1
|
Hao S, Hou L, Wang JH, Yan JH, Niu YF, Cai ZH, Li F, Meng FH. Design, synthesis and biological evaluation of novel benzimidazole-derived p21-activited kinase 4 (PAK4) inhibitors bearing a 4-(4-methylpiperazin-1-yl)phenyl scaffold as potential antitumor agents. Eur J Med Chem 2024; 280:116971. [PMID: 39427518 DOI: 10.1016/j.ejmech.2024.116971] [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: 08/22/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024]
Abstract
A series of novel 6-(4-(4-methylpiperazin-1-yl)phenyl)-1H-benzo[d]imidazole-based p21-activited kinase 4 (PAK4) inhibitors were designed and synthesized based on the structure of lead compound GNE-2861 and that of anticancer inhibitors reported in our previous studies. All target compounds so designed were preliminarily screened in vitro for anti-tumor potency through kinase inhibitory assays and MTT assays, which revealed that most compounds exhibited significant inhibitory effects on PAK4 enzyme as well as prominent antiproliferative activities against four cancer cell models (A549, NCI-H1975, MDA-MB-231 and SK-BR-3) and low damage to healthy cells. In particular, the hit compound 12i was identified as the most effective and rather selective compound both at the enzyme and cellular level. Meanwhile, molecular docking and dynamic studies disclosed that compound 12i could bind to PAK4 stably via multiple interactions applied between the compound and the enzyme. Further mechanism studies indicated that compound 12i could inhibit the proliferation and suppress the migratory potential of MDA-MB-231 cells by inhibiting the phosphorylation of PAK4 and LIMK1, arresting cell cycle in the G0/G1 phase, inducing apoptosis and promoting ROS production. Notably, compound 12i could effectively inhibit triple-negative breast cancer (TNBC) growth with little toxicity in the MDA-MB-231 cell xenograft model. Taken together, in vitro and in vivo results demonstrated that compound 12i possessed high drug potential as an inhibitor of PAK4 to inhibit the growth and metastasis of TNBC.
Collapse
Affiliation(s)
- Shuang Hao
- School of Pharmacy, Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang 110122, PR China
| | - Liang Hou
- School of Pharmacy, Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang 110122, PR China
| | - Jia-Hui Wang
- School of Pharmacy, Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang 110122, PR China
| | - Jing-Han Yan
- School of Pharmacy, Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang 110122, PR China
| | - Yi-Fan Niu
- School of Pharmacy, Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang 110122, PR China
| | - Zheng-Hao Cai
- School of Pharmacy, Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang 110122, PR China
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Cell Biology of Ministry of Public Health, and Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang 110122, PR China.
| | - Fan-Hao Meng
- School of Pharmacy, Key Laboratory of Research and Development of Small Molecule Targeted Antitumor Drugs, China Medical University, Shenyang 110122, PR China.
| |
Collapse
|
2
|
Hao S, Wang JH, Hou L, Liang JW, Yan JH, Niu YF, Li XY, Sun Q, Meng FH. Design, synthesis and biological evaluation of novel quinazoline-derived EGFR/HER-2 dual-target inhibitors bearing a heterocyclic-containing tail as potential anti-tumor agents. Bioorg Chem 2024; 151:107686. [PMID: 39111120 DOI: 10.1016/j.bioorg.2024.107686] [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: 06/27/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 08/30/2024]
Abstract
A series of novel quinazoline-derived EGFR/HER-2 dual-target inhibitors were designed and synthesized by heterocyclic-containing tail approach. The inhibitory activities against four human epidermal growth factor receptor (HER) isozymes (EGFR, HER-2, HER-3 and HER-4) of all new compounds so designed were investigated in vitro. Compound 12k was found to be the most effective and rather selective dual-target inhibitor of EGFR and HER-2 with inhibitory constant (IC50) values of 6.15 and 9.78 nM, respectively, which was more potent than the clinical used agent Lapatinib (IC50 = 8.41 and 9.41 nM). Meanwhile, almost all compounds showed excellent antiproliferative activities against four cancer cell models (A549, NCI-H1975, SK-BR-3 and MCF-7) and low damage to healthy cells. Among them, compound 12k also exhibited the most prominent antitumor activity. Moreover, the hit compound 12k could bind to EGFR and HER-2 stably in molecular docking and dynamics studies. The following wound healing assay revealed that compound 12k could inhibit the migration of SK-BR-3 cells. Further studies found that compound 12k could arrest cell cycle in the G0/G1 phase and induce SK-BR-3 cells apoptosis. Notably, compound 12k could effectively inhibit breast cancer growth with little toxicity in the SK-BR-3 cell xenograft model. Taken together, in vitro and in vivo results disclosed that compound 12k had high drug potential as a dual-target inhibitor of EGFR/HER-2 to inhibit breast cancer growth.
Collapse
Affiliation(s)
- Shuang Hao
- School of Pharmacy, China Medical University, Shenyang 110122, PR China
| | - Jia-Hui Wang
- School of Pharmacy, China Medical University, Shenyang 110122, PR China
| | - Liang Hou
- School of Pharmacy, China Medical University, Shenyang 110122, PR China
| | - Jing-Wei Liang
- School of Pharmacy, China Medical University, Shenyang 110122, PR China; School of Pharmacy, Hainan Medical University, Haikou 571199, PR China
| | - Jing-Han Yan
- School of Pharmacy, China Medical University, Shenyang 110122, PR China
| | - Yi-Fan Niu
- School of Pharmacy, China Medical University, Shenyang 110122, PR China
| | - Xin-Yang Li
- School of Pharmacy, China Medical University, Shenyang 110122, PR China; Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Qi Sun
- School of Pharmacy, China Medical University, Shenyang 110122, PR China.
| | - Fan-Hao Meng
- School of Pharmacy, China Medical University, Shenyang 110122, PR China.
| |
Collapse
|
3
|
El-Fakharany ZS, Nissan YM, Sedky NK, Arafa RK, Abou-Seri SM. New proapoptotic chemotherapeutic agents based on the quinolone-3-carboxamide scaffold acting by VEGFR-2 inhibition. Sci Rep 2023; 13:11346. [PMID: 37443185 PMCID: PMC10444817 DOI: 10.1038/s41598-023-38264-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
In the current study, we designed and synthesized a series of new quinoline derivatives 10a-p as antiproliferative agents targeting cancer through inhibition of VEGFR-2. Preliminary molecular docking to assess the interactions of the designed derivatives with the binding site of VEGFR-2 (PDB code: 4ASD) displayed binding poses and interactions comparable to sorafenib. The synthesized compounds exhibited VEGFR-2 inhibitory activity with IC50 ranging from 36 nM to 2.23 μM compared to sorafenib (IC50 = 45 nM), where derivative 10i was the most potent. Additionally, the synthesized derivatives were evaluated in vitro for their cytotoxic activity against HepG2 cancer cell line. Seven compounds 10a, 10c, 10d, 10e, 10i, 10n and 10o (IC50 = 4.60, 4.14, 1.07, 0.88, 1.60, 2.88 and 2.76 μM respectively) displayed better antiproliferative activity than sorafenib (IC50 = 8.38 μM). Compound 10i was tested against Transformed Human Liver Epithelial-2 normal cell line (THLE-2) to evaluate its selective cytotoxicity. Furthermore, 10i, as a potent representative of the series, was assayed for its apoptotic activity and cell cycle kinetics' influence on HepG2, its effects on the gene expression of VEGFR-2, and protein expression of the apoptotic markers Caspase-7 and Bax. Compound 10i proved to have a potential role in apoptosis by causing significant increase in the early and late apoptotic quartiles, a remarkable activity in elevating the relative protein expression of Bax and Caspase-7 and a significant reduction of VEGFR-2 gene expression. Collectively, the obtained results indicate that compound 10i has a promising potential as a lead compound for the development of new anticancer agents.
Collapse
Affiliation(s)
- Zeinab S El-Fakharany
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, October University for Modern Sciences and Arts, Cairo, 12451, Egypt
| | - Yassin M Nissan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, October University for Modern Sciences and Arts, Cairo, 12451, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt
| | - Nada K Sedky
- Department of Biochemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted By Global Academic Foundation, New Administrative Capital, Cairo, Egypt
| | - Reem K Arafa
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, October Gardens, 6th of October City, Giza, 12578, Egypt.
- Drug Design and Discovery Lab, Zewail City of Science and Technology, Cairo, 12578, Egypt.
| | - Sahar M Abou-Seri
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, 11562, Egypt.
| |
Collapse
|
4
|
Nazreen S, Elbehairi SEI, Malebari AM, Alghamdi N, Alshehri RF, Shati AA, Ali NM, Alfaifi MY, Elhenawy AA, Alam MM. New Natural Eugenol Derivatives as Antiproliferative Agents: Synthesis, Biological Evaluation, and Computational Studies. ACS OMEGA 2023; 8:18811-18822. [PMID: 37273621 PMCID: PMC10233844 DOI: 10.1021/acsomega.3c00933] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/05/2023] [Indexed: 06/06/2023]
Abstract
Semisynthetic modifications of natural products have bestowed us with many anticancer drugs. In the present work, a natural product, eugenol, has been modified synthetically to generate new anticancer agents. The final compounds were structurally confirmed by NMR, IR, and mass techniques. From the cytotoxicity results, compound 17 bearing morpholine was found to be the most active cytotoxic agent with IC50 1.71 (MCF-7), 1.84 (SKOV3), and 1.1 μM (PC-3) and a thymidylate synthase (TS) inhibitor with an IC50 of 0.81 μM. Further cellular studies showed that compound 17 could induce apoptosis and arrest the cell cycle at the S phase in PC-3 carcinoma. The docking study strongly favors compound 17 to be a TS inhibitor as it displayed a similar interaction to 5-fluorouracil. The in silico pharmacokinetics and DFT computational studies support the results obtained from docking and biological evaluation and displayed favorable pharmacokinetic profile for a drug to be orally available. Compound 17 was found to be a promising TS inhibitor which could suppress DNA synthesis and consequently DNA damage in prostate cancer cells.
Collapse
Affiliation(s)
- Syed Nazreen
- Department
of Chemistry, Faculty of Science, Al-Baha
University, Al-Baha 65799, Kingdom of Saudi Arabia
| | - Serag Eldin I. Elbehairi
- Department
of Biology, Faculty of Science, King Khalid
University, Abha 9004, Saudi Arabia
- Cell
Culture Laboratory, Egyptian Organization for Biological Products
and Vaccines, VACSERA Holding Company, Giza 2311, Egypt
| | - Azizah M. Malebari
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Kingdom
of Saudi Arabia
| | - Nuha Alghamdi
- Department
of Chemistry, Faculty of Science, Al-Baha
University, Al-Baha 65799, Kingdom of Saudi Arabia
| | - Reem F. Alshehri
- Chemistry
Department, Faculty of Science and Art, Taibah University, Al Ula, Madinah 16857, Kingdom of Saudi Arabia
| | - Ali A. Shati
- Department
of Biology, Faculty of Science, King Khalid
University, Abha 9004, Saudi Arabia
| | - Nada M. Ali
- Department
of Chemistry, Faculty of Science, Al-Baha
University, Al-Baha 65799, Kingdom of Saudi Arabia
| | - Mohammad Y. Alfaifi
- Department
of Biology, Faculty of Science, King Khalid
University, Abha 9004, Saudi Arabia
| | - Ahmed A. Elhenawy
- Chemistry
Department, Faculty of Science, Al-Azhar
Unuversity, 11884 Nasr
City, Cairo 11751, Egypt
| | - Mohammad Mahboob Alam
- Department
of Chemistry, Faculty of Science, Al-Baha
University, Al-Baha 65799, Kingdom of Saudi Arabia
| |
Collapse
|
5
|
Lu G, Nie W, Xin M, Meng Y, Gu J, Miao H, Cheng X, Chan AS, Zou Y. Design, synthesis, biological evaluation and molecular docking study of novel urea-based benzamide derivatives as potent poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors. Eur J Med Chem 2022; 243:114790. [DOI: 10.1016/j.ejmech.2022.114790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/13/2022] [Accepted: 09/18/2022] [Indexed: 11/03/2022]
|
6
|
Almalki ASA, Nazreen S, Elbehairi SEI, Asad M, Shati AA, Alfaifi MY, Alhadhrami A, Elhenawy AA, Alorabi AQ, Asiri AM, Alam MM. Design, synthesis, anticancer activity and molecular docking studies of new benzimidazole derivatives bearing 1,3,4-oxadiazole moieties as potential thymidylate synthase inhibitors. NEW J CHEM 2022. [DOI: 10.1039/d2nj01980a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Compounds 10 and 14 arrest the cell cycle at the G1 phase and induce apoptosis without any necrosis in MDA-MB-231 cells.
Collapse
Affiliation(s)
- Abdulraheem SA Almalki
- Department of Chemistry, Faculty of Science, Taif University, Taif, Kingdom of Saudi Arabia
| | - Syed Nazreen
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
| | - Serag Eldin I. Elbehairi
- Department of Biology, Faculty of Science, King Khalid University, Abha 9004, Saudi Arabia
- Cell Culture Laboratory, Egyptian Organization for Biological Products and Vaccines, VACSERA Holding Company, Giza 2311, Egypt
| | - Mohammad Asad
- Center of Excellence for Advanced Materials Research (CEAMR), King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
- Chemistry Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
| | - Ali A. Shati
- Department of Biology, Faculty of Science, King Khalid University, Abha 9004, Saudi Arabia
| | - Mohammad Y. Alfaifi
- Department of Biology, Faculty of Science, King Khalid University, Abha 9004, Saudi Arabia
| | - Abdulrahman Alhadhrami
- Department of Chemistry, Faculty of Science, Taif University, Taif, Kingdom of Saudi Arabia
| | - Ahmed A. Elhenawy
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
- Chemistry Department, Faculty of Science, Al-Azhar University, 11884 Nasr City, Cairo, Egypt
| | - Ali Q. Alorabi
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
| | - Abdullah M. Asiri
- Center of Excellence for Advanced Materials Research (CEAMR), King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
- Chemistry Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia
| | - Mohammad Mahboob Alam
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
| |
Collapse
|
7
|
Yang L, Hou X, Zhang Y, Wang D, Liu J, Huang F, Liu J. NIR-activated self-sensitized polymeric micelles for enhanced cancer chemo-photothermal therapy. J Control Release 2021; 339:114-129. [PMID: 34536448 DOI: 10.1016/j.jconrel.2021.09.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 01/20/2023]
Abstract
NIR-activated therapies based on light-responsive drug delivery systems are emerging as a remote-controlled method for cancer precise therapy. In this work, fluorescent dye indocyanine green (ICG)-conjugated and bioactive compound gambogic acid (GA)-loaded polymeric micelles (GA@PEG-TK-ICG PMs) were smoothly fabricated via the self-assembly of the reactive oxygen species (ROS)-responsive thioketal (TK)-linked amphiphilic polymer poly(ethyleneglycol)-thioketal-(indocyanine green) (PEG-TK-ICG). The resultant micelles demonstrated increased resistance to photobleaching, enhanced photothermal conversion efficiency, NIR-controlled drug release behavior, preferable biocompatibility, and excellent tumor accumulation performance. Moreover, upon an 808 nm laser irradiation, the micellar photoactive chromophore ICG converted the absorbed optical energy to both hyperthermia for photothermal therapy (PTT) and ROS as the feedback trigger to the micelles for the tumor-specific release of GA, which could serve as not only a chemotherapeutic drug to directly kill tumor cells but also a heat shock protein 90 (HSP90) inhibitor to realize the photothermal sensitization. As a result, an extremely high tumor inhibition rate (97.9%) of mouse 4 T1 breast cancer models was achieved with negligible side effects after the chemo-photothermal synergistic therapy. This NIR-activated nanosystem with photothermal self-sensitization function may provide a feasible option for the effective treatment of aggressive breast cancers.
Collapse
Affiliation(s)
- Lijun Yang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Xiaoxue Hou
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Yumin Zhang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Dianyu Wang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Jinjian Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| | - Fan Huang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| |
Collapse
|
8
|
Li XY, Wang DP, Li S, Xue WH, Qian XH, Liu KL, Li YH, Lin QQ, Dong G, Meng FH, Jian LY. Discovery of N-(1,3,4-thiadiazol-2-yl)benzamide derivatives containing a 6,7-methoxyquinoline structure as novel EGFR/HER-2 dual-target inhibitors against cancer growth and angiogenesis. Bioorg Chem 2021; 119:105469. [PMID: 34915285 DOI: 10.1016/j.bioorg.2021.105469] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/26/2021] [Accepted: 11/02/2021] [Indexed: 12/27/2022]
Abstract
Targeting EGFR and HER-2 is an essential direction for cancer treatment. Here, a series of N-(1,3,4-thiadiazol-2-yl)benzamide derivatives containing a 6,7-methoxyquinoline structure was designed and synthesized to serve as EGFR/HER-2 dual-target inhibitors. The kinase assays verified that target compounds could inhibit the kinase activity of EGFR and HER-2 selectively. The results of CCK-8 and 3D cell viability assays confirmed that target compounds had excellent anti-proliferation ability against breast cancer cells (MCF-7 and SK-BR-3) and lung cancer cells (A549 and H1975), particularly against SK-BR-3 cells, while the inhibitory effect on healthy breast cells (MCF-10A) and lung cells (Beas-2B) was weak. Among them, the hit compound YH-9 binded to EGFR and HER-2 stably in molecular dynamics studies. Further studies found thatYH-9could induce the release of cytochrome c and inhibit proliferation by promoting ROS expression in SK-BR-3 cells. Moreover,YH-9could diminish the secretion of VEGF and bFGF factors in SK-BR-3 cells, then inhibited tube formation and angiogenesis. Notably,YH-9could effectively inhibit breast cancer growth and angiogenesis with little toxicity in the SK-BR-3 cell xenograft model. Taken together,in vitroandin vivoresults revealed that YH-9 had high drug potential as a dual-target inhibitor of EGFR/HER-2 to inhibit breast cancer growth and angiogenesis.
Collapse
Affiliation(s)
- Xin-Yang Li
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang 110004, PR China; School of Pharmacy, China Medical University, Shenyang 110122, PR China
| | - De-Pu Wang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang 110004, PR China; School of Pharmacy, China Medical University, Shenyang 110122, PR China
| | - Shuai Li
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang 110004, PR China; School of Pharmacy, China Medical University, Shenyang 110122, PR China
| | - Wen-Han Xue
- School of Pharmacy, China Medical University, Shenyang 110122, PR China
| | - Xin-Hua Qian
- School of Pharmacy, China Medical University, Shenyang 110122, PR China
| | - Kai-Li Liu
- School of Pharmacy, China Medical University, Shenyang 110122, PR China
| | - Yu-Heng Li
- School of Pharmacy, China Medical University, Shenyang 110122, PR China
| | - Qi-Qi Lin
- School of Pharmacy, China Medical University, Shenyang 110122, PR China
| | - Gang Dong
- School of Pharmacy, China Medical University, Shenyang 110122, PR China
| | - Fan-Hao Meng
- School of Pharmacy, China Medical University, Shenyang 110122, PR China
| | - Ling-Yan Jian
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| |
Collapse
|
9
|
Zuccolo M, Arrighetti N, Perego P, Colombo D. Recent Progresses in Conjugation with Bioactive Ligands to Improve the Anticancer Activity of Platinum Compounds. Curr Med Chem 2021; 29:2566-2601. [PMID: 34365939 DOI: 10.2174/0929867328666210806110857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/09/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
Platinum (Pt) drugs, including cisplatin, are widely used for the treatment of solid tumors. Despite the clinical success, side effects and occurrence of resistance represent major limitations to the use of clinically available Pt drugs. To overcome these problems, a variety of derivatives have been designed and synthetized. Here, we summarize the recent progress in the development of Pt(II) and Pt(IV) complexes with bioactive ligands. The development of Pt(II) and Pt(IV) complexes with targeting molecules, clinically available agents, and other bioactive molecules is an active field of research. Even if none of the reported Pt derivatives has been yet approved for clinical use, many of these compounds exhibit promising anticancer activities with an improved pharmacological profile. Thus, planning hybrid compounds can be considered as a promising approach to improve the available Pt-based anticancer agents and to obtain new molecular tools to deepen the knowledge of cancer progression and drug resistance mechanisms.
Collapse
Affiliation(s)
- Marco Zuccolo
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan. Italy
| | - Noemi Arrighetti
- Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan. Italy
| | - Paola Perego
- Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan. Italy
| | - Diego Colombo
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Milan. Italy
| |
Collapse
|
10
|
Wang DP, Liu KL, Li XY, Lu GQ, Xue WH, Qian XH, Mohamed O K, Meng FH. Design, synthesis, and in vitro and in vivo anti-angiogenesis study of a novel vascular endothelial growth factor receptor-2 (VEGFR-2) inhibitor based on 1,2,3-triazole scaffold. Eur J Med Chem 2020; 211:113083. [PMID: 33340911 DOI: 10.1016/j.ejmech.2020.113083] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/01/2020] [Accepted: 12/01/2020] [Indexed: 01/08/2023]
Abstract
In the past five years, our team had been committed to click chemistry research, exploring the biological activity of 1,2,3-triazole by synthesizing different target inhibitors. In this study, a series of novel indole-2-one derivatives based on 1,2,3-triazole scaffolds were synthesized for the first time, and their inhibitory activity on vascular endothelial growth factor receptor-2 (VEGFR-2) was tested. Most of the compounds had shown promising activity in the VEGFR-2 kinase assay and had low toxicity to human umbilical vein endothelial cells (HUVECs). The compound 13d (IC50 = 26.38 nM) had better kinase activity inhibition ability than sunitinib (IC50 = 83.20 nM) and was less toxic to HUVECs. Moreover, it had an excellent inhibitory effect on HT-29 and MKN-45 cells. On the one hand, by tube formation assay, transwell, and Western blot analysis, compound 13d could inhibit VEGFR-2 protein phosphorylate on HUVECs, thereby inhibiting HUVECs migration and tube formation. In vivo study, the zebrafish model with VEGFR-2 labeling also verified that compound 13d had more anti-angiogenesis ability than sunitinib. On the other hand, molecular docking and molecular dynamics (MD) simulation results showed that compound 13d could stably bind to the active site of VEGFR-2. Based on the above findings, compound 13d could be considered an effective anti-angiogenesis drug and has more development value than sunitinib.
Collapse
Affiliation(s)
- De-Pu Wang
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Kai-Li Liu
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Xin-Yang Li
- School of Pharmacy, China Medical University, Shenyang, 110122, China; Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Guo-Qing Lu
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Wen-Han Xue
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Xin-Hua Qian
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Kamara Mohamed O
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Fan-Hao Meng
- School of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
11
|
Li XY, Wang DP, Lu GQ, Liu KL, Zhang TJ, Li S, Mohamed O K, Xue WH, Qian XH, Meng FH. Development of a novel thymidylate synthase (TS) inhibitor capable of up-regulating P53 expression and inhibiting angiogenesis in NSCLC. J Adv Res 2020; 26:95-110. [PMID: 33133686 PMCID: PMC7584679 DOI: 10.1016/j.jare.2020.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/16/2020] [Accepted: 07/16/2020] [Indexed: 11/24/2022] Open
Abstract
Introduction The development of a new type of Thymidylate synthase (TS) inhibitor that could inhibit cancer cells' proliferation and anti-angiogenesis is of great significance for cancer's clinical treatment. Objectives Our research hopes to develop a TS inhibitor that is more effective than the current first-line clinical treatment of pemetrexed (PTX) and provide a new reference for the clinical treatment of non-small cell lung cancer (NSCLC). Methods We obtained a series of novel TS inhibitors by chemical synthesis. Moreover, TS assay and molecular docking to verify the target compound's inhibitory mode. Use MTT assay, colony-forming assay, flow cytometry, and western blot to verify the compound's inhibitory effect on cancer cell proliferation and its mechanism; and explore the compound’s effect on angiogenesis in vitro and in vivo. Further, explore the hit compound's anti-cancer ability through the xenograft tumor model and the orthotopic cancer murine model. Results A series of N-(3-(5-phenyl-1,3,4-oxadiazole-2-yl) phenyl)-2,4-dihydroxypyrimidine-5-sulfamide derivatives were synthesized as TS inhibitors for the first time. All target compounds significantly inhibited hTS enzyme activity and demonstrated significant antitumor activity against five cancer cell lines. Notably, 7f had a high selectivity index (SI) and unique inhibitory effects on eight NSCLC cells. In-depth research indicated that 7f could induce apoptosis by the mitochondrial pathway in A549 and PC-9 cells through the upregulation of wild-type P53 protein expression. Additionally, 7f was shown to inhibit angiogenesis in vitro and in vivo. In vivo studies, compared to PTX, 7f significantly inhibited tumor growth in A549 cell xenografts and had a higher therapeutic index (TGI). Moreover, 7f could prolong the survival of the orthotopic lung cancer murine model more effectively than PTX. Conclusion The anti-angiogenic effect of 7f provides a new reference for the development of TS inhibitors and the clinical treatment of NSCLC.
Collapse
Affiliation(s)
- Xin-Yang Li
- School of Pharmacy, China Medical University, Shenyang 110122, China.,Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang 110122, China
| | - De-Pu Wang
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Guo-Qing Lu
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Kai-Li Liu
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Ting-Jian Zhang
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Shuai Li
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Kamara Mohamed O
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Wen-Han Xue
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Xin-Hua Qian
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Fan-Hao Meng
- School of Pharmacy, China Medical University, Shenyang 110122, China
| |
Collapse
|