resistance to chemotherapy. Thus, multiple Akt inhibitors have been developed and tested in adult tumors, and some of them are currently undergoing phase I, II, and III clinical trials for distinct cancers that arise during adulthood. Despite that, the impact of these inhibitors is still not fully understood in pediatric tumors, and Akt-specific targeting seems to be a promising approach to treat children affected by cancers. This review summarizes recent available evidence of Akt inhibitors in pediatric cancers, from both preclinical and clinical studies. In short, we demonstrate the impact that Akt inhibition provides in tumorigenesis, and we suggest targeting the PI3K/Akt/mTOR signaling pathway, alone or in combination with other inhibitors, is a feasible tool to achieve better outcomes in pediatric tumors.
Collapse
Affiliation(s)
- Bruno Toson
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Isadora S Fortes
- Pharmaceutical Synthesis Group (PHARSG), College of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Pharmaceutical Sciences Graduate Program, Federal University of Rio Grande do Sul (UFRGS), Av. Ipiranga, 2752, Porto Alegre, RS 90610-000, Brazil
| | - Rafael Roesler
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Department of Pharmacology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Saulo F Andrade
- Pharmaceutical Synthesis Group (PHARSG), College of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Pharmaceutical Sciences Graduate Program, Federal University of Rio Grande do Sul (UFRGS), Av. Ipiranga, 2752, Porto Alegre, RS 90610-000, Brazil.
| |
Collapse
|
17
|
Sinha K, Kumar S, Rawat B, Singh R, Purohit R, Kumar D, Padwad Y. Kutkin, iridoid glycosides enriched fraction of Picrorrhiza kurroa promotes insulin sensitivity and enhances glucose uptake by activating PI3K/Akt signaling in 3T3-L1 adipocytes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 103:154204. [PMID: 35671635 DOI: 10.1016/j.phymed.2022.154204] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/21/2022] [Accepted: 05/24/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Therapeutic failure and drug resistance are common sequelae to insulin resistance associated with type 2 diabetes mellitus (T2DM). Consequently, there is an unmet need of alternative strategies to overcome insulin resistance associated complications. PURPOSE To demonstrate whether Kutkin (KT), iridoid glycoside enriched fraction of Picrorhiza kurroa extract (PKE) has potential to increase the insulin sensitivity vis à vis glucose uptake in differentiated adipocytes. METHODS Molecular interaction of KT phytoconstituents, picroside-I (P-I) & picroside- II (P-II) with peroxisome proliferator-activated receptor gamma (PPARγ), phosphatidylinositol 3-kinase (PI3K) and protein kinase B (Akt) were analyzed in silico. Cellular viability and adipogenesis were determined by following 3-(4, 5-Dimethylthiazol-2-Yl)-2, 5-Diphenyltetrazolium bromide (MTT) assay and Oil Red-O staining. Further, ELISA kit based triglycerides and diacylglycerol-O-Acyltransferase-1 (DGAT1) were assessed in differentiated adipocytes. ELISA based determination were performed to check the levels of adiponectin and tumor necrosis factor alpha (TNF-α). However, Flow cytometry and immunofluorescence based assays were employed to measure the glucose uptake and glucose transporter 4 (glut4) expression in differentiated adipocytes, respectively. Further to explore the targeted signaling axis, mRNA expression levels of PPARγ, CCAAT/enhancer binding protein α (CEBPα), and glut4 were determined using qRT-PCR and insulin receptor substrate-1 (IRS-1), Insulin receptor substrate-2 (IRS-2), PI3K/Akt, AS160, glut4 followed by protein validation using immunoblotting in differentiated adipocytes. RESULTS In silico analysis revealed the binding affinities of major constituents of KT (P-I& P-II) with PPARγ/PI3K/Akt. The enhanced intracellular accumulation of triglycerides with concomitant activation of PPARγ and C/EBPα in KT treated differentiated adipocytes indicates augmentation of adipogenesis in a concentration-dependent manner. Additionally, at cellular level, KT upregulated the expression of DAGT1, and decreases fatty acid synthase (FAS), and lipoprotein lipase (LPL), further affirmed improvement in lipid milieu. It was also observed that KT upregulated the levels of adiponectin and reduced TNFα expression, thus improving the secretory functions of adipocytes along with enhanced insulin sensitivity. Furthermore, KT significantly promoted insulin mediated glucose uptake by increasing glut4 translocation to the membrane via PI3/Akt signaling cascade. The results were further validated using PI3K specific inhibitor, wortmannin and findings revealed that KT treatment significantly enhanced the expression and activation of p-PI3K/PI3K and p-Akt/Akt even in case of treatment with PI3K inhibitor wortmannin alone and co-treatment with KT in differentiated adipocytes and affirmed that KT as activator of PI3K/Akt axis in the presence of inhibitor as well. CONCLUSION Collectively, KT fraction of PKE showed anti-diabetic effects by enhancing glucose uptake in differentiated adipocytes via activation of PI3K/Akt signaling cascade. Therefore, KT may be used as a promising novel natural therapeutic agent for managing T2DMand to the best of our knowledge, this is the first report, showing the efficacy and potential molecular mechanism of KT in enhancing insulin sensitivity and glucose uptake in differentiated adipocytes.
Collapse
Affiliation(s)
- Kajal Sinha
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061 H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Shiv Kumar
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061 H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Bindu Rawat
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061 HP., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Rahul Singh
- Structural Bioinformatics Lab, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061 H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Rituraj Purohit
- Structural Bioinformatics Lab, Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061 H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Dinesh Kumar
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061 HP., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Yogendra Padwad
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061 H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
18
|
Preparation of Novel Pyrazolo[4,3- e]tetrazolo[1,5- b][1,2,4]triazine Sulfonamides and Their Experimental and Computational Biological Studies. Int J Mol Sci 2022; 23:ijms23115892. [PMID: 35682571 PMCID: PMC9180621 DOI: 10.3390/ijms23115892] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023] Open
Abstract
Pyrazolo[4,3-e]tetrazolo[1,5-b][1,2,4]triazine sulfonamides constitute a novel class of heterocyclic compounds with broad biological activity, including anticancer properties. Investigated in this study, MM-compounds (MM134, MM136, MM137, and MM139) exhibited cytotoxic and proapoptotic activity against cancer cell lines (BxPC-3, PC-3, and HCT-116) in nanomolar concentrations without causing cytotoxicity in normal cells (L929 and WI38). In silico predictions indicate that tested compounds exhibit favorable pharmacokinetic profiles and may exert anticancer activity through the inhibition of BTK kinase, the AKT-mTOR pathway and PD1-PD-L1 interaction. Our findings point out that these sulfonamide derivatives may constitute a source of new anticancer drugs after optimization.
Collapse
|
19
|
Huang W, Li G, He XH, Li HP, Zhao Q, Li DA, Zhu HP, Zhang YH, Zhan G. Design, synthesis, and biological evaluation of tetrahydro-αcarbolines as Akt1 inhibitors that inhibit colorectal cancer cells proliferation. ChemMedChem 2022; 17:e202200104. [PMID: 35355421 DOI: 10.1002/cmdc.202200104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/25/2022] [Indexed: 11/09/2022]
Abstract
A series of densely functionalized THαCs were designed and synthesized as Akt1 inhibitors. Organocatalytic [3+3] annulation between indolin-2-imines 1 and nitroallylic acetates 2 provided rapid access to this pharmacologically interesting framework. In vitro kinase inhibitory abilities and cytotoxicity assays revealed that compound 3af was the most potent Akt1 inhibitor, and mechanistic study indicated that compound 3af suppressed the proliferation of colorectal cancer cells via inducing apoptosis and autophagy. Molecular docking suggested that the indole fragment of 3af was inserted into the hydrophobic pocket of Akt1 protein, and the H-bond between 3af and residue Lys179 also contributed to the stable binding. This article provides an efficient strategy to design and synthesize biologically important compounds as novel Akt1 inhibitors.
Collapse
Affiliation(s)
- Wei Huang
- Chengdu University of Traditional Chinese Medicine, School of Pharmacy, 1166 Liu Tai Av., 610000, Chengdu, CHINA
| | - Guo Li
- Chengdu University of Traditional Chinese Medicine Wenjiang Campus: Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, CHINA
| | - Xiang-Hong He
- Chengdu University of Traditional Chinese Medicine Wenjiang Campus: Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, CHINA
| | - He-Ping Li
- Chengdu University of Traditional Chinese Medicine Wenjiang Campus: Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, CHINA
| | - Qian Zhao
- Chengdu University of Traditional Chinese Medicine Wenjiang Campus: Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, 610000, Chengdu, CHINA
| | - Dong-Ai Li
- Chengdu University of Traditional Chinese Medicine Wenjiang Campus: Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, CHINA
| | - Hong-Ping Zhu
- Chengdu University of Traditional Chinese Medicine Wenjiang Campus: Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, CHINA
| | - Yue-Hua Zhang
- Sichuan University, State Key Laboratory of Biotherapy and Department of Pharmacy, CHINA
| | - Gu Zhan
- Chengdu University of Traditional Chinese Medicine Wenjiang Campus: Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, CHINA
| |
Collapse
|
20
|
Shanak S, Bassalat N, Barghash A, Kadan S, Ardah M, Zaid H. Drug Discovery of Plausible Lead Natural Compounds That Target the Insulin Signaling Pathway: Bioinformatics Approaches. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2832889. [PMID: 35356248 PMCID: PMC8958086 DOI: 10.1155/2022/2832889] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/16/2022] [Accepted: 02/09/2022] [Indexed: 12/11/2022]
Abstract
The growing smooth talk in the field of natural compounds is due to the ancient and current interest in herbal medicine and their potentially positive effects on health. Dozens of antidiabetic natural compounds were reported and tested in vivo, in silico, and in vitro. The role of these natural compounds, their actions on the insulin signaling pathway, and the stimulation of the glucose transporter-4 (GLUT4) insulin-responsive translocation to the plasma membrane (PM) are all crucial in the treatment of diabetes and insulin resistance. In this review, we collected and summarized a group of available in vivo and in vitro studies which targeted isolated phytochemicals with possible antidiabetic activity. Moreover, the in silico docking of natural compounds with some of the insulin signaling cascade key proteins is also summarized based on the current literature. In this review, hundreds of recent studies on pure natural compounds that alleviate type II diabetes mellitus (type II DM) were revised. We focused on natural compounds that could potentially regulate blood glucose and stimulate GLUT4 translocation through the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway. On attempt to point out potential new natural antidiabetic compounds, this review also focuses on natural ingredients that were shown to interact with proteins in the insulin signaling pathway in silico, regardless of their in vitro/in vivo antidiabetic activity. We invite interested researchers to test these compounds as potential novel type II DM drugs and explore their therapeutic mechanisms.
Collapse
Affiliation(s)
- Siba Shanak
- Faculty of Sciences, Arab American University, P.O Box 240, Jenin, State of Palestine
| | - Najlaa Bassalat
- Faculty of Sciences, Arab American University, P.O Box 240, Jenin, State of Palestine
- Faculty of Medicine, Arab American University, P.O Box 240, Jenin, State of Palestine
| | - Ahmad Barghash
- Computer Science Department, German Jordanian University, Madaba Street. P.O. Box 35247, Amman 11180, Jordan
| | - Sleman Kadan
- Qasemi Research Center, Al-Qasemi Academic College, P.O Box 124, Baqa El-Gharbia 30100, Israel
| | - Mahmoud Ardah
- Faculty of Sciences, Arab American University, P.O Box 240, Jenin, State of Palestine
| | - Hilal Zaid
- Faculty of Medicine, Arab American University, P.O Box 240, Jenin, State of Palestine
- Qasemi Research Center, Al-Qasemi Academic College, P.O Box 124, Baqa El-Gharbia 30100, Israel
| |
Collapse
|
21
|
Yu X, Xu J, Shen Y, Cahuzac KM, Park KS, Dale B, Liu J, Parsons RE, Jin J. Discovery of Potent, Selective, and In Vivo Efficacious AKT Kinase Protein Degraders via Structure-Activity Relationship Studies. J Med Chem 2022; 65:3644-3666. [PMID: 35119851 PMCID: PMC8900464 DOI: 10.1021/acs.jmedchem.1c02165] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We recently reported a potent, selective, and in vivo efficacious AKT degrader, MS21, which is a von Hippel-Lindau (VHL)-recruiting proteolysis targeting chimera (PROTAC) based on the AKT inhibitor AZD5363. However, no structure-activity relationship (SAR) studies that resulted in this discovery have been reported. Herein, we present our SAR studies that led to the discovery of MS21, another VHL-recruiting AKT degrader, MS143 (compound 20) with similar potency as MS21, and a novel cereblon (CRBN)-recruiting PROTAC, MS5033 (compound 35). Compounds 20 and 35 induced rapid and robust AKT degradation in a concentration- and time-dependent manner via hijacking the ubiquitin-proteasome system. Compound 20 suppressed cell growth more effectively than AZD5363 in multiple cancer cell lines. Furthermore, 20 and 35 displayed good plasma exposure levels in mice and are suitable for in vivo efficacy studies. Lastly, compound 20 effectively suppressed tumor growth in vivo in a xenograft model without apparent toxicity.
Collapse
Affiliation(s)
| | | | - Yudao Shen
- Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences and Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Kaitlyn M. Cahuzac
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Kwang-su Park
- Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences and Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Brandon Dale
- Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences and Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Jing Liu
- Corresponding Authors: Jing Liu – Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences and Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States; , Ramon E. Parsons – Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States; , Jian Jin – Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences and Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States;
| | - Ramon E. Parsons
- Corresponding Authors: Jing Liu – Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences and Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States; , Ramon E. Parsons – Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States; , Jian Jin – Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences and Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States;
| | - Jian Jin
- Corresponding Authors: Jing Liu – Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences and Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States; , Ramon E. Parsons – Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States; , Jian Jin – Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences and Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States;
| |
Collapse
|
22
|
Yu X, Xu J, Xie L, Wang L, Shen Y, Cahuzac KM, Chen X, Liu J, Parsons RE, Jin J. Design, Synthesis, and Evaluation of Potent, Selective, and Bioavailable AKT Kinase Degraders. J Med Chem 2021; 64:18054-18081. [PMID: 34855399 PMCID: PMC8819633 DOI: 10.1021/acs.jmedchem.1c01476] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The serine/threonine kinase AKT functions as a critical node of the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (m-TOR) signaling pathway. Aberrant activation and overexpression of AKT are strongly correlated with numerous human cancers. To date, only two AKT degraders with no structure-activity relationship (SAR) results have been reported. Through extensive SAR studies on various linkers, E3 ligase ligands, and AKT binding moieties, we identified two novel and potent AKT proteolysis targeting chimera (PROTAC) degraders: von Hippel-Lindau (VHL)-recruiting degrader 13 (MS98) and cereblon (CRBN)-recruiting degrader 25 (MS170). These two compounds selectively induced robust AKT protein degradation, inhibited downstream signaling, and suppressed cancer cell proliferation. Moreover, these two degraders exhibited good plasma exposure levels in mice through intraperitoneal injection. Overall, our comprehensive SAR studies led to the discovery of degraders 13 and 25, which are potentially useful chemical tools to investigate biological and pathogenic functions of AKT in vitro and in vivo.
Collapse
Affiliation(s)
| | | | - Ling Xie
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Li Wang
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Yudao Shen
- Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States; Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Kaitlyn M. Cahuzac
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Xian Chen
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Jing Liu
- Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States; Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Ramon E. Parsons
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States; Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
23
|
Li HL, Cheng Y, Zhou ZW, Long HZ, Luo HY, Wen DD, Cheng L, Gao LC. Isoliensinine induces cervical cancer cell cycle arrest and apoptosis by inhibiting the AKT/GSK3α pathway. Oncol Lett 2021; 23:8. [PMID: 34820007 PMCID: PMC8607237 DOI: 10.3892/ol.2021.13126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/20/2021] [Indexed: 12/26/2022] Open
Abstract
Isoliensinine is a bis-benzylisoquinoline alkaloid that can be isolated from the lotus Nelumbo nucifera Gaertn. It has been reported to exert a variety of anti-cancer properties. In the present study, the potential effects of isoliensinine on cervical cancer Siha, HeLa, Caski and C33A cell lines were investigated by using Cell Counting Kit-8 (CCK-8), flow cytometry, western blotting and reverse transcription-PCR (RT-PCR) to measure cell proliferation, the cell cycle and apoptosis, in addition to elucidating the underlying molecular mechanism. Protein levels of p21, CDK2, Cyclin E, Mcl-1, cleaved Caspase-9, AKT, phosphorylated-AKT, glycogen synthase kinase (Gsk)3α, PTEN, and mRNA levels of p21, p15, p27, CDK2, CDK4, Cyclin E, Cyclin D, Gsk3α, Gsk3β and PTEN were measured. Molecular docking assays were used to calculate the strength of binding of isoliensinine to AKT using AutoDock 4.0. Isoliensinine was found to induce cell cycle arrest at the G0/G1 phase by upregulating p21 expression and downregulating CDK2 and cyclin E in cervical cancer cells. In addition, in previous research, isoliensinine promoted cell apoptosis by downregulating myeloid-cell leukemia 1 expression and activating caspase-9. Upstream, isoliensinine significantly downregulated AKT (S473) phosphorylation and GSK3α expression in a dose- and time-dependent manner. The AKT inhibitor AKTi-1/2 enhanced the function of isoliensinine on cell cycle arrest and apoptosis through the AKT/GSK3α pathway. AutoDock analysis showed that isoliensinine can bind to the AKT protein. These findings suggest that isoliensinine can induce cervical cancer cell cycle arrest and apoptosis by inhibiting the AKT/GSK3α pathway, which represents a novel strategy for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Hong-Li Li
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, Hunan 410000, P.R. China.,School of Life Science, Hunan University of Science and Technology, Xiangtan, Hunan 411201, P.R. China
| | - Yan Cheng
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, Hunan 410000, P.R. China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research Affiliated to School of Pharmacy, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zi-Wei Zhou
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, Hunan 410000, P.R. China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research Affiliated to School of Pharmacy, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hui-Zhi Long
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, Hunan 410000, P.R. China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research Affiliated to School of Pharmacy, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hong-Yu Luo
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, Hunan 410000, P.R. China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research Affiliated to School of Pharmacy, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Dan-Dan Wen
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, Hunan 410000, P.R. China
| | - Lin Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Li-Chen Gao
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, Hunan 410000, P.R. China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research Affiliated to School of Pharmacy, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
24
|
Irie T, Asami T, Sawa A, Uno Y, Taniyama C, Funakoshi Y, Masai H, Sawa M. Discovery of AS-0141, a Potent and Selective Inhibitor of CDC7 Kinase for the Treatment of Solid Cancers. J Med Chem 2021; 64:14153-14164. [PMID: 34607435 DOI: 10.1021/acs.jmedchem.1c01319] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
CDC7, a serine-threonine kinase, plays conserved and important roles in regulation of DNA replication and has been recognized as a potential anticancer target. We report here the optimization of a series of furanone analogues starting from compound 1 with a focus on ADME properties suitable for clinical development. By replacing the 2-chlorobenzene moiety in 1 with various aliphatic groups, we identified compound 24 as a potent CDC7 inhibitor with excellent kinase selectivity and favorable oral bioavailability in multiple species. Oral administration of 24 demonstrated robust in vivo antitumor efficacy in a colorectal cancer xenograft model. Compound 24 (AS-0141) is currently in phase I clinical trials for the treatment of solid cancers.
Collapse
Affiliation(s)
- Takayuki Irie
- Research and Development, Carna Biosciences, Inc., 3F BMA, 1-5-5 minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Tokiko Asami
- Research and Development, Carna Biosciences, Inc., 3F BMA, 1-5-5 minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Ayako Sawa
- Research and Development, Carna Biosciences, Inc., 3F BMA, 1-5-5 minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Yuko Uno
- Research and Development, Carna Biosciences, Inc., 3F BMA, 1-5-5 minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Chika Taniyama
- Ginkgo Biomedical Research Institute, Research and Development Department, SBI Biotech Co., Ltd., Izumi Garden Tower 15F, 1-6- Roppongi, Minato-ku, Tokyo 106-6015, Japan
| | - Yoko Funakoshi
- Ginkgo Biomedical Research Institute, Research and Development Department, SBI Biotech Co., Ltd., Izumi Garden Tower 15F, 1-6- Roppongi, Minato-ku, Tokyo 106-6015, Japan
| | - Hisao Masai
- Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Masaaki Sawa
- Research and Development, Carna Biosciences, Inc., 3F BMA, 1-5-5 minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| |
Collapse
|
25
|
Junaid M, Akter Y, Afrose SS, Tania M, Khan MA. Biological Role of AKT and Regulation of AKT Signaling Pathway by Thymoquinone: Perspectives in Cancer Therapeutics. Mini Rev Med Chem 2021; 21:288-301. [PMID: 33019927 DOI: 10.2174/1389557520666201005143818] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 09/02/2020] [Accepted: 09/08/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND AKT/PKB is an important enzyme with numerous biological functions, and its overexpression is related to carcinogenesis. AKT stimulates different signaling pathways that are downstream of activated tyrosine kinases and phosphatidylinositol 3-kinase, hence functions as an important target for anti-cancer drugs. OBJECTIVE In this review article, we have interpreted the role of AKT signaling pathway in cancer and the natural inhibitory effect of Thymoquinone (TQ) in AKT and its possible mechanisms. METHOD We have collected the updated information and data on AKT, its role in cancer and the inhibitory effect of TQ in AKT signaling pathway from Google Scholar, PubMed, Web of Science, Elsevier, Scopus, and many more. RESULTS Many drugs are already developed, which can target AKT, but very few among them have passed clinical trials. TQ is a natural compound, mainly found in black cumin, which has been found to have potential anti-cancer activities. TQ targets numerous signaling pathways, including AKT, in different cancers. In fact, many studies revealed that AKT is one of the major targets of TQ. The preclinical success of TQ suggests its clinical studies on cancer. CONCLUSION This review article summarizes the role of AKT in carcinogenesis, its potent inhibitors in clinical trials, and how TQ acts as an inhibitor of AKT and TQ's future as a cancer therapeutic drug.
Collapse
Affiliation(s)
- Md Junaid
- Molecular Modeling Drug-design and Discovery Laboratory, Pharmacology Research Division, Bangladesh Council of Scientific and Industrial Research, Chattogram, Bangladesh
| | - Yeasmin Akter
- Department of Biotechnology and Genetic Engineering, Noakhali Science & Technology University, Noakhali, Bangladesh
| | | | - Mousumi Tania
- Division of Molecular Cancer, Red Green Research Center, Dhaka, Bangladesh
| | - Md Asaduzzaman Khan
- The research center for preclinical medicine, Southwest Medical University, Luzhou, China
| |
Collapse
|
26
|
Wiechmann S, Ruprecht B, Siekmann T, Zheng R, Frejno M, Kunold E, Bajaj T, Zolg DP, Sieber SA, Gassen NC, Kuster B. Chemical Phosphoproteomics Sheds New Light on the Targets and Modes of Action of AKT Inhibitors. ACS Chem Biol 2021; 16:631-641. [PMID: 33755436 DOI: 10.1021/acschembio.0c00872] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Due to its important roles in oncogenic signaling, AKT has been subjected to extensive drug discovery efforts leading to small molecule inhibitors investigated in advanced clinical trials. To better understand how these drugs exert their therapeutic effects at the molecular level, we combined chemoproteomic target affinity profiling using kinobeads and phosphoproteomics to analyze the five clinical AKT inhibitors AZD5363 (Capivasertib), GSK2110183 (Afuresertib), GSK690693, Ipatasertib, and MK-2206 in BT-474 breast cancer cells. Kinobead profiling identified between four and 29 nM targets for these compounds and showed that AKT1 and AKT2 were the only common targets. Similarly, measuring the response of the phosphoproteome to the same inhibitors identified ∼1700 regulated phosphorylation sites, 276 of which were perturbed by all five compounds. This analysis expanded the known AKT signaling network by 119 phosphoproteins that may represent direct or indirect targets of AKT. Within this new network, 41 regulated phosphorylation sites harbor the AKT substrate motif, and recombinant kinase assays validated 16 as novel AKT substrates. These included CEP170 and FAM83H, suggesting a regulatory function of AKT in mitosis and cytoskeleton organization. In addition, a specific phosphorylation pattern on the ULK1-FIP200-ATG13-VAPB complex was found to determine the active state of ULK1, leading to elevated autophagy in response to AKT inhibition.
Collapse
Affiliation(s)
- Svenja Wiechmann
- Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
- German Cancer Consortium (DKTK), 80336 Munich, Germany
- German Cancer Center (DKFZ), 69120 Heidelberg, Germany
| | - Benjamin Ruprecht
- Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
| | - Theresa Siekmann
- Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
| | - Runsheng Zheng
- Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
| | - Martin Frejno
- Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
| | - Elena Kunold
- Organic Chemistry II, Technical University of Munich, 85748 Garching, Germany
| | - Thomas Bajaj
- Department of Psychiatry, Bonn Clinical Center, 53127 Bonn, Germany
| | - Daniel P. Zolg
- Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
| | - Stephan A. Sieber
- Organic Chemistry II, Technical University of Munich, 85748 Garching, Germany
| | - Nils C. Gassen
- Department of Psychiatry, Bonn Clinical Center, 53127 Bonn, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
- German Cancer Consortium (DKTK), 80336 Munich, Germany
- German Cancer Center (DKFZ), 69120 Heidelberg, Germany
- Bavarian Center for Biomolecular Mass Spectrometry, 85354 Freising, Germany
| |
Collapse
|
27
|
Abstract
Incorporation of heterocycles into drug molecules can enhance physical properties and biological activity. A variety of heterocyclic groups is available to medicinal chemists, many of which have been reviewed in detail elsewhere. Oxadiazoles are a class of heterocycle containing one oxygen and two nitrogen atoms, available in three isomeric forms. While the 1,2,4- and 1,3,4-oxadiazoles have seen widespread application in medicinal chemistry, 1,2,5-oxadiazoles (furazans) are less common. This Review provides a summary of the application of furazan-containing molecules in medicinal chemistry and drug development programs from analysis of both patent and academic literature. Emphasis is placed on programs that reached clinical or preclinical stages of development. The examples provided herein describe the pharmacology and biological activity of furazan derivatives with comparative data provided where possible for other heterocyclic groups and pharmacophores commonly used in medicinal chemistry.
Collapse
Affiliation(s)
| | | | - Donald F Weaver
- Department of Fundamental Neurobiology, Krembil Research Institute, Toronto, Ontario M5T 0S8, Canada.,Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | - Mark A Reed
- Treventis Corporation, Toronto, Ontario M5T 0S8, Canada.,Department of Fundamental Neurobiology, Krembil Research Institute, Toronto, Ontario M5T 0S8, Canada
| |
Collapse
|
28
|
Ethyl Acetate Fraction of Helianthus tuberosus L. Induces Anti-Diabetic, and Wound-Healing Activities in Insulin-Resistant Human Liver Cancer and Mouse Fibroblast Cells. Antioxidants (Basel) 2021; 10:antiox10010099. [PMID: 33445702 PMCID: PMC7828129 DOI: 10.3390/antiox10010099] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/28/2020] [Accepted: 01/05/2021] [Indexed: 12/14/2022] Open
Abstract
Traditional, complementary, and integrative medicine are globally accepted alternative methods for the treatment of diabetes mellitus (DM). However, the mechanism of anti-diabetic effects of Helianthus tuberosus L. remains unproven. In the present study, antioxidant and anti-diabetic activity of the tubers of H. tuberosus were studied in detail. Methanolic extracts of H. tuberosus tubers were subjected to solvent fractionation method by increasing the polarity of the solvent using n-hexane, and ethyl acetate. The obtained methanol extracts and its fractions were subjected to free radical scavenging activity (DPPH and ABTS assay) and in vitro enzyme (α-amylase and α-glucosidase) inhibition assay. Moreover, glucose uptake in insulin-resistant HepG2 cell line was analyzed. The preliminary phytochemical analysis confirmed the presence of phenolic and flavonoid compounds in the active fraction. The radical scavenging and in vitro diabetic related enzyme inhibitory activities were found to be dose dependent. The maximum ABTS+ and DPPH scavenging activity was documented in ethyl acetate fraction of the H. tuberosus followed by methanol extract, hexane fraction, and methanol fraction. We also found that H. tuberosus showed a less toxicity in mouse fibroblast cells and enhance the glucose uptake in insulin-resistant HepG2 cells. Besides, the ethyl acetate fraction of the H. tuberosus analyzed by UPLC-QTOF-MS-MS and GC/MS revealed the presence of phenolic compounds such as neochlorogenic acid, chlorogenic acid, caffeic acid, 5-O-(4-coumaroyl)-quinic acid, feruloylquinic acid, caffeoylquinic acid, isoxazolidine, salicylic acid β-D-glucoside, dicaffeoylquinic acid isomers, salvianolic acid derivative isomers, and 1,4 dicaffeoylquinic acid etc. Among the identified phytochemicals, six were chosen for molecular docking study to explore their its inhibitory interactions with α-amylase and α-glucosidase. Taken together, the findings of the present study suggested that phytocompounds of EAF were responsible for the significant in vitro antioxidant, wound-healing, and anti-diabetic activities.
Collapse
|
29
|
Alqawlaq S, Livne-Bar I, Williams D, D'Ercole J, Leung SW, Chan D, Tuccitto A, Datti A, Wrana JL, Corbett AH, Schmitt-Ulms G, Sivak JM. An endogenous PI3K interactome promoting astrocyte-mediated neuroprotection identifies a novel association with RNA-binding protein ZC3H14. J Biol Chem 2021; 296:100118. [PMID: 33234594 PMCID: PMC7948738 DOI: 10.1074/jbc.ra120.015389] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 11/06/2022] Open
Abstract
Astrocytes can support neuronal survival through a range of secreted signals that protect against neurotoxicity, oxidative stress, and apoptotic cascades. Thus, analyzing the effects of the astrocyte secretome may provide valuable insight into these neuroprotective mechanisms. Previously, we characterized a potent neuroprotective activity mediated by retinal astrocyte conditioned media (ACM) on retinal and cortical neurons in metabolic stress models. However, the molecular mechanism underlying this complex activity in neuronal cells has remained unclear. Here, a chemical genetics screen of kinase inhibitors revealed phosphoinositide 3-kinase (PI3K) as a central player transducing ACM-mediated neuroprotection. To identify additional proteins contributing to the protective cascade, endogenous PI3K was immunoprecipitated from neuronal cells exposed to ACM or control media, followed by MS/MS proteomic analyses. These data pointed toward a relatively small number of proteins that coimmunoprecipitated with PI3K, and surprisingly only five were regulated by the ACM signal. These hits included expected PI3K interactors, such as the platelet-derived growth factor receptor A (PDGFRA), as well as novel RNA-binding protein interactors ZC3H14 (zinc finger CCCH-type containing 14) and THOC1 (THO complex protein 1). In particular, ZC3H14 has recently emerged as an important RNA-binding protein with multiple roles in posttranscriptional regulation. In validation studies, we show that PI3K recruitment of ZC3H14 is necessary for PDGF-induced neuroprotection and that this interaction is present in primary retinal ganglion cells. Thus, we identified a novel non-cell autonomous neuroprotective signaling cascade mediated through PI3K that requires recruitment of ZC3H14 and may present a promising strategy to promote astrocyte-secreted prosurvival signals.
Collapse
Affiliation(s)
- Samih Alqawlaq
- Department of Vision Science, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Ophthalmology and Vision Science, University of Toronto School of Medicine, Toronto, Ontario, Canada
| | - Izhar Livne-Bar
- Department of Vision Science, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Ophthalmology and Vision Science, University of Toronto School of Medicine, Toronto, Ontario, Canada
| | - Declan Williams
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Ontario, Canada
| | - Joseph D'Ercole
- Department of Vision Science, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Ophthalmology and Vision Science, University of Toronto School of Medicine, Toronto, Ontario, Canada
| | - Sara W Leung
- Department of Biology, Emory University, Atlanta, Georgia, USA
| | - Darren Chan
- Department of Vision Science, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Ophthalmology and Vision Science, University of Toronto School of Medicine, Toronto, Ontario, Canada
| | - Alessandra Tuccitto
- Department of Vision Science, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Ophthalmology and Vision Science, University of Toronto School of Medicine, Toronto, Ontario, Canada
| | - Alessandro Datti
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Jeffrey L Wrana
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Anita H Corbett
- Department of Biology, Emory University, Atlanta, Georgia, USA
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Ontario, Canada
| | - Jeremy M Sivak
- Department of Vision Science, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Ophthalmology and Vision Science, University of Toronto School of Medicine, Toronto, Ontario, Canada.
| |
Collapse
|
30
|
Cavanah P, Itou J, Rusman Y, Tahara N, Williams JM, Salomon CE, Kawakami Y. A nontoxic fungal natural product modulates fin regeneration in zebrafish larvae upstream of FGF-WNT developmental signaling. Dev Dyn 2020; 250:160-174. [PMID: 32857425 DOI: 10.1002/dvdy.244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The regeneration of larvae zebrafish fin emerged as a new model of regeneration in the last decade. In contrast to genetic tools to study fin regeneration, chemical probes to modulate and interrogate regeneration processes are not well developed. RESULTS We set up a zebrafish larvae fin regeneration assay system and tested activities of natural product compounds and extracts, prepared from various microbes. Colomitide C, a recently isolated product from a fungus obtained from Antarctica, inhibited larvae fin regeneration. Using fluorescent reporter transgenic lines, we show that colomitide C inhibited fibroblast growth factor (FGF) signaling and WNT/β-catenin signaling, which were activated after larvae fin amputation. By using the endothelial cell reporter line and immunofluorescence, we showed that colomitide C did not affect migration of the blood vessel and nerve into the injured larvae fin. Colomitide C did not show any cytotoxic activities when tested against FGF receptor-amplified human cancer cell lines. CONCLUSION Colomitide C, a natural product, modulated larvae fin regeneration likely acting upstream of FGF and WNT signaling. Colomitide C may serve as a template for developing new chemical probes to study regeneration and other biological processes.
Collapse
Affiliation(s)
- Paul Cavanah
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Junji Itou
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA.,Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA.,Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yudi Rusman
- Center for Drug Design, University of Minnesota, Minneapolis, Minnesota, USA
| | - Naoyuki Tahara
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA.,Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA.,Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jessica M Williams
- Center for Drug Design, University of Minnesota, Minneapolis, Minnesota, USA
| | - Christine E Salomon
- Center for Drug Design, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yasuhiko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA.,Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA.,Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
31
|
Kostaras E, Kaserer T, Lazaro G, Heuss SF, Hussain A, Casado P, Hayes A, Yandim C, Palaskas N, Yu Y, Schwartz B, Raynaud F, Chung YL, Cutillas PR, Vivanco I. A systematic molecular and pharmacologic evaluation of AKT inhibitors reveals new insight into their biological activity. Br J Cancer 2020; 123:542-555. [PMID: 32439931 PMCID: PMC7435276 DOI: 10.1038/s41416-020-0889-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 04/07/2020] [Accepted: 04/24/2020] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND AKT, a critical effector of the phosphoinositide 3-kinase (PI3K) signalling cascade, is an intensely pursued therapeutic target in oncology. Two distinct classes of AKT inhibitors have been in clinical development, ATP-competitive and allosteric. Class-specific differences in drug activity are likely the result of differential structural and conformational requirements governing efficient target binding, which ultimately determine isoform-specific potency, selectivity profiles and activity against clinically relevant AKT mutant variants. METHODS We have carried out a systematic evaluation of clinical AKT inhibitors using in vitro pharmacology, molecular profiling and biochemical assays together with structural modelling to better understand the context of drug-specific and drug-class-specific cell-killing activity. RESULTS Our data demonstrate clear differences between ATP-competitive and allosteric AKT inhibitors, including differential effects on non-catalytic activity as measured by a novel functional readout. Surprisingly, we found that some mutations can cause drug resistance in an isoform-selective manner despite high structural conservation across AKT isoforms. Finally, we have derived drug-class-specific phosphoproteomic signatures and used them to identify effective drug combinations. CONCLUSIONS These findings illustrate the utility of individual AKT inhibitors, both as drugs and as chemical probes, and the benefit of AKT inhibitor pharmacological diversity in providing a repertoire of context-specific therapeutic options.
Collapse
Affiliation(s)
- Eleftherios Kostaras
- Division of Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, SM2 5NG, London, UK
| | - Teresa Kaserer
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, SW7 3RP, UK
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, A-6020, Austria
| | - Glorianne Lazaro
- Division of Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, SM2 5NG, London, UK
| | - Sara Farrah Heuss
- Division of Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, SM2 5NG, London, UK
| | - Aasia Hussain
- Division of Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, SM2 5NG, London, UK
| | - Pedro Casado
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Angela Hayes
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Cihangir Yandim
- Division of Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, SM2 5NG, London, UK
- Department of Genetics and Bioengineering, Faculty of Engineering, Izmir University of Economics, 35330, Balçova, Izmir, Turkey
| | - Nicolaos Palaskas
- Division of Hematology and Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yi Yu
- ArQule, Inc. (a wholly-owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA), Burlington, MA, 01803, USA
| | - Brian Schwartz
- ArQule, Inc. (a wholly-owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA), Burlington, MA, 01803, USA
| | - Florence Raynaud
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Yuen-Li Chung
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research London and Royal Marsden Hospital, London, SW7 3RP, UK
| | - Pedro R Cutillas
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - Igor Vivanco
- Division of Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, SM2 5NG, London, UK.
| |
Collapse
|
32
|
Ginsenoside Rh4 suppresses aerobic glycolysis and the expression of PD-L1 via targeting AKT in esophageal cancer. Biochem Pharmacol 2020; 178:114038. [PMID: 32422139 DOI: 10.1016/j.bcp.2020.114038] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022]
Abstract
Ginsenoside Rh4, as a bioactive component obtained from Panax notoginseng, has excellent pharmacological efficacy especially antitumor effects. However, its anticancer effects and target mechanisms in regulating human esophageal cancer are still poorly understood. Here, the results suggested that Rh4 exhibited potent anti-esophageal cancer effects in vivo and in vitro. Flow cytometric analysis and Western Blot showed that Rh4 significantly inhibited the growth by inducing G1 phase arrest. In parallel, Rh4 inhibited aerobic glycolysis in esophageal cancer cells by hindering lactate production, glucose uptake and ATP production; reducing the extracellular acidification rate (ECAR) and oxygen consumption rate (OCR); suppressing aerobic glycolysis-related protein expression. Mechanistic studies demonstrated that AKT is a possible target of Rh4, which suppresses aerobic glycolysis. Rh4 administration resulted in AKT deregulation, whereas treatment with insulin abolished the inhibitory effect of Rh4 on aerobic glycolysis. In contrast, treatment with AKT inhibitors or siRNA that silenced AKT enhanced the inhibitory effect of Rh4 on aerobic glycolysis. Moreover, molecular docking results indicated that Rh4 was able to bind to the interdomain region of AKT. Interestingly, the results revealed that Rh4 also inhibited the expression of PD-L1 via the AKT/mTOR pathway. Taken together, our findings provide important insights into the anti-esophageal cancer effects of Rh4 via suppressing aerobic glycolysis and PD-L1 expression, which indicated Rh4 could be as promising drug for clinical treatment.
Collapse
|
33
|
Markovics A, Angyal Á, Tóth KF, Ádám D, Pénzes Z, Magi J, Pór Á, Kovács I, Törőcsik D, Zouboulis CC, Bíró T, Oláh A. GPR119 Is a Potent Regulator of Human Sebocyte Biology. J Invest Dermatol 2020; 140:1909-1918.e8. [PMID: 32142797 DOI: 10.1016/j.jid.2020.02.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 01/31/2020] [Accepted: 02/12/2020] [Indexed: 12/16/2022]
Abstract
We have shown previously that endocannabinoids promote sebaceous lipogenesis, and sebocytes are involved in the metabolism of the endocannabinoid-like substance oleoylethanolamide (OEA). OEA is an endogenous activator of GPR119, a recently deorphanized receptor, which currently is being investigated as a promising antidiabetic drug target. In this study, we investigated the effects of OEA as well as the expression and role of GPR119 in human sebocytes. We found that OEA promoted differentiation of human SZ95 sebocytes (elevated lipogenesis, enhanced granulation, and the induction of early apoptotic events), and it switched the cells to a proinflammatory phenotype (increased expression and release of several proinflammatory cytokines). Moreover, we could also demonstrate that GPR119 was expressed in human sebocytes, and its small interfering RNA-mediated gene silencing suppressed OEA-induced sebaceous lipogenesis, which was mediated via c-Jun N-terminal kinase, extracellular signal-regulated kinase 1/2, protein kinase B, and CRE-binding protein activation. Finally, our pilot data demonstrated that GPR119 was downregulated in the sebaceous glands of patients with acne, arguing that GPR119 signaling may indeed be disturbed in acne. Collectively, our findings introduce the OEA/GPR119 signaling as a positive regulator of sebocyte differentiation and highlight the possibility that dysregulation of this pathway may contribute to the development of seborrhea and acne.
Collapse
Affiliation(s)
- Arnold Markovics
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; University of Debrecen, Doctoral School of Molecular Medicine, Debrecen, Hungary
| | - Ágnes Angyal
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; University of Debrecen, Doctoral School of Molecular Medicine, Debrecen, Hungary
| | - Kinga Fanni Tóth
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; University of Debrecen, Doctoral School of Molecular Medicine, Debrecen, Hungary
| | - Dorottya Ádám
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; University of Debrecen, Doctoral School of Molecular Medicine, Debrecen, Hungary
| | - Zsófia Pénzes
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; University of Debrecen, Doctoral School of Molecular Medicine, Debrecen, Hungary; Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - József Magi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ágnes Pór
- Department of Pathology, Gyula Kenézy University Hospital, University of Debrecen, Debrecen, Hungary
| | - Ilona Kovács
- Department of Pathology, Gyula Kenézy University Hospital, University of Debrecen, Debrecen, Hungary
| | - Dániel Törőcsik
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Christos C Zouboulis
- Departments of Dermatology, Venereology, Allergology and Immunology, Dessau Medical Center, Brandenburg Medical School Theodor Fontane, Dessau, Germany
| | - Tamás Bíró
- DE-MTA Lendület Cellular Physiology Research Group, Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Oláh
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
34
|
Zhang L, Chu Y, Ma P, Zhao S, Li Q, Chen B, Hong X, Sun J. Visible-light-mediated photocatalytic cross-coupling of acetenyl ketones with benzyl trifluoroborate. Org Biomol Chem 2020; 18:1073-1077. [PMID: 31960883 DOI: 10.1039/c9ob02624j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In this report, we describe a simple visible light-triggered Barbier-type reaction by employing acetenyl ketones with benzyl trifluoroborates. Through a radical-radical cross-coupling process, this photocatalytic protocol furnished a wide range of tertiary propargyl alcohols. Mechanistic investigation indicated that proton-coupled electron transfer (PCET) might be involved in the photochemical transformations.
Collapse
Affiliation(s)
- Lingchun Zhang
- Department of Pharmacy, Henan Provincial People's Hospital; Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, P. R. China.
| | - Yanle Chu
- Department of Pharmacy, The Second Affiliated Hospital of Zhengzhou University, 450000, P. R. China
| | - Peizhi Ma
- Department of Pharmacy, Henan Provincial People's Hospital; Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, P. R. China.
| | - Shujuan Zhao
- Department of Pharmacy, Henan Provincial People's Hospital; Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, P. R. China.
| | - Qiaoyan Li
- Department of Pharmacy, Henan Provincial People's Hospital; Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, P. R. China.
| | - Boya Chen
- Department of Pharmacy, Henan Provincial People's Hospital; Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, P. R. China.
| | - Xuejiao Hong
- Department of Pharmacy, Henan Provincial People's Hospital; Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, P. R. China.
| | - Jun Sun
- Department of Pharmacy, Henan Provincial People's Hospital; Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, P. R. China.
| |
Collapse
|
35
|
Makhova NN, Belen’kii LI, Gazieva GA, Dalinger IL, Konstantinova LS, Kuznetsov VV, Kravchenko AN, Krayushkin MM, Rakitin OA, Starosotnikov AM, Fershtat LL, Shevelev SA, Shirinian VZ, Yarovenko VN. Progress in the chemistry of nitrogen-, oxygen- and sulfur-containing heterocyclic systems. RUSSIAN CHEMICAL REVIEWS 2020. [DOI: 10.1070/rcr4914] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
36
|
Reinecke M, Heinzlmeir S, Wilhelm M, Médard G, Klaeger S, Kuster B. Kinobeads: A Chemical Proteomic Approach for Kinase Inhibitor Selectivity Profiling and Target Discovery. ACTA ACUST UNITED AC 2019. [DOI: 10.1002/9783527818242.ch4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
37
|
Eberl HC, Werner T, Reinhard FB, Lehmann S, Thomson D, Chen P, Zhang C, Rau C, Muelbaier M, Drewes G, Drewry D, Bantscheff M. Chemical proteomics reveals target selectivity of clinical Jak inhibitors in human primary cells. Sci Rep 2019; 9:14159. [PMID: 31578349 PMCID: PMC6775116 DOI: 10.1038/s41598-019-50335-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/05/2019] [Indexed: 12/15/2022] Open
Abstract
Kinobeads are a set of promiscuous kinase inhibitors immobilized on sepharose beads for the comprehensive enrichment of endogenously expressed protein kinases from cell lines and tissues. These beads enable chemoproteomics profiling of kinase inhibitors of interest in dose-dependent competition studies in combination with quantitative mass spectrometry. We present improved bead matrices that capture more than 350 protein kinases and 15 lipid kinases from human cell lysates, respectively. A multiplexing strategy is suggested that enables determination of apparent dissociation constants in a single mass spectrometry experiment. Miniaturization of the procedure enabled determining the target selectivity of the clinical BCR-ABL inhibitor dasatinib in peripheral blood mononuclear cell (PBMC) lysates from individual donors. Profiling of a set of Jak kinase inhibitors revealed kinase off-targets from nearly all kinase families underpinning the need to profile kinase inhibitors against the kinome. Potently bound off-targets of clinical inhibitors suggest polypharmacology, e.g. through MRCK alpha and beta, which bind to decernotinib with nanomolar affinity.
Collapse
Affiliation(s)
- H Christian Eberl
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany.
| | - Thilo Werner
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Friedrich B Reinhard
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Stephanie Lehmann
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Douglas Thomson
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Peiling Chen
- GlaxoSmithKline, Upper Merion, 709 Swedeland Rd #1539, King of Prussia, PA, 19406, United States
| | - Cunyu Zhang
- GlaxoSmithKline, Upper Merion, 709 Swedeland Rd #1539, King of Prussia, PA, 19406, United States
| | - Christina Rau
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Marcel Muelbaier
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Gerard Drewes
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - David Drewry
- GlaxoSmithKline, Research Triangle Park, 5 Moore Drive, North Carolina, 27709, United States.,UNC Eshelman School of Pharmacy, Structural Genomics Consortium, University of North Carolina at Chapel Hill, 120 Mason Farm Rd, Chapel Hill, NC, 27599, United States
| | - Marcus Bantscheff
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany.
| |
Collapse
|
38
|
Hirozane Y, Toyofuku M, Yogo T, Tanaka Y, Sameshima T, Miyahisa I, Yoshikawa M. Structure-based rational design of staurosporine-based fluorescent probe with broad-ranging kinase affinity for kinase panel application. Bioorg Med Chem Lett 2019; 29:126641. [PMID: 31526603 DOI: 10.1016/j.bmcl.2019.126641] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/20/2019] [Accepted: 08/24/2019] [Indexed: 02/08/2023]
Abstract
Selectivity profiling of compounds is important for kinase drug discovery. To this end, we aimed to develop a broad-range protein kinase assay by synthesizing a novel staurosporine-derived fluorescent probe based on staurosporine and kinase-binding related structural information. Upon structural analysis of staurosporine with kinases, a 4'-methylamine moiety of staurosporine was found to be located on the solvent side of the kinases, to which several linker units can be conjugated by either alkylation or acylation. However, such conjugation was suggested to reduce the binding affinities of the modified compound for several kinases, owing to the elimination of hydrogen bond donor moiety of NH-group from 4'-methylamine and/or steric hindrance by acyl moiety. Based on this structural information, we designed and synthesized a novel staurosporine-based probe without methyl group in order to retain the hydrogen bond donor, similar to unmodified staurosporine. The broad range of the kinase binding assay demonstrated that our novel fluorescent probe is an excellent tool for developing broad-ranging kinase binding assay.
Collapse
Affiliation(s)
- Yoshihiko Hirozane
- innovative Biology Laboratories, Neuroscience Drug Discovery Unit, Japan; Biomolecular Research Laboratories, Pharmaceutical Research Division, Japan.
| | - Masashi Toyofuku
- Drug Discovery Chemistry Laboratories, Neuroscience Drug Discovery Unit, Japan
| | - Takatoshi Yogo
- Drug Discovery Chemistry Laboratories, Neuroscience Drug Discovery Unit, Japan
| | - Yukiya Tanaka
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Japan
| | - Tomoya Sameshima
- Drug Safety Research Laboratories, Japan; Biomolecular Research Laboratories, Pharmaceutical Research Division, Japan
| | - Ikuo Miyahisa
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Japan
| | - Masato Yoshikawa
- Drug Discovery Chemistry Laboratories, Neuroscience Drug Discovery Unit, Japan
| |
Collapse
|
39
|
Dong X, Zhan W, Zhao M, Che J, Dai X, Wu Y, Xu L, Zhou Y, Zhao Y, Tian T, Cheng G, Jin Z, Li J, Shao Y, He Q, Yang B, Weng Q, Hu Y. Discovery of 3,4,6-Trisubstituted Piperidine Derivatives as Orally Active, Low hERG Blocking Akt Inhibitors via Conformational Restriction and Structure-Based Design. J Med Chem 2019; 62:7264-7288. [DOI: 10.1021/acs.jmedchem.9b00891] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
| | | | | | | | | | | | - Lei Xu
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yubo Zhou
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | | | | | - Gang Cheng
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 311402, China
| | | | - Jia Li
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yanfei Shao
- Department of Pharmacy, Zhejiang Provincial People’s Hospital, Hangzhou 310014, China
| | | | | | | | | |
Collapse
|
40
|
Amable G, Martínez-León E, Picco ME, Di Siervi N, Davio C, Rozengurt E, Rey O. Metformin inhibits β-catenin phosphorylation on Ser-552 through an AMPK/PI3K/Akt pathway in colorectal cancer cells. Int J Biochem Cell Biol 2019; 112:88-94. [PMID: 31082618 DOI: 10.1016/j.biocel.2019.05.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/06/2019] [Accepted: 05/08/2019] [Indexed: 02/08/2023]
Abstract
Several epidemiologic studies have revealed strong inverse associations between metformin use and risk of colorectal cancer development. Nevertheless, the underlying mechanisms are still uncertain. The Wnt/β-catenin pathway, which plays a central role in intestinal homeostasis and sporadic colorectal cancer development, is regulated by phosphorylation cascades that are dependent and independent of Wnt. Here we report that a non-canonical Ser552 phosphorylation in β-catenin, which promotes its nuclear accumulation and transcriptional activity, is blocked by metformin via AMPK-mediated PI3K/Akt signaling inhibition.
Collapse
Affiliation(s)
- Gastón Amable
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín", Caba, 1120, Argentina
| | - Eduardo Martínez-León
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín", Caba, 1120, Argentina
| | - María Elisa Picco
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín", Caba, 1120, Argentina
| | - Nicolas Di Siervi
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Investigaciones Farmacológicas, Facultad de Farmacia y Bioquímica, Argentina
| | - Carlos Davio
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Investigaciones Farmacológicas, Facultad de Farmacia y Bioquímica, Argentina; Departamento de Farmacología, Caba, 1113, Argentina
| | - Enrique Rozengurt
- Unit of Signal Transduction and Gastrointestinal Cancer, Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, Molecular Biology Institute and Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles, CA, 90095-1786, USA
| | - Osvaldo Rey
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín", Caba, 1120, Argentina.
| |
Collapse
|
41
|
Song M, Bode AM, Dong Z, Lee MH. AKT as a Therapeutic Target for Cancer. Cancer Res 2019; 79:1019-1031. [PMID: 30808672 DOI: 10.1158/0008-5472.can-18-2738] [Citation(s) in RCA: 491] [Impact Index Per Article: 98.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/15/2018] [Accepted: 12/26/2018] [Indexed: 11/16/2022]
Abstract
Many cellular processes in cancer are attributed to kinase signaling networks. V-akt murine thymoma viral oncogene homolog (AKT) plays a major role in the PI3K/AKT signaling pathways. AKT is activated by PI3K or phosphoinositide-dependent kinases (PDK) as well as growth factors, inflammation, and DNA damage. Signal transduction occurs through downstream effectors such as mTOR, glycogen synthase kinase 3 beta (GSK3β), or forkhead box protein O1 (FOXO1). The abnormal overexpression or activation of AKT has been observed in many cancers, including ovarian, lung, and pancreatic cancers, and is associated with increased cancer cell proliferation and survival. Therefore, targeting AKT could provide an important approach for cancer prevention and therapy. In this review, we discuss the rationale for targeting AKT and also provide details regarding synthetic and natural AKT-targeting compounds and their associated studies.
Collapse
Affiliation(s)
- Mengqiu Song
- Basic Medical College, Zhengzhou University, Zhengzhou, Henan, China.,China-US (Henan) Hormel Cancer Institute, Jinshui District, Zhengzhou, Henan, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Zigang Dong
- Basic Medical College, Zhengzhou University, Zhengzhou, Henan, China. .,China-US (Henan) Hormel Cancer Institute, Jinshui District, Zhengzhou, Henan, China.,The Hormel Institute, University of Minnesota, Austin, Minnesota.,The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, China
| | - Mee-Hyun Lee
- Basic Medical College, Zhengzhou University, Zhengzhou, Henan, China. .,China-US (Henan) Hormel Cancer Institute, Jinshui District, Zhengzhou, Henan, China.,The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, China
| |
Collapse
|
42
|
Mahajan P, Wadhwa B, Barik MR, Malik F, Nargotra A. Combining ligand- and structure-based in silico methods for the identification of natural product-based inhibitors of Akt1. Mol Divers 2019; 24:45-60. [PMID: 30798436 DOI: 10.1007/s11030-019-09924-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/29/2019] [Indexed: 01/08/2023]
Abstract
The traditional method of drug discovery process has been surpassed by a rational approach where computer-aided drug designing plays a vital role in the identification of leads from large compound databases. Further, natural products have an important role in drug discovery as these have been the source of most active ingredients of medicines. Herein, in silico structure- and ligand-based approaches have been applied to screen in-house IIIM natural product repository for Akt1 (serine/threonine protein kinases) which is a well-known therapeutic target for cancer due to its overexpression and preventing the cells from undergoing apoptosis. Combined ligand-based and structure-based strategies were applied on to the existing library comprising of about 700 pure natural products, and the compounds identified from screening were biologically evaluated for Akt1 inhibition using Akt1 kinase activity assay. Fourteen promising compounds showed significant inhibition at 500 nM through in vitro screening, and from them, eight were new for Akt1 inhibition. Through the MD studies of Akt1 with the most active compound IN00145, it was inferred that Lys179, Glu191, Glu228, Ala230, Glu234 and Asp292 are the important amino acid residues which provide stability to the Akt1-IN00145 complex. Lead optimization studies were also performed around the actives to design better and selective inhibitors for Akt1. The results emphasized the successful application of virtual screening to identify new Akt1 inhibitor scaffolds that can be developed into a drug candidate in drug discovery programme.
Collapse
Affiliation(s)
- Priya Mahajan
- Discovery Informatics Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Bhumika Wadhwa
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Manas Ranjan Barik
- Discovery Informatics Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Fayaz Malik
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Amit Nargotra
- Discovery Informatics Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India. .,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India.
| |
Collapse
|
43
|
Lv Y, Du T, Ji M, Wang C, Lin S, Xue N, Jin J, Xu H, Chen X. A novel PI3K/mTOR dual inhibitor XH002 exhibited robust antitumor activity in NSCLC. J Drug Target 2018; 27:451-459. [DOI: 10.1080/1061186x.2018.1542533] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Yuanhao Lv
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tingting Du
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming Ji
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunyang Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Songwen Lin
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nina Xue
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Jin
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Heng Xu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoguang Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
44
|
Wlodarchak N, Teachout N, Beczkiewicz J, Procknow R, Schaenzer AJ, Satyshur K, Pavelka M, Zuercher W, Drewry D, Sauer JD, Striker R. In Silico Screen and Structural Analysis Identifies Bacterial Kinase Inhibitors which Act with β-Lactams To Inhibit Mycobacterial Growth. Mol Pharm 2018; 15:5410-5426. [PMID: 30285456 PMCID: PMC6648700 DOI: 10.1021/acs.molpharmaceut.8b00905] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
New tools and concepts are needed to combat antimicrobial resistance. Actinomycetes and firmicutes share several eukaryotic-like Ser/Thr kinases (eSTK) that offer antibiotic development opportunities, including PknB, an essential mycobacterial eSTK. Despite successful development of potent biochemical PknB inhibitors by many groups, clinically useful microbiologic activity has been elusive. Additionally, PknB kinetics are not fully described, nor are structures with specific inhibitors available to inform inhibitor design. We used computational modeling with available structural information to identify human kinase inhibitors predicted to bind PknB, and we selected hits based on drug-like characteristics intended to increase the likelihood of cell entry. The computational model suggested a family of inhibitors, the imidazopyridine aminofurazans (IPAs), bind PknB with high affinity. We performed an in-depth characterization of PknB and found that these inhibitors biochemically inhibit PknB, with potency roughly following the predicted models. A novel X-ray structure confirmed that the inhibitors bound as predicted and made favorable protein contacts with the target. These inhibitors also have antimicrobial activity toward mycobacteria and nocardia. We demonstrated that the inhibitors are uniquely potentiated by β-lactams but not antibiotics traditionally used to treat mycobacteria, consistent with PknB's role in sensing cell wall stress. This is the first demonstration in the phylum actinobacteria that some β-lactam antibiotics could be more effective if paired with a PknB inhibitor. Collectively, our data show that in silico modeling can be used as a tool to discover promising drug leads, and the inhibitors we discovered can act with clinically relevant antibiotics to restore their efficacy against bacteria with limited treatment options.
Collapse
Affiliation(s)
- Nathan Wlodarchak
- Department of Medicine, University of Wisconsin-Madison, 3341 Microbial Sciences Building, 1550 Linden Dr., Madison, WI 53706
| | - Nathan Teachout
- Department of Medicine, University of Wisconsin-Madison, 3341 Microbial Sciences Building, 1550 Linden Dr., Madison, WI 53706
| | - Jeffrey Beczkiewicz
- Department of Medicine, University of Wisconsin-Madison, 3341 Microbial Sciences Building, 1550 Linden Dr., Madison, WI 53706
| | - Rebecca Procknow
- Department of Medicine, University of Wisconsin-Madison, 3341 Microbial Sciences Building, 1550 Linden Dr., Madison, WI 53706
| | - Adam J. Schaenzer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, 4203 Microbial Sciences Building, 1550 Linden Dr., Madison, WI 53706
| | - Kenneth Satyshur
- Small Molecule Screening Facility, Carbone Cancer Center, University of Wisconsin-Madison, 1111Highland Ave., Madison, WI 53705
| | - Martin Pavelka
- School of Medicine and Dentistry, University of Rochester Medical Center, 601 Elmwood Ave., Rochester, NY 14620
| | - William Zuercher
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, SGC Center for Chemical Biology, 120 Mason Farm Rd., Chapel Hill, NC 27599
| | - David Drewry
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, SGC Center for Chemical Biology, 120 Mason Farm Rd., Chapel Hill, NC 27599
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, 4203 Microbial Sciences Building, 1550 Linden Dr., Madison, WI 53706
| | - Rob Striker
- Department of Medicine, University of Wisconsin-Madison, 3341 Microbial Sciences Building, 1550 Linden Dr., Madison, WI 53706,William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terr., Madison, WI 53705,To whom correspondence should be addressed Rob Striker, Department of Medicine, University of Wisconsin-Madison, 3301 Microbial Sciences Building, 1550 Linden Dr., Madison, WI 53706, 608-263-2994,
| |
Collapse
|
45
|
Zhao S, Jiang Y, Zhao J, Li H, Yin X, Wang Y, Xie Y, Chen X, Lu J, Dong Z, Liu K. Quercetin‐3‐methyl ether inhibits esophageal carcinogenesis by targeting the AKT/mTOR/p70S6K and MAPK pathways. Mol Carcinog 2018; 57:1540-1552. [DOI: 10.1002/mc.22876] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/06/2018] [Accepted: 07/19/2018] [Indexed: 11/12/2022]
Affiliation(s)
- Simin Zhao
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
| | - Yanan Jiang
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
| | - Jimin Zhao
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
| | - Honglin Li
- East China University of Science and TechnologyShanghaiChina
| | - Xueshan Yin
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
- The First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yanhong Wang
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
| | - Yifei Xie
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
| | - Xinhuan Chen
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
| | - Jing Lu
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
| | - Ziming Dong
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
| | - Kangdong Liu
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
| |
Collapse
|
46
|
Zhang D, Tong D, Yang D, Sun J, Zhang F, Zhao G. Design, synthesis and biological evaluation of AKT inhibitors bearing a piperidin-4-yl appendant. MEDCHEMCOMM 2018; 9:1340-1350. [PMID: 30151089 DOI: 10.1039/c8md00197a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/28/2018] [Indexed: 11/21/2022]
Abstract
A series of AKT inhibitors possessing a piperidin-4-yl side chain was designed and synthesized. Some of them showed high AKT1 inhibitory activity and potent anti-proliferative effect on PC-3 prostate cancer cells in the preliminary screening. Further studies revealed the most potent compound, 10h, as a pan-AKT inhibitor. Compound 10h was able to inhibit the cellular phosphorylation of AKT effectively and induce apoptosis of PC-3 cells. It also showed high metabolic stability in human liver microsomes. Preclinical characterization of 10h, a promising lead AKT inhibitor, as a potential anti-prostate cancer therapeutic needs to be further investigated.
Collapse
Affiliation(s)
- Daoguang Zhang
- Department of Medicinal Chemistry , Key Laboratory of Chemical Biology (Ministry of Education) , School of Pharmaceutical Sciences , Shandong University , Jinan , Shandong 250012 , PR China . ; ; Tel: +86 531 88382009
| | - Dongdong Tong
- Department of Oral and Maxillofacial Surgery , School of Stomatology , Shandong Provincial Key Laboratory of Oral Tissue Regeneration , Shandong University , Jinan , Shandong 250012 , PR China . ; ; Tel: +86 531 88382961
| | - Dezhi Yang
- National Pharmaceutical Experimental Teaching Demonstration Center, School of Pharmacy , Zunyi Medical University , Zunyi , Guizhou 563003 , PR China
| | - Jing Sun
- Department of Bone Metabolism , School of Stomatology, Shandong Provincial Key Laboratory of Oral Tissue Regeneration , Shandong University , Jinan , Shandong 250012 , PR China
| | - Fenghe Zhang
- Department of Oral and Maxillofacial Surgery , School of Stomatology , Shandong Provincial Key Laboratory of Oral Tissue Regeneration , Shandong University , Jinan , Shandong 250012 , PR China . ; ; Tel: +86 531 88382961
| | - Guisen Zhao
- Department of Medicinal Chemistry , Key Laboratory of Chemical Biology (Ministry of Education) , School of Pharmaceutical Sciences , Shandong University , Jinan , Shandong 250012 , PR China . ; ; Tel: +86 531 88382009
| |
Collapse
|
47
|
Pérez Ortiz JM, Mollema N, Toker N, Adamski CJ, O'Callaghan B, Duvick L, Friedrich J, Walters MA, Strasser J, Hawkinson JE, Zoghbi HY, Henzler C, Orr HT, Lagalwar S. Reduction of protein kinase A-mediated phosphorylation of ATXN1-S776 in Purkinje cells delays onset of Ataxia in a SCA1 mouse model. Neurobiol Dis 2018; 116:93-105. [PMID: 29758256 DOI: 10.1016/j.nbd.2018.05.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/17/2018] [Accepted: 05/09/2018] [Indexed: 12/27/2022] Open
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a polyglutamine (polyQ) repeat neurodegenerative disease in which a primary site of pathogenesis are cerebellar Purkinje cells. In addition to polyQ expansion of ataxin-1 protein (ATXN1), phosphorylation of ATXN1 at the serine 776 residue (ATXN1-pS776) plays a significant role in protein toxicity. Utilizing a biochemical approach, pharmacological agents and cell-based assays, including SCA1 patient iPSC-derived neurons, we examine the role of Protein Kinase A (PKA) as an effector of ATXN1-S776 phosphorylation. We further examine the implications of PKA-mediated phosphorylation at ATXN1-S776 on SCA1 through genetic manipulation of the PKA catalytic subunit Cα in Pcp2-ATXN1[82Q] mice. Here we show that pharmacologic inhibition of S776 phosphorylation in transfected cells and SCA1 patient iPSC-derived neuronal cells lead to a decrease in ATXN1. In vivo, reduction of PKA-mediated ATXN1-pS776 results in enhanced degradation of ATXN1 and improved cerebellar-dependent motor performance. These results provide evidence that PKA is a biologically important kinase for ATXN1-pS776 in cerebellar Purkinje cells.
Collapse
Affiliation(s)
- Judit M Pérez Ortiz
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States; Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Nissa Mollema
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Nicholas Toker
- Skidmore College Neuroscience Program, Saratoga Springs, NY, United States
| | - Carolyn J Adamski
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, and Department of Molecular and Human Genetics, Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, United States
| | - Brennon O'Callaghan
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Lisa Duvick
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Jillian Friedrich
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Michael A Walters
- Department of Medicinal Chemistry, Institute for Therapeutics Discovery and Development, University of Minnesota, United States
| | - Jessica Strasser
- Department of Medicinal Chemistry, Institute for Therapeutics Discovery and Development, University of Minnesota, United States
| | - Jon E Hawkinson
- Department of Medicinal Chemistry, Institute for Therapeutics Discovery and Development, University of Minnesota, United States
| | - Huda Y Zoghbi
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, and Department of Molecular and Human Genetics, Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, United States
| | - Christine Henzler
- RISS Bioinformatics, Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, United States
| | - Harry T Orr
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States.
| | - Sarita Lagalwar
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States; Skidmore College Neuroscience Program, Saratoga Springs, NY, United States.
| |
Collapse
|
48
|
Konno H, Chinn IK, Hong D, Orange JS, Lupski JR, Mendoza A, Pedroza LA, Barber GN. Pro-inflammation Associated with a Gain-of-Function Mutation (R284S) in the Innate Immune Sensor STING. Cell Rep 2018; 23:1112-1123. [PMID: 29694889 PMCID: PMC6092751 DOI: 10.1016/j.celrep.2018.03.115] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/30/2018] [Accepted: 03/26/2018] [Indexed: 12/22/2022] Open
Abstract
The cellular sensor stimulator of interferon genes (STING) initiates type I interferon (IFN) and cytokine production following association with cyclic dinucleotides (CDNs) generated from intracellular bacteria or via a cellular synthase, cGAS, after binding microbial or self-DNA. Although essential for protecting the host against infection, unscheduled STING signaling is now known to be responsible for a variety of autoinflammatory disorders. Here, we report a gain-of-function mutation in STING (R284S), isolated from a patient who did not require CDNs to augment activity and who manifested a constitutively active phenotype. Control of the Unc-51-like autophagy activating kinase 1 (ULK1) pathway, which has previously been shown to influence STING function, was potently able to suppress STING (R284S) activity to alleviate cytokine production. Our findings add to the growing list of inflammatory syndromes associated with spontaneous STING signaling and provide a therapeutic strategy for the treatment of STING-induced inflammatory disease.
Collapse
Affiliation(s)
- Hiroyasu Konno
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ivan K Chinn
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Division of Immunology/Allergy/Rheumatology, Texas Children's Hospital, Houston, TX 77030, USA; Center for Human Immunobiology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - Diana Hong
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Division of Immunology/Allergy/Rheumatology, Texas Children's Hospital, Houston, TX 77030, USA; Center for Human Immunobiology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - Jordan S Orange
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Division of Immunology/Allergy/Rheumatology, Texas Children's Hospital, Houston, TX 77030, USA; Center for Human Immunobiology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - James R Lupski
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Hospital, Houston, TX 77030, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alejandra Mendoza
- Colegio de Ciencias de la Salud-Hospital de los Valles, Universidad San Francisco de Quito, Quito, Ecuador
| | - Luis A Pedroza
- Colegio de Ciencias de la Salud-Hospital de los Valles, Universidad San Francisco de Quito, Quito, Ecuador
| | - Glen N Barber
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
49
|
Kadioglu O, Saeed M, Kuete V, Greten HJ, Efferth T. Oridonin Targets Multiple Drug-Resistant Tumor Cells as Determined by in Silico and in Vitro Analyses. Front Pharmacol 2018; 9:355. [PMID: 29713280 PMCID: PMC5911471 DOI: 10.3389/fphar.2018.00355] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/27/2018] [Indexed: 01/08/2023] Open
Abstract
Drug resistance is one of the main reasons of chemotherapy failure. Therefore, overcoming drug resistance is an invaluable approach to identify novel anticancer drugs that have the potential to bypass or overcome resistance to established drugs and to substantially increase life span of cancer patients for effective chemotherapy. Oridonin is a cytotoxic diterpenoid isolated from Rabdosia rubescens with in vivo anticancer activity. In the present study, we evaluated the cytotoxicity of oridonin toward a panel of drug-resistant cancer cells overexpressing ABCB1, ABCG2, or ΔEGFR or with a knockout deletion of TP53. Interestingly, oridonin revealed lower degree of resistance than the control drug, doxorubicin. Molecular docking analyses pointed out that oridonin can interact with Akt/EGFR pathway proteins with comparable binding energies and similar docking poses as the known inhibitors. Molecular dynamics results validated the stable conformation of oridonin docking pose on Akt kinase domain. Western blot experiments clearly revealed dose-dependent downregulation of Akt and STAT3. Pharmacogenomics analyses pointed to a mRNA signature that predicted sensitivity and resistance to oridonin. In conclusion, oridonin bypasses major drug resistance mechanisms and targets Akt pathway and might be effective toward drug refractory tumors. The identification of oridonin-specific gene expressions may be useful for the development of personalized treatment approaches.
Collapse
Affiliation(s)
- Onat Kadioglu
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Mohamed Saeed
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Victor Kuete
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Henry J Greten
- Abel Salazar Institute of Biomedical Sciences, University of Porto, Porto, Portugal.,Heidelberg School of Chinese Medicine, Heidelberg, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
50
|
Copeland RA, Boriack-Sjodin PA. The Elements of Translational Chemical Biology. Cell Chem Biol 2018; 25:128-134. [DOI: 10.1016/j.chembiol.2017.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/01/2017] [Accepted: 11/08/2017] [Indexed: 02/06/2023]
|