1
|
Van Dort ME, McDonald L, Jang Y, Heist K, Bonham CA, Abraskin K, Chenevert TL, Ross BD. Synthesis and Biological Evaluation of MEK/mTOR Multifunctional Inhibitors as Novel Anticancer Agents. J Med Chem 2025. [PMID: 40375738 DOI: 10.1021/acs.jmedchem.5c00376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
The mitogen-activated protein kinase (MAPK) and mechanistic target of rapamycin (mTOR) signaling nodes play a crucial role in many human cancers. Due to the molecular reciprocity between MAPK and mTOR signaling nodes, development of compounds with multikinase targeting was explored. A series of mTOR inhibitor analogs of AZD8055 and AZD2014 were designed to allow for covalent linking to a potent MAPK kinase (MEK) inhibitor to produce a single, bivalent chemical entity. Dual-acting agents (i.e., compound LP-65) were synthesized displaying high in vitro inhibition of both MEK (IC50 = 83.2 nM) and mTOR (IC50 = 40.5 nM). Additionally, compound LP-65 demonstrated significant modulation of MEK and mTOR signaling activity in human glioma cells (D54) and human melanoma cells (A375), with a corresponding decrease in cellular proliferation and migration. Treatment of mice with LP-65 (40 mg/kg) having a myeloproliferative neoplasm, myelofibrosis, revealed down modulation of in vivo signaling pathways and therapeutic efficacy.
Collapse
Affiliation(s)
- Marcian E Van Dort
- Center for Molecular Imaging, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
- Department of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Lucas McDonald
- Center for Molecular Imaging, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
- Department of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Youngsoon Jang
- Center for Molecular Imaging, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
- Department of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Kevin Heist
- Center for Molecular Imaging, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
- Department of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Christopher A Bonham
- Center for Molecular Imaging, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
- Department of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Kamryn Abraskin
- Center for Molecular Imaging, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
- Department of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Thomas L Chenevert
- Center for Molecular Imaging, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
- Department of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Brian D Ross
- Center for Molecular Imaging, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
- Department of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
- Department of Biological Chemistry, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
2
|
Hu J, Fu S, Zhan Z, Zhang J. Advancements in dual-target inhibitors of PI3K for tumor therapy: Clinical progress, development strategies, prospects. Eur J Med Chem 2024; 265:116109. [PMID: 38183777 DOI: 10.1016/j.ejmech.2023.116109] [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: 12/05/2023] [Revised: 12/24/2023] [Accepted: 12/28/2023] [Indexed: 01/08/2024]
Abstract
Phosphoinositide 3-kinases (PI3Ks) modify lipids by the phosphorylation of inositol phospholipids at the 3'-OH position, thereby participating in signal transduction and exerting effects on various physiological processes such as cell growth, metabolism, and organism development. PI3K activation also drives cancer cell growth, survival, and metabolism, with genetic dysregulation of this pathway observed in diverse human cancers. Therefore, this target is considered a promising potential therapeutic target for various types of cancer. Currently, several selective PI3K inhibitors and one dual-target PI3K inhibitor have been approved and launched on the market. However, the majority of these inhibitors have faced revocation or voluntary withdrawal of indications due to concerns regarding their adverse effects. This article provides a comprehensive review of the structure and biological functions, and clinical status of PI3K inhibitors, with a specific emphasis on the development strategies and structure-activity relationships of dual-target PI3K inhibitors. The findings offer valuable insights and future directions for the development of highly promising dual-target drugs targeting PI3K.
Collapse
Affiliation(s)
- Jiarui Hu
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Siyu Fu
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zixuan Zhan
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jifa Zhang
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
3
|
Dai Q, Sun Q, Ouyang X, Liu J, Jin L, Liu A, He B, Fan T, Jiang Y. Antitumor Activity of s-Triazine Derivatives: A Systematic Review. Molecules 2023; 28:molecules28114278. [PMID: 37298753 DOI: 10.3390/molecules28114278] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/15/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
1,3,5-triazine derivatives, also called s-triazines, are a series of containing-nitrogen heterocyclic compounds that play an important role in anticancer drug design and development. To date, three s-triazine derivatives, including altretamine, gedatolisib, and enasidenib, have already been approved for refractory ovarian cancer, metastatic breast cancer, and leukemia therapy, respectively, demonstrating that the s-triazine core is a useful scaffold for the discovery of novel anticancer drugs. In this review, we mainly focus on s-triazines targeting topoisomerases, tyrosine kinases, phosphoinositide 3-kinases, NADP+-dependent isocitrate dehydrogenases, and cyclin-dependent kinases in diverse signaling pathways, which have been extensively studied. The medicinal chemistry of s-triazine derivatives as anticancer agents was summarized, including discovery, structure optimization, and biological applications. This review will provide a reference to inspire new and original discoveries.
Collapse
Affiliation(s)
- Qiuzi Dai
- The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, China
| | - Qinsheng Sun
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen 518055, China
| | - Xiaorong Ouyang
- The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, China
| | - Jinyang Liu
- The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, China
| | - Liye Jin
- The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, China
| | - Ahao Liu
- The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, China
| | - Binsheng He
- The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, China
| | - Tingting Fan
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Yuyang Jiang
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen 518055, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518132, China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
4
|
Wang H, Chi L, Yu F, Dai H, Si X, Gao C, Wang Z, Liu L, Zheng J, Ke Y, Liu H, Zhang Q. The overview of Mitogen-activated extracellular signal-regulated kinase (MEK)-based dual inhibitor in the treatment of cancers. Bioorg Med Chem 2022; 70:116922. [PMID: 35849914 DOI: 10.1016/j.bmc.2022.116922] [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: 05/24/2022] [Revised: 07/01/2022] [Accepted: 07/06/2022] [Indexed: 11/02/2022]
Abstract
Mitogen-activated extracellular signal-regulated kinase 1 and 2 (MEK1/2) are the critical components of the mitogen-activated protein kinase/extracellular signal-regulated kinase 1 and 2 (MAPK/ERK1/2) signaling pathway which is one of the well-characterized kinase cascades regulating cell proliferation, differentiation, growth, metabolism, survival and mobility both in normal and cancer cells. The aberrant activation of MAPK/ERK1/2 pathway is a hallmark of numerous human cancers, therefore targeting the components of this pathway to inhibit its dysregulation is a promising strategy for cancer treatment. Enormous efforts have been done in the development of MEK1/2 inhibitors and encouraging advancements have been made, including four inhibitors approved for clinical use. However, due to the multifactorial property of cancer and rapidly arising drug resistance, the clinical efficacy of these MEK1/2 inhibitors as monotherapy are far from ideal. Several alternative strategies have been developed to improve the limited clinical efficacy, including the dual inhibitor which is a single drug molecule able to simultaneously inhibit two targets. In this review, we first introduced the activation and function of the MAPK/ERK1/2 components and discussed the advantages of MEK1/2-based dual inhibitors compared with the single inhibitors and combination therapy in the treatment of cancers. Then, we overviewed the MEK1/2-based dual inhibitors for the treatment of cancers and highlighted the theoretical basis of concurrent inhibition of MEK1/2 and other targets for development of these dual inhibitors. Besides, the status and results of these dual inhibitors in both preclinical and clinical studies were also the focus of this review.
Collapse
Affiliation(s)
- Hao Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Lingling Chi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Fuqiang Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Hongling Dai
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Xiaojie Si
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Chao Gao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Zhengjie Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Limin Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Jiaxin Zheng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China
| | - Yu Ke
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China.
| | - Hongmin Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450052, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China.
| | - Qiurong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation of Henan Province, Zhengzhou 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China.
| |
Collapse
|
5
|
Van Dort ME, Jang Y, Bonham CA, Heist K, Palagama DSW, McDonald L, Zhang EZ, Chenevert TL, Luker GD, Ross BD. Structural effects of morpholine replacement in ZSTK474 on Class I PI3K isoform inhibition: Development of novel MEK/PI3K bifunctional inhibitors. Eur J Med Chem 2022; 229:113996. [PMID: 34802837 PMCID: PMC8792322 DOI: 10.1016/j.ejmech.2021.113996] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 02/07/2023]
Abstract
Established roles for PI3K and MAPK signaling pathways in tumorigenesis has prompted extensive research towards the discovery of small-molecule inhibitors as cancer therapeutics. However, significant compensatory regulation exists between these two signaling cascades, leading to redundancy among survival pathways. Consequently, initial clinical trials aimed at either PI3K or MEK inhibition alone have proven ineffective and highlight the need for development of targeted and innovative therapeutic combination strategies. We designed a series of PI3K inhibitor derivatives wherein a single morpholine group of the PI3K inhibitor ZSTK474 was substituted with a variety of 2-aminoethyl functional groups. Analogs with pendant hydroxyl or methoxy groups maintained low nanomolar inhibition towards PI3Kα, PI3Kγ, and PI3Kδ isoforms in contrast to those with pendant amino groups which were significantly less inhibitory. Synthesis of prototype PI3K/MEK bifunctional inhibitors (6r, 6s) was guided by the structure-activity data, where a MEK-targeting inhibitor was tethered directly via a short PEG linker to the triazine core of the PI3K inhibitor analogs. These compounds (6r, 6s) displayed nanomolar inhibition towards PI3Kα, δ, and MEK (IC50 ∼105-350 nM), and low micromolar inhibition for PI3Kβ and PI3Kγ (IC50 ∼1.5-3.9 μM) in enzymatic inhibition assays. Cell viability assays demonstrated superior anti-proliferative activity for 6s over 6r in three tumor-derived cell lines (A375, D54, SET-2), which correlated with inhibition of downstream AKT and ERK1/2 phosphorylation. Compounds 6r and 6s also demonstrated in vivo tolerability with therapeutic efficacy through reduction of kinase activation and amelioration of disease phenotypes in the JAK2V617F mutant myelofibrosis mouse cancer model. Taken together, these results support further structure optimization of 6r and 6s as promising leads for combination therapy in human cancer as a new class of PI3K/MEK bifunctional inhibitors.
Collapse
Affiliation(s)
- Marcian E Van Dort
- Center for Molecular Imaging, The University of Michigan Medical School, MI, 48109, USA; Department of Radiology, The University of Michigan Medical School, MI, 48109, USA.
| | - Youngsoon Jang
- Center for Molecular Imaging, The University of Michigan Medical School, MI, 48109, USA; Department of Radiology, The University of Michigan Medical School, MI, 48109, USA.
| | - Christopher A Bonham
- Center for Molecular Imaging, The University of Michigan Medical School, MI, 48109, USA; Department of Radiology, The University of Michigan Medical School, MI, 48109, USA.
| | - Kevin Heist
- Center for Molecular Imaging, The University of Michigan Medical School, MI, 48109, USA; Department of Radiology, The University of Michigan Medical School, MI, 48109, USA.
| | - Dilrukshika S W Palagama
- Center for Molecular Imaging, The University of Michigan Medical School, MI, 48109, USA; Department of Radiology, The University of Michigan Medical School, MI, 48109, USA.
| | - Lucas McDonald
- Center for Molecular Imaging, The University of Michigan Medical School, MI, 48109, USA; Department of Radiology, The University of Michigan Medical School, MI, 48109, USA.
| | - Edward Z Zhang
- Center for Molecular Imaging, The University of Michigan Medical School, MI, 48109, USA; Department of Radiology, The University of Michigan Medical School, MI, 48109, USA.
| | - Thomas L Chenevert
- Center for Molecular Imaging, The University of Michigan Medical School, MI, 48109, USA; Department of Radiology, The University of Michigan Medical School, MI, 48109, USA.
| | - Gary D Luker
- Center for Molecular Imaging, The University of Michigan Medical School, MI, 48109, USA; Department of Radiology, The University of Michigan Medical School, MI, 48109, USA.
| | - Brian D Ross
- Center for Molecular Imaging, The University of Michigan Medical School, MI, 48109, USA; Department of Radiology, The University of Michigan Medical School, MI, 48109, USA; Department of Biological Chemistry, The University of Michigan Medical School, MI, 48109, USA.
| |
Collapse
|
6
|
Gharat R, Prabhu A, Khambete MP. Potential of triazines in Alzheimer's disease: A versatile privileged scaffold. Arch Pharm (Weinheim) 2022; 355:e2100388. [DOI: 10.1002/ardp.202100388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/10/2022]
Affiliation(s)
- Ruchita Gharat
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy Mumbai Maharashtra India
| | - Arati Prabhu
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy Mumbai Maharashtra India
| | - Mihir. P. Khambete
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy Mumbai Maharashtra India
| |
Collapse
|
7
|
Guo H, Diao QP. 1,3,5-Triazine-azole Hybrids and their Anticancer Activity. Curr Top Med Chem 2020; 20:1481-1492. [DOI: 10.2174/1568026620666200310122741] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/12/2020] [Accepted: 02/17/2020] [Indexed: 12/24/2022]
Abstract
1,3,5-Triazine and azole can interact with various therapeutic targets, and their derivatives
possess promising in vitro and in vivo anticancer activity. Hybrid molecules have the potential to enhance
efficiency, overcome drug resistance and reduce side effects, and many hybrid molecules are under
different phases of clinical trials, so hybridization of 1,3,5-triazine with azole may provide valuable
therapeutic intervention for the treatment of cancer. Substantial efforts have been made to develop
azole-containing 1,3,5-triazine hybrids as novel anticancer agents, and some of them exhibited excellent
activity. This review emphasizes azole-containing 1,3,5-triazine hybrids with potential anticancer activity,
and the structure-activity relationships as well as the mechanisms of action are also discussed to
provide comprehensive and target-oriented information for the development of this kind of anticancer
drugs.
Collapse
Affiliation(s)
- Hua Guo
- School of Chemistry and Life Science, Anshan Normal University, Anshan, Liaoning, China
| | - Quan-Ping Diao
- School of Chemistry and Life Science, Anshan Normal University, Anshan, Liaoning, China
| |
Collapse
|
8
|
Li S, Xu G, Zhu Y, Zhao J, Gou S. Bifunctional ruthenium(ii) polypyridyl complexes of curcumin as potential anticancer agents. Dalton Trans 2020; 49:9454-9463. [PMID: 32598409 DOI: 10.1039/d0dt01040e] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ru(ii)-polypyridyl complexes have been widely studied and well established for their antitumor properties. Modifications of the coordination environment around the Ru atom through a proper choice of the ligand can lead to different modes of action and result in greatly improved anticancer efficacy. Herein, two Ru(ii)-polypyridyl complexes of curcumin were synthesized and characterized as potential anticancer agents. In vitro tests indicated that complexes 1 and 2 displayed excellent antiproliferative activity against the tested cancer cell lines, especially complex 2, which exhibited superior cytotoxicity compared to curcumin and cisplatin. Further biological evaluations demonstrated that complexes 1 and 2 can cause cell apoptosis via DNA interaction and MEK/ERK signaling pathway, which is the first example of a Ru(ii)-polypyridyl complex inhibiting the MEK/ERK signaling pathway and DNA intercalation. Overall, this work suggests that coordination with bioactive agents may endow Ru(ii)-polypyridyl complexes with improved pharmaceutical properties and synergistic effects for cancer therapy.
Collapse
Affiliation(s)
- Shuang Li
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research and Pharmaceutical Research Center, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, P. R. China.
| | | | | | | | | |
Collapse
|
9
|
Tao Q, Fang F, Li J, Wang Y, Zhao C, Liang J, Ma X, Wang H. A conjugated mTOR/MEK bifunctional inhibitor as potential polypharmacological anticancer agent: the prototype compound discovery. Med Chem Res 2020; 29:519-527. [PMID: 32435126 PMCID: PMC7223898 DOI: 10.1007/s00044-020-02502-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 01/06/2020] [Indexed: 02/07/2023]
Abstract
mTOR/MEK bifunctional inhibitors have the potential to surmount the drug resistance aroused from cross talk between PI3K/Akt/mTOR (PAM) and Ras/MEK/ERK pathways. Herein, we report the discovery of a conjugated dual-targeted molecule, compound 13, as the prototype mTOR/MEK bifunctional inhibitor. It exhibited moderately high inhibitory activity against mTOR and MEK1 with IC50 values of 0.19 μM and 0.98 μM, respectively. In particular, it displayed attractive antiproliferative activity against both A549 (GI50 = 4.66 μM) and HCT116 (GI50 = 5.47 μM) cell lines. To our knowledge, it has been the first example of a conjugated mTOR/MEK bifunctional inhibitor. In addition, from this proof-of-principle study, it has become evident that the single-agent dual inhibition of mTOR and MEK can be fulfilled via covalently attaching mTOR kinase inhibitor to an allosteric MEK inhibitor.
Collapse
Affiliation(s)
- Qiangqiang Tao
- 1School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012 China
| | - Fang Fang
- 1School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012 China.,Department of Medicinal Chemistry, Anhui Academy of Chinese Medicine, Hefei, 230012 China
| | - Jiaming Li
- 1School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012 China.,Department of Medicinal Chemistry, Anhui Academy of Chinese Medicine, Hefei, 230012 China
| | - Yong Wang
- 1School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012 China
| | - Can Zhao
- 1School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012 China
| | - Jingtai Liang
- 1School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012 China
| | - Xiaodong Ma
- 1School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012 China.,Department of Medicinal Chemistry, Anhui Academy of Chinese Medicine, Hefei, 230012 China
| | - Hao Wang
- 3Department of Clinical Laboratory, The First Affiliated Hospital of University of Science and Technology of China, Hefei, 230001 China
| |
Collapse
|
10
|
Jiang X, Yu J, Zhou Z, Kongsted J, Song Y, Pannecouque C, De Clercq E, Kang D, Poongavanam V, Liu X, Zhan P. Molecular design opportunities presented by solvent‐exposed regions of target proteins. Med Res Rev 2019; 39:2194-2238. [PMID: 31002405 DOI: 10.1002/med.21581] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 03/09/2019] [Accepted: 03/16/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Xiangyi Jiang
- Department of Medicinal ChemistryKey Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University Jinan Shandong People's Republic of China
| | - Ji Yu
- Department of Medicinal ChemistryKey Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University Jinan Shandong People's Republic of China
| | - Zhongxia Zhou
- Department of Medicinal ChemistryKey Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University Jinan Shandong People's Republic of China
| | - Jacob Kongsted
- Department of Physics, Chemistry and PharmacyUniversity of Southern Denmark Odense Denmark
| | - Yuning Song
- Department of Clinical PharmacyQilu Hospital of Shandong University Jinan China
| | - Christophe Pannecouque
- Rega Institute for Medical ResearchLaboratory of Virology and Chemotherapy Leuven Belgium
| | - Erik De Clercq
- Rega Institute for Medical ResearchLaboratory of Virology and Chemotherapy Leuven Belgium
| | - Dongwei Kang
- Department of Medicinal ChemistryKey Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University Jinan Shandong People's Republic of China
| | | | - Xinyong Liu
- Department of Medicinal ChemistryKey Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University Jinan Shandong People's Republic of China
| | - Peng Zhan
- Department of Medicinal ChemistryKey Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University Jinan Shandong People's Republic of China
| |
Collapse
|
11
|
Yu Z, Chen Z, Su Q, Ye S, Yuan H, Kuai M, Lv M, Tu Z, Yang X, Liu R, Hu G, Li Q. Dual inhibitors of RAF-MEK-ERK and PI3K-PDK1-AKT pathways: Design, synthesis and preliminary anticancer activity studies of 3-substituted-5-(phenylamino) indolone derivatives. Bioorg Med Chem 2019; 27:944-954. [PMID: 30777660 DOI: 10.1016/j.bmc.2019.01.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 01/21/2019] [Accepted: 01/23/2019] [Indexed: 12/31/2022]
Abstract
The dysfunction and mutual compensatory activation of RAF-MEK-ERK and PI3K-PDK1-AKT pathways have been demonstrated as the hallmarks in several primary and recurrent cancers. The strategy of concurrent blocking of these two pathways shows clinical merits on effective cancer therapy, such as combinatory treatments and dual-pathway inhibitors. Herein, we report a novel prototype of dual-pathway inhibitors by means of merging the core structural scaffolds of a MEK1 inhibitor and a PDK1 inhibitor. A library of 43 compounds that categorized into three series (Series I-III) was synthesized and tested for antitumor activity in lung cancer cells. The results from structure-activity relationship (SAR) analysis showed the following order of antitumor activity that 3-hydroxy-5-(phenylamino) indolone (Series III) > 3-alkenyl-5-(phenylamino) indolone (Series I) > 3-alkyl-5-(phenylamino) indolone (Series II). A lead compound 9za in Series III showed most potent antitumor activity with IC50 value of 1.8 ± 0.8 µM in A549 cells. Moreover, antitumor mechanism study demonstrated that 9za exerted significant apoptotic effect, and cellular signal pathway analysis revealed the potent blockage of phosphorylation levels of ERK and AKT in RAF-MEK-ERK and PI3K-PDK1-AKT pathways, respectively. The results reported here provide robust experimental basis for the discovery and optimization of dual pathway agents for anti-lung cancer therapy.
Collapse
Affiliation(s)
- Zutao Yu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, PR China
| | - Zhuo Chen
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, PR China
| | - Qiongli Su
- Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha 410013, Hunan, PR China
| | - Shiqi Ye
- School of Medicine, Shenzhen University, Shenzhen 518060, Guangdong, PR China
| | - Hongbo Yuan
- Hunan Qianjin Xiangjiang Pharmaceutical Co., Ltd, Changsha 410013, Hunan, PR China
| | - Mengni Kuai
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, PR China; Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha 410013, Hunan, PR China
| | - Meng Lv
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, PR China
| | - Zhijun Tu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, PR China
| | - Xiaoping Yang
- Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha 410013, Hunan, PR China
| | - RangRu Liu
- Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, PR China
| | - Gaoyun Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, PR China
| | - Qianbin Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, PR China.
| |
Collapse
|
12
|
Smith A, Pawar M, Van Dort ME, Galbán S, Welton AR, Thurber GM, Ross BD, Besirli CG. Ocular Toxicity Profile of ST-162 and ST-168 as Novel Bifunctional MEK/PI3K Inhibitors. J Ocul Pharmacol Ther 2018; 34:477-485. [PMID: 29708810 DOI: 10.1089/jop.2017.0126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE ST-162 and ST-168 are small-molecule bifunctional inhibitors of MEK and PI3K signaling pathways that are being developed as novel antitumor agents. Previous small-molecule and biologic MEK inhibitors demonstrated ocular toxicity events that were dose limiting in clinical studies. We evaluated in vitro and in vivo ocular toxicity profiles of ST-162 and ST-168. METHODS Photoreceptor cell line 661W and adult retinal pigment epithelium cell line ARPE-19 were treated with increasing concentrations of bifunctional inhibitors. Western blots, cell viability, and caspase activity assays were performed to evaluate MEK and PI3K inhibition and dose-dependent in vitro toxicity, and compared with monotherapy. In vivo toxicity profile was assessed by intravitreal injection of ST-162 and ST-168 in Dutch-Belted rabbits, followed by ocular examination and histological analysis of enucleated eyes. RESULTS Retinal cell lines treated with ST-162 or ST-168 exhibited dose-dependent inhibition of MEK and PI3K signaling. Compared with inhibition by monotherapies and their combinations, bifunctional inhibitors demonstrated reduced cell death and caspase activity. In vivo, both bifunctional inhibitors exhibited a more favorable toxicity profile when compared with MEK inhibitor PD0325901. CONCLUSIONS Novel MEK and PI3K bifunctional inhibitors ST-162 and ST-168 demonstrate favorable in vitro and in vivo ocular toxicity profiles, supporting their further development as potential therapeutic agents targeting multiple aggressive tumors.
Collapse
Affiliation(s)
- Andrew Smith
- 1 Department of Ophthalmology and Visual Sciences, University of Michigan , Medical School, Ann Arbor, Michigan
| | - Mercy Pawar
- 1 Department of Ophthalmology and Visual Sciences, University of Michigan , Medical School, Ann Arbor, Michigan
| | - Marcian E Van Dort
- 2 Department of Radiology and Biological Chemistry, Center for Molecular Imaging, University of Michigan , Medical School, Ann Arbor, Michigan.,3 Department of Radiology, University of Michigan , Medical School, Ann Arbor, Michigan
| | - Stefanie Galbán
- 2 Department of Radiology and Biological Chemistry, Center for Molecular Imaging, University of Michigan , Medical School, Ann Arbor, Michigan.,3 Department of Radiology, University of Michigan , Medical School, Ann Arbor, Michigan
| | - Amanda R Welton
- 2 Department of Radiology and Biological Chemistry, Center for Molecular Imaging, University of Michigan , Medical School, Ann Arbor, Michigan.,3 Department of Radiology, University of Michigan , Medical School, Ann Arbor, Michigan
| | - Greg M Thurber
- 4 Department of Chemical Engineering, University of Michigan , Medical School, Ann Arbor, Michigan
| | - Brian D Ross
- 2 Department of Radiology and Biological Chemistry, Center for Molecular Imaging, University of Michigan , Medical School, Ann Arbor, Michigan.,3 Department of Radiology, University of Michigan , Medical School, Ann Arbor, Michigan.,5 Department of Biological Chemistry, University of Michigan , Medical School, Ann Arbor, Michigan
| | - Cagri G Besirli
- 1 Department of Ophthalmology and Visual Sciences, University of Michigan , Medical School, Ann Arbor, Michigan
| |
Collapse
|