1
|
Mehra A, Sangwan R. A Promising Paradigm Shift in Cancer Treatment with FGFR Inhibitors. Anticancer Agents Med Chem 2025; 25:2-23. [PMID: 39192641 DOI: 10.2174/0118715206318833240819031953] [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: 04/18/2024] [Revised: 07/04/2024] [Accepted: 07/12/2024] [Indexed: 08/29/2024]
Abstract
FGFR have been demonstrated to perform a crucial role in biological processes but their overexpression has been perceived as the operator component in the occurrence and progression of different types of carcinoma. Out of all the interest around cancer, FGFR inhibitors have assembled pace over the past few years. Therefore, FGFR inhibitors are one of the main fundamental tools to reverse drug resistance, tumor growth, and angiogenesis. Currently, many FGFR inhibitors are under the development stage or have been developed. Due to great demand and hotspots, different pharmacophores were approached to access structurally diverse FGFR inhibitors. Here, we have selected to present several representative examples such as Naphthyl, Pyrimidine, Pyridazine, Indole, and Quinoline derivatives that illustrate the diversity and advances of FGFR inhibitors in medicinal chemistry. This review focuses on the SAR study of FGFR inhibitors last five years which will be a great future scope that influences the medicinal chemist to work towards more achievements in this area.
Collapse
Affiliation(s)
- Anuradha Mehra
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road (NH-1), Phagwara (Punjab), 144411, India
| | - Rekha Sangwan
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road (NH-1), Phagwara (Punjab), 144411, India
| |
Collapse
|
2
|
Gan X, Wu Y, Zhu M, Liu B, Kong M, Xi Z, Li K, Wang H, Su T, Yao J, Khushafah F, Yi B, Wang J, Li W, Wu J. Design, synthesis, and evaluation of cyclic C7-bridged monocarbonyl curcumin analogs containing an o-methoxy phenyl group as potential agents against gastric cancer. J Enzyme Inhib Med Chem 2024; 39:2314233. [PMID: 38385332 PMCID: PMC10885745 DOI: 10.1080/14756366.2024.2314233] [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: 08/01/2023] [Accepted: 01/02/2024] [Indexed: 02/23/2024] Open
Abstract
The structure-activity relationship (SAR) between toxicity and the types of linking ketones of C7 bridged monocarbonyl curcumin analogs (MCAs) was not clear yet. In the pursuit of effective and less cytotoxic chemotherapeutics, we conducted a SAR analysis using various diketene skeletons of C7-bridged MCAs, synthesized cyclic C7-bridged MCAs containing the identified low-toxicity cyclopentanone scaffold and an o-methoxy phenyl group, and assessed their anti-gastric cancer activity and safety profile. Most compounds exhibited potent cytotoxic activities against gastric cancer cells. We developed a quantitative structure-activity relationship model (R2 > 0.82) by random Forest method, providing important information for optimizing structure. An optimized compound 2 exhibited in vitro and in vivo anti-gastric cancer activity partly through inhibiting the AKT and STAT3 pathways, and displayed a favorable in vivo safety profile. In summary, this paper provided a promising class of MCAs and a potential compound for the development of chemotherapeutic drugs.
Collapse
Affiliation(s)
- Xin Gan
- The Second Affiliated Hospital and Yuying Children's Hospital of the Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yuna Wu
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, China
| | - Min Zhu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Bo Liu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Miaomiao Kong
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zixuan Xi
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ke Li
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haibao Wang
- Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Tiande Su
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiali Yao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Fatehi Khushafah
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Baozhu Yi
- The Second Affiliated Hospital and Yuying Children's Hospital of the Wenzhou Medical University, Wenzhou, China
| | - Jiabing Wang
- Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Wulan Li
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianzhang Wu
- The Second Affiliated Hospital and Yuying Children's Hospital of the Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
3
|
Wen J, Zhao L, Li Z, Pi C, Feng X, Shi P, Yang H, Chen L, Wang X, Liu F, Wei Y, Zhao L. Preparation and anti-colon cancer effect of a novel curcumin analogue (CA8): in vivo and in vitro evaluation. Front Pharmacol 2024; 15:1464626. [PMID: 39600365 PMCID: PMC11589483 DOI: 10.3389/fphar.2024.1464626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Chemotherapy remains the first choice of treatment for colon cancer despite the inevitable adverse effects. Curcumin (CU) possesses antitumor activity but has poor aqueous solubility, low bioavailability, and weak activity. To address this, nine novel monocarbonyl CU analogues were designed, synthesized, and evaluated in the present study. Among them, CA8 exhibited the highest water solubility, which was approximately 2.37 × 106 times that of CU. In addition, compared with CU, its cytotoxicity on Caco-2 cells (19.2 times/48 h) was stronger. Of note, CA8 arrestedthe cell cycle of Caco-2 cells at the G2/M phase and induced apoptosis. Meanwhile, acute toxicity experiments indicated that KM mice tolerated CA8 for up to 300 mg/kg CA8 (oral administration) and 50 mg/kg CA8 (intraperitoneal injection). The oral administration of CA8 to Sprague Dawley rats exhibited higher AUC (0-t) (6.23-fold) and longer MRT (0-t) (3.35-fold) than that of CU. CA8 also inhibited the proliferation and angiogenesis of tumor cells more than CU and tegafur. Finally, CA8 may exert anti-tumor effects through the activation of JNK pathway and inhibition of AKT pathway. These results suggest that CA8 is a safe and highly effective new drug for colon cancer treatment.
Collapse
Affiliation(s)
- Jie Wen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Lingmao Zhao
- Luzhou Longmatan District People’s Hospital, Luzhou Third People’s Hospital, Luzhou, Sichuan, China
| | - Zhuohan Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Chao Pi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xianhu Feng
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, China
- Nanchong Key Laboratory of Individualized Drug Therapy, Department of Pharmacy, Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Peng Shi
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Hongru Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaodong Wang
- Department of Hepatobiliary Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Furong Liu
- Department of Oncology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Yumeng Wei
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Ling Zhao
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
4
|
Chen X, Zhang P, Zhang H, Ma X, Zhang Y, Wu Y, Jin K, Wang J, Wu J. Discovery of cinnamylaldehyde-derived mono-carbonyl curcumin analogs as anti-gastric cancer agents via suppression of STAT3 and AKT pathway. Bioorg Chem 2024; 146:107306. [PMID: 38531150 DOI: 10.1016/j.bioorg.2024.107306] [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: 01/10/2024] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 03/28/2024]
Abstract
The structural modification of curcumin has always been a hotspot in drug development. In this paper, a class of cinnamylaldehyde-derived mono-carbonyl curcumin analogs (MCAs) with 7-carbon-links were designed and synthesized and their anticancer properties were evaluated. Through screening anti-gastric cancer activity of these compounds, H1 exhibited the strongest cytotoxic activity by inhibiting cell viability and colony formation, inducing cell cycle G2/M phase arrest in vitro (SGC-7901 and AGS gastric cancer cells). Moreover, the SGC-7901 subcutaneous tumor-bearing mice studies revealed that H1 significantly inhibited the tumor growth of gastric cancer. We explored the possible potential targets of H1 through network pharmacology. Mechanistically, our results demonstrated that H1 showed potential anti-gastric cancer activity through suppression of the STAT3 and AKT signaling pathway in vitro and in vivo, which was validated by molecular docking. Overall, our results indicate the potential of H1 as a potent chemotherapeutic drug against gastric cancer.
Collapse
Affiliation(s)
- Xi Chen
- School of Medicine, Taizhou University, Taizhou Zhejiang, 318000, China
| | - Peiqin Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou Zhejiang, 325000, China
| | - Huating Zhang
- School of Medicine, Taizhou University, Taizhou Zhejiang, 318000, China
| | - Xueqiang Ma
- Municipal Hospital Affiliated to Taizhou University, Taizhou 318000, Zhejiang, China
| | - Ye Zhang
- School of Medicine, Taizhou University, Taizhou Zhejiang, 318000, China
| | - Yajie Wu
- School of Medicine, Taizhou University, Taizhou Zhejiang, 318000, China
| | - Kaiwen Jin
- School of Medicine, Taizhou University, Taizhou Zhejiang, 318000, China
| | - Jiabing Wang
- Municipal Hospital Affiliated to Taizhou University, Taizhou 318000, Zhejiang, China.
| | - Jianzhang Wu
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, 325000, Zhejiang, China
| |
Collapse
|
5
|
Nian C, Gan X, Liu Q, Wu Y, Kong M, Zhang P, Jin M, Dong Z, Li W, Wang L, He W, Li X, Wu J. Synthesis and Anti-gastric Cancer Activity by Targeting FGFR1 Pathway of Novel Asymmetric Bis-chalcone Compounds. Curr Med Chem 2024; 31:6521-6541. [PMID: 38847254 DOI: 10.2174/0109298673298420240530093525] [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: 12/15/2023] [Revised: 04/03/2024] [Accepted: 04/19/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND Bis-chalcone compounds with symmetrical structures, either isolated from natural products or chemically synthesized, have multiple pharmacological activities. Asymmetric Bis-chalcone compounds have not been reported before, which might be attributed to the synthetic challenges involved, and it remains unknown whether these compounds possess any potential pharmacological activities. AIMS The aim of this study is to investigate the synthesis route of asymmetric bis-chalcone compounds and identify potential candidates with efficient anti-tumor activity. METHODS The two-step structural optimization of the bis-chalcone compounds was carried out sequentially, guided by the screening of the compounds for their growth inhibitory activity against gastric cancer cells by MTT assay. The QSAR model of compounds was established through random forest (RF) algorithm. The activities of the optimal compound J3 on growth inhibition, apoptosis, and apoptosis-inducing protein expression in gastric cancer cells were investigated sequentially by colony formation assay, flow cytometry, and western blotting. Further, the inhibitory effects of J3 on the FGFR1 signaling pathway were explored by Western Blotting, shRNA, and MTT assays. Finally, the in vivo anti-tumor activity and mechanism of J3 were studied through nude mice xenograft assay, western blotting. RESULTS 27 asymmetric bis-chalcone compounds, including two types (N and J) were sequentially designed and synthesized. Some N-class compounds have good inhibitory activity on the growth of gastric cancer cells. The vast majority of J-class compounds optimized on the basis of N3 exhibit excellent inhibitory activity on gastric cancer cell growth. We established a QSAR model (R2 = 0.851627) by applying random forest algorithms. The optimal compound J3, which had better activity, concentration-dependently inhibited the formation of gastric cancer cell colonies and led to cell apoptosis by inducing the expression of the pro-apoptotic protein cleaved PARP in a dose-dependent manner. J3 may exert anti-gastric cancer effects by inhibiting the activation of FGFR1/ERK pathway. Moreover, at a dose of 10 mg/kg/day, J3 inhibited tumor growth in nude mice by nearly 70% in vivo with no significant toxic effect on body weight and organs. CONCLUSION In summary, this study outlines a viable method for the synthesis of novel asymmetric bischalcone compounds. Furthermore, the compound J3 demonstrates substantial promise as a potential candidate for an anti-tumor drug.
Collapse
Affiliation(s)
- Chunhui Nian
- The Second Affiliated Hospital and Yuying Children's Hospital of the Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xin Gan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Department of Pharmacy, Ezhou Central Hospital, Ezhou, Hubei, 436000, China
| | - Qunpeng Liu
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, Zhejiang, 325035, China
| | - Yuna Wu
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University; Wenzhou, 325027, China
| | - Miaomiao Kong
- The 1st affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Peiqin Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Mingming Jin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Zhaojun Dong
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wulan Li
- The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Ledan Wang
- The Second Affiliated Hospital and Yuying Children's Hospital of the Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Wenfei He
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, Zhejiang, 325035, China
| | - Jianzhang Wu
- The Second Affiliated Hospital and Yuying Children's Hospital of the Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University; Wenzhou, 325027, China
| |
Collapse
|
6
|
Lu T, Li T, Wu MK, Zheng CC, He XM, Zhu HL, Li L, Man RJ. Molecular simulations required to target novel and potent inhibitors of cancer invasion. Expert Opin Drug Discov 2023; 18:1367-1377. [PMID: 37676052 DOI: 10.1080/17460441.2023.2254695] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 08/30/2023] [Indexed: 09/08/2023]
Abstract
INTRODUCTION Computer-aided drug design (CADD) is a computational approach used to discover, develop, and analyze drugs and active molecules with similar biochemical properties. Molecular simulation technology has significantly accelerated drug research and reduced manufacturing costs. It is an optimized drug discovery method that greatly improves the efficiency of novel drug development processes. AREASCOVERED This review discusses the development of molecular simulations of effective cancer inhibitors and traces the main outcomes of in silico studies by introducing representative categories of six important anticancer targets. The authors provide views on this topic from the perspective of both medicinal chemistry and artificial intelligence, indicating the major challenges and predicting trends. EXPERT OPINION The goal of introducing CADD into cancer treatment is to realize a highly efficient, accurate, and desired approach with a high success rate for identifying potent drug candidates. However, the major challenge is the lack of a sophisticated data-filtering mechanism to verify bottom data from mixed-quality references. Consequently, despite the continuous development of algorithms, computer power, and interface optimization, specific data filtering mechanisms will become an urgent and crucial issue in the future.
Collapse
Affiliation(s)
| | - Tong Li
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi University for Nationalities, Nanning, China
| | - Meng-Ke Wu
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi University for Nationalities, Nanning, China
| | - Chi-Chong Zheng
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi University for Nationalities, Nanning, China
| | - Xue-Mei He
- Agro-food Science and Technology Research Institute, Guangxi Academy of Agricultural Science, Nanning, China
| | - Hai-Liang Zhu
- School of Life Sciences, Nanjing University, Nanjing, China
| | - Li Li
- Agro-food Science and Technology Research Institute, Guangxi Academy of Agricultural Science, Nanning, China
| | - Ruo-Jun Man
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi University for Nationalities, Nanning, China
| |
Collapse
|
7
|
Zahran SS, Ragab FA, El-Gazzar MG, Soliman AM, Mahmoud WR, Ghorab MM. Antiproliferative, antiangiogenic and apoptotic effect of new hybrids of quinazoline-4(3H)-ones and sulfachloropyridazine. Eur J Med Chem 2022; 245:114912. [DOI: 10.1016/j.ejmech.2022.114912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/26/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022]
|
8
|
Kumar V, Bala R, Dhawan S, Singh P, Karpoormath R. The Multi‐Biological Targeted Role of Dehydrozingerone and its Analogues. ChemistrySelect 2022. [DOI: 10.1002/slct.202201938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Vishal Kumar
- Department of Pharmaceutical Chemistry Discipline of Pharmaceutical Sciences College of Health Sciences University of KwaZulu-Natal (Westville Campus) Durban 4000 South Africa
| | - Renu Bala
- Department of Pharmaceutical Chemistry Discipline of Pharmaceutical Sciences College of Health Sciences University of KwaZulu-Natal (Westville Campus) Durban 4000 South Africa
| | - Sanjeev Dhawan
- Department of Pharmaceutical Chemistry Discipline of Pharmaceutical Sciences College of Health Sciences University of KwaZulu-Natal (Westville Campus) Durban 4000 South Africa
| | - Parvesh Singh
- School of Chemistry and Physics University of KwaZulu-Natal (Westville campus) Private Bag X01, Scottsville Durban South Africa
| | - Rajshekhar Karpoormath
- Department of Pharmaceutical Chemistry Discipline of Pharmaceutical Sciences College of Health Sciences University of KwaZulu-Natal (Westville Campus) Durban 4000 South Africa
| |
Collapse
|
9
|
Chen J, Wen K, Wu Y, Deng J, Chen H, Yao X, Tang X. Synthesis of 3,4,5‐Triarylcyclohexanones from Dienones and 2‐Methylquinolines Based on a [5+1] Annulation. ChemistrySelect 2021. [DOI: 10.1002/slct.202103317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Jiewen Chen
- Guangdong Provincial Key Laboratory of New Drug Screening School of Pharmaceutical Sciences Southern Medical University 1023 South Shatai Road, Baiyun District Guangzhou 510515 P. R. China
| | - Kangmei Wen
- Guangdong Provincial Key Laboratory of New Drug Screening School of Pharmaceutical Sciences Southern Medical University 1023 South Shatai Road, Baiyun District Guangzhou 510515 P. R. China
| | - Yinrong Wu
- Guangdong Provincial Key Laboratory of New Drug Screening School of Pharmaceutical Sciences Southern Medical University 1023 South Shatai Road, Baiyun District Guangzhou 510515 P. R. China
| | - Jie Deng
- Guangdong Provincial Key Laboratory of New Drug Screening School of Pharmaceutical Sciences Southern Medical University 1023 South Shatai Road, Baiyun District Guangzhou 510515 P. R. China
| | - Hongyue Chen
- Guangdong Provincial Key Laboratory of New Drug Screening School of Pharmaceutical Sciences Southern Medical University 1023 South Shatai Road, Baiyun District Guangzhou 510515 P. R. China
| | - Xingang Yao
- Guangdong Provincial Key Laboratory of New Drug Screening School of Pharmaceutical Sciences Southern Medical University 1023 South Shatai Road, Baiyun District Guangzhou 510515 P. R. China
| | - Xiaodong Tang
- Guangdong Provincial Key Laboratory of New Drug Screening School of Pharmaceutical Sciences Southern Medical University 1023 South Shatai Road, Baiyun District Guangzhou 510515 P. R. China
| |
Collapse
|
10
|
Zhao Y, Zheng Z, Zhang M, Wang Y, Hu R, Lin W, Huang C, Xu C, Wu J, Deng H. Design, synthesis, and evaluation of mono-carbonyl analogues of curcumin (MCACs) as potential antioxidants against periodontitis. J Periodontal Res 2021; 56:656-666. [PMID: 33604902 DOI: 10.1111/jre.12862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/23/2020] [Accepted: 01/28/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND OBJECTIVE The application of curcumin is limited by its instability. Mono-carbonyl analogues of curcumin (MCACs) are structurally stable, yet the intermediate bridging ketones in their skeletons account for increased toxicity. This study aimed to synthesize and screen MCACs that exhibit low cytotoxicity and high antioxidant ability, and the effects of MCACs on experimental periodontitis were also investigated. MATERIALS AND METHODS The cytotoxicity of MCACs on MC3 T3-E1 was determined by MTT assay. The antioxidant capacity was investigated by the cell viability against H2 O2 -induced damage and the level of reactive oxygen species (ROS) and malondialdehyde (MDA). The localization and protein expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) was detected by immunofluorescence and western blot, respectively. In addition, MCAC was intragastrically administrated in rats with ligature-induced experimental periodontitis. The effects were assessed by bone resorption, as well as the immunohistology staining of inflammatory and oxidative stress markers. RESULTS MCACs with cyclopentanone and containing pyrone showed lower toxicity than natural curcumin were synthesized (1A-10A, 1H-10H), among which, 1A exhibited the most potent cytoprotective effect against H2 O2 -induced damage. Such effects could be explained by the reduced MDA and ROS level, possibly through the nucleus translocation of Nrf2 and the induction of HO-1. Micro-CT results further indicated that 1A significantly reduced bone loss, along with an increased level of Nrf2 and HO-1, and decreased TNF-α and IL-1β. CONCLUSION The present study has synthesized a novel antioxidant MCAC 1A with good biosafety and stability. MCAC 1A could serve as a host response modulator with preventive and protective effects on periodontitis.
Collapse
Affiliation(s)
- Ya Zhao
- Department of Periodontics, School of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Zhiwei Zheng
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Menghan Zhang
- Department of Orthodontics, School of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yi Wang
- Department of Orthodontics, School of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Rongdang Hu
- Department of Orthodontics, School of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Weijia Lin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Chenyang Huang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Chuchu Xu
- Department of Periodontics, School of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Jianzhang Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hui Deng
- Department of Periodontics, School of Stomatology, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
11
|
Peng R, Chen Y, Wei L, Li G, Feng D, Liu S, Jiang R, Zheng S, Chen Y. Resistance to FGFR1-targeted therapy leads to autophagy via TAK1/AMPK activation in gastric cancer. Gastric Cancer 2020; 23:988-1002. [PMID: 32617693 DOI: 10.1007/s10120-020-01088-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Fibroblast growth factor receptor 1 (FGFR1) is frequently dysregulated in various tumors. FGFR inhibitors have shown promising therapeutic value in several preclinical models. However, tumors resistant to FGFR inhibitors have emerged, compromising therapeutic outcomes by demonstrating markedly aggressive metastatic progression; however, the underlying signaling mechanism of resistance remains unknown. METHODS We established FGFR inhibitor-resistant cell models using two gastric cancer (GC) cell lines, MGC-803 and BGC-823. RNA-seq was performed to determine the continuous cellular transcriptome changes between parental and resistant cells. We explored the mechanism of resistance to FGFR inhibitor, using a subcutaneous tumor model and GC patient-derived tumor organotypic culture. RESULTS We observed that FGFR1 was highly expressed in GC and FGFR1 inhibitor-resistant cell lines, demonstrating elevated levels of autophagic activity. These resistant cells were characterized by epithelial-mesenchymal transition (EMT) required to facilitate metastatic outgrowth. In drug-resistant cells, the FGFR1 inhibitor regulated GC cell autophagy via AMPK/mTOR signal activation, which could be blocked using either pharmacological inhibitors or essential gene knockdown. Furthermore, TGF-β-activated kinase 1 (TAK1) amplification and metabolic restrictions led to AMPK pathway activation and autophagy. In vitro and in vivo results demonstrated that the FGFR inhibitor AZD4547 and TAK1 inhibitor NG25 synergistically inhibited proliferation and autophagy in AZD4547-resistant cell lines and patient-derived GC organotypic cultures. CONCLUSIONS We elucidated the molecular mechanisms underlying primary resistance to FGFR1 inhibitors in GC, and revealed that the inhibition of FGFR1 and TAK1 signaling could present a potential novel therapeutic strategy for FGFR1 inhibitor-resistant GC patients.
Collapse
Affiliation(s)
- Rui Peng
- Department of General Surgery, Research Center for Clinical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, 210009, Jiangsu, China
| | - Yan Chen
- Key Laboratory of Emergency and Trauma of Ministry of Education, Tumor Institute of the First Affiliated Hospital, Hainan Medical University, Haikou, 571199, China
- Department of Ultrasound, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570311, China
| | - Liangnian Wei
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Gang Li
- Department of General Surgery, Research Center for Clinical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, 210009, Jiangsu, China
| | - Dongju Feng
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Siru Liu
- Key Laboratory of Emergency and Trauma of Ministry of Education, Tumor Institute of the First Affiliated Hospital, Hainan Medical University, Haikou, 571199, China
| | - Runqiu Jiang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210093, Jiangsu, China.
| | - Shaojiang Zheng
- Key Laboratory of Emergency and Trauma of Ministry of Education, Tumor Institute of the First Affiliated Hospital, Hainan Medical University, Haikou, 571199, China.
| | - Yun Chen
- Department of General Surgery, Research Center for Clinical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, 210009, Jiangsu, China.
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
12
|
Chen Q, Zhu M, Xie J, Dong Z, Khushafah F, Yun D, Fu W, Wang L, Wei T, Liu Z, Qiu P, Wu J, Li W. Design and Synthesis of Novel Nordihydroguaiaretic Acid (NDGA) Analogues as Potential FGFR1 Kinase Inhibitors With Anti-Gastric Activity and Chemosensitizing Effect. Front Pharmacol 2020; 11:518068. [PMID: 33041789 PMCID: PMC7517944 DOI: 10.3389/fphar.2020.518068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 08/13/2020] [Indexed: 12/31/2022] Open
Abstract
Aberrant fibroblast growth factor receptor-1 (FGFR1), a key driver promoting gastric cancer (GC) progression and chemo-resistance, has been increasingly recognized as a potential therapeutic target in GC. Hereon, we designed and synthesized a series of asymmetric analogues using Af23 and NDGA as lead compounds by retaining the basic structural framework (bisaryl-1,4-dien-3-one) and the unilateral active functional groups (3,4-dihydroxyl). Thereinto, Y14 showed considerable inhibitory activity against FGFR1. Next, pharmacological experiments showed that Y14 could significantly inhibit the phosphorylation of FGFR1 and its downstream kinase AKT and ERK, thus inhibiting the growth, survival, and migration of gastric cancer cells. Furthermore, compared with 5-FU treatment alone, the combination of Y14 and 5-FU significantly reduced the phosphorylation level of FGFR1, and enhanced the anti-cancer effect by inhibiting the viability and colony formation in two gastric cancer cell lines. These results confirmed that Y14 exerted anti-gastric activity and chemosensitizing effect by inhibiting FGFR1 phosphorylation and its downstream signaling pathway in vitro. This work also provides evidence that Y14, an effective FGFR1 inhibitor, could be used alone or in combination with chemotherapy to treat gastric cancer in the future.
Collapse
Affiliation(s)
- Qian Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Min Zhu
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jingwen Xie
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhaojun Dong
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Fatehi Khushafah
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Di Yun
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Weitao Fu
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ledan Wang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tao Wei
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhiguo Liu
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Peihong Qiu
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jianzhang Wu
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wulan Li
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
13
|
Recent advance in the development of novel, selective and potent FGFR inhibitors. Eur J Med Chem 2020; 186:111884. [DOI: 10.1016/j.ejmech.2019.111884] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/12/2019] [Accepted: 11/12/2019] [Indexed: 12/28/2022]
|
14
|
Design, synthesis and preliminary in-vitro studies of novel boronated monocarbonyl analogues of Curcumin (BMAC) for antitumor and β-amiloyd disaggregation activity. Bioorg Chem 2019; 93:103324. [DOI: 10.1016/j.bioorg.2019.103324] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/23/2019] [Accepted: 09/26/2019] [Indexed: 12/20/2022]
|
15
|
Li G, Zheng Y, Yao J, Hu L, Liu Q, Ke F, Feng W, Zhao Y, Yan P, He W, Deng H, Qiu P, Li W, Wu J. Design and Green Synthesis of Piperlongumine Analogs and Their Antioxidant Activity against Cerebral Ischemia-Reperfusion Injury. ACS Chem Neurosci 2019; 10:4545-4557. [PMID: 31491086 DOI: 10.1021/acschemneuro.9b00402] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The supplementation of exogenous antioxidants to scavenge excessive reactive oxygen species (ROS) is an effective treatment for cerebral ischemia-reperfusion injury (CIRI) in stroke. Piperlongumine (PL), a natural alkaloid, has a great potential as a neuroprotective agent, but it also has obvious toxicity. Moreover, its neuroprotective effects remain to be improved. In this study, we designed a series of novel PL analogs by hybridizing the screened low-toxicity diketene skeleton with antioxidant effect and the 3,4,5-trimethoxyphenyl group, which may increase the antioxidant activity of PL. The intermediate was synthesized by a novel green synthesis method, and 34 compounds were obtained. The compounds without obvious cytotoxicity have remarkable antioxidant effects, especially compared with diketene skeletons and PL. The cytoprotection of the active compound decreased significantly by reduction of the carbon-carbon double bonds of the Michael acceptor in the diketene skeleton. More importantly, further study revealed that compound A9, which has the best activity, can confer protection for cells against oxidative stress and attenuate brain injury in vivo. Overall, this study provided a promising drug candidate for the treatment of CIRI and guided the further development of drug research in oxidative stress-mediated diseases.
Collapse
Affiliation(s)
- Ge Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Yuantie Zheng
- Chemical Biology Research Center, School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Jiali Yao
- Chemical Biology Research Center, School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Linya Hu
- Chemical Biology Research Center, School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Qunpeng Liu
- Chemical Biology Research Center, School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
- College of Chemistry and Materials Engineering , Wenzhou University , Wenzhou , Zhejiang 325035 , China
| | - Furong Ke
- Chemical Biology Research Center, School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Weixiao Feng
- Chemical Biology Research Center, School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
- The First Affiliated Hospital of Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Ya Zhao
- Chemical Biology Research Center, School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
- Department of Periodontics, Hospital & School of Stomatology , Wenzhou Medical University , No. 373 West Xueyuan Road , Wenzhou , Zhejiang 325035 , China
| | - Pencheng Yan
- Chemical Biology Research Center, School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Wenfei He
- Chemical Biology Research Center, School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Hui Deng
- Department of Periodontics, Hospital & School of Stomatology , Wenzhou Medical University , No. 373 West Xueyuan Road , Wenzhou , Zhejiang 325035 , China
| | - Peihong Qiu
- Chemical Biology Research Center, School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Wulan Li
- The First Affiliated Hospital of Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| | - Jianzhang Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences , Wenzhou Medical University , Wenzhou , Zhejiang 325035 , China
| |
Collapse
|
16
|
Curcumin analogues and their hybrid molecules as multifunctional drugs. Eur J Med Chem 2019; 182:111631. [DOI: 10.1016/j.ejmech.2019.111631] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 08/02/2019] [Accepted: 08/16/2019] [Indexed: 02/07/2023]
|
17
|
Shimizu D, Saito T, Ito S, Masuda T, Kurashige J, Kuroda Y, Eguchi H, Kodera Y, Mimori K. Overexpression of FGFR1 Promotes Peritoneal Dissemination Via Epithelial-to-Mesenchymal Transition in Gastric Cancer. Cancer Genomics Proteomics 2018; 15:313-320. [PMID: 29976636 DOI: 10.21873/cgp.20089] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/27/2018] [Accepted: 04/30/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Peritoneal dissemination (PD) is one of the most common causes of cancer-related mortality in gastric cancer (GC). We aimed to identify PD-associated genes and investigate their role in GC. MATERIALS AND METHODS We identified FGFR1 as a putative PD-associated gene using a bioinformatics approach. The biological significance of FGFR1 in epithelial-to-mesenchymal transition (EMT) was evaluated according to the correlation with genes that participated in EMT and FGFR1 knockdown experiments. The associations between FGFR1 expression and the clinicopathological features were examined. RESULTS FGFR1 expression positively correlated with SNAI1, VIM and ZEB1 expression, and negatively correlated with CDH1 expression. Knockdown of FGFR1 suppressed the malignant phenotype of GC cells. High FGFR1 expression significantly correlated with the peritoneal lavage cytology and synchronous PD positivity as well as poor prognosis. CONCLUSION High FGFR1 expression was associated with PD via promotion of EMT and led to a poor prognosis of GC patients.
Collapse
Affiliation(s)
- Dai Shimizu
- Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara, Japan.,Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoko Saito
- Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara, Japan
| | - Shuhei Ito
- Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara, Japan
| | - Takaaki Masuda
- Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara, Japan
| | - Junji Kurashige
- Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara, Japan.,Department of Surgery, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Yosuke Kuroda
- Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara, Japan
| | - Hidetoshi Eguchi
- Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Tsurumihara, Japan
| |
Collapse
|
18
|
Design, synthesis, anti-lung cancer activity, and chemosensitization of tumor-selective MCACs based on ROS-mediated JNK pathway activation and NF-κB pathway inhibition. Eur J Med Chem 2018; 151:508-519. [DOI: 10.1016/j.ejmech.2018.03.051] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/28/2018] [Accepted: 03/18/2018] [Indexed: 12/14/2022]
|