1
|
Li T, Zhang R, Gong H, Tang Z, Li X, Gong Z, Challa M, Zou C, Zhang SL, Guo J, He Y. Synthesis and antibacterial evaluations of novel vancomycin analogues targeting bacteria membrane to combat Gram-negative infections. Eur J Med Chem 2025; 289:117483. [PMID: 40056801 DOI: 10.1016/j.ejmech.2025.117483] [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: 01/16/2025] [Revised: 02/20/2025] [Accepted: 03/04/2025] [Indexed: 03/10/2025]
Abstract
Vancomycin is primarily used to treat severe infections caused by Gram-positive bacteria and is often considered as the last-resort therapy in the life-threatening situation. However, it is inherently ineffective against Gram-negative bacteria. Herein, we report the design, synthesis, and biological evaluation of novel vancomycin analogues incorporated with lipophilic cationic groups. Through structural optimization and structure-activity relationship (SAR) studies, we identified vancomycin analogue 18b, which exhibited remarkable antibacterial activity against A. baumannii ATCC 17978, with a MIC of 8 μg/mL. In contrast, vancomycin showed no activity against this strain, even at concentration as high as 128 μg/mL. Further investigations revealed that 18b possesses rapid bactericidal properties, low toxicity, and a reduced propensity to induce bacterial resistance. The exceptional antibacterial performance of 18b is partially attributed to the presence of membrane-targeting, lipophilic piperazine cationic groups. In a mouse model infected with A. baumannii ATCC 17978, 18b exhibited excellent efficacy at a dose of 20 mg/kg, while no toxicity was observed. These findings highlight 18b as a promising candidate for further development in the fight against Gram-negative bacterial infections.
Collapse
Affiliation(s)
- Tao Li
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Ruixue Zhang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Hongzhi Gong
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Ziyi Tang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 266 Fangzheng Ave, Beibei, Chongqing, 400714, PR China; Chongqing Institute for Food and Drug Control, Chongqing, 401120, PR China
| | - Xinyu Li
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Zhi Gong
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Mahesh Challa
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Cheng Zou
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China
| | - Shao-Lin Zhang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China.
| | - Jian Guo
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China.
| | - Yun He
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Shuitu Technology Development Zone, No. 55 Daxuecheng South Rd., Shapingba, Chongqing, 401331, PR China; BayRay Innovation Center, Shenzhen Bay Laboratory, Shenzhen, 518132, PR China.
| |
Collapse
|
2
|
Yang X, Shi G, Lin Z, Qiu Y, Liu F, Hu K, Guo J, Peng H, He Y. Pathogen-targeting biomineralized bacterial outer membrane vesicles for eradicating both intracellular and extracellular Staphylococcus aureus. J Control Release 2025; 382:113702. [PMID: 40189054 DOI: 10.1016/j.jconrel.2025.113702] [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: 10/01/2024] [Revised: 02/07/2025] [Accepted: 04/03/2025] [Indexed: 04/13/2025]
Abstract
Intracellular Staphylococcus aureus is associated with recurrent infections and antibiotic resistance. Conventional antibiotics are ineffective against such intracellular bacterial pathogens, which calls for exploration of new approaches to treat these infections. Here, we report the development of pathogen-targeting biomineralized bacterial outer membrane vesicle (OMV) for targeted antibiotic delivery and eradicating both intracellular and extracellular S. aureus. These OMVs were derived from E. coli, and chemically modified with hydroxamate-type siderophore to target the intracellular S. aureus. The surface of OMV was coated with pH-sensitive calcium carbonate (CaCO3) to target the infection microenvironment. The CaCO3-coated siderophore-OMV (SOMV@CaCO3) was loaded with the antimicrobial drugs lysostaphin (Lsn) and mupirocin (Mup) (Lsn-SOMV@CaCO3-Mup) and administration of these OMVs resulted in effective eradication of both extracellular and intracellular S. aureus. Thus, Lsn-SOMV@CaCO3-Mup provides a novel and promising strategy for the treatment of invasive S. aureus infections.
Collapse
Affiliation(s)
- Xiaohong Yang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Gongming Shi
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Zihua Lin
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Yanfei Qiu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Feiyang Liu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Kecui Hu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Jian Guo
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China.
| | - Haibo Peng
- Chongqing Academy of Science and Technology, Chongqing 401123, China.
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China; Translational Innovation Center, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| |
Collapse
|
3
|
Marten I, Dilanas MEA, Podlech J. Fluorescent Carbazole-Derived Aza[5]Helicenes: Synthesis, Functionalization, and Characterization. Chemistry 2025:e202501081. [PMID: 40163366 DOI: 10.1002/chem.202501081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 03/31/2025] [Indexed: 04/02/2025]
Abstract
5,8-Dihydroindolo[2,3-c]carbazole (ICz), 9H-cinnolino[3,4-c]carbazole (CnCz), and variously alkyl-, alkenyl-, and aryl-substituted indolo[2,3-k]- and -[3,2-a]phenanthridines (IPs) were synthesized using an ortho fusion strategy with Suzuki cross couplings, intramolecular nitrene insertions, diazo couplings, and Morgan-Walls cyclizations as key reactions. The IPs were additionally transformed into organoboranes and helicene conjugates with tetraphenylethylene derivatives. The compounds fluoresce with large Stokes shifts, exhibit strong acidochromism, and show a good to excellent aggregation-induced emission. Their helical structure was elucidated by x-ray crystallographic analysis and by quantum chemical calculations. HOMO-LUMO gaps of 3.96-4.06 eV and S1-T1 gaps were calculated, with CnCz showing a small singlet-triplet inversion. Relative pKa values of 6.65-9.55 were estimated for the different types of azahelicenes.
Collapse
Affiliation(s)
- Inka Marten
- Institute of Organic Chemistry, Karlsruhe Institute of Technology (KIT), Kaiserstraße 12, Karlsruhe, Germany
| | - Melina E A Dilanas
- Institute of Inorganic Chemistry, Karlsruhe Institute of Technology (KIT), Kaiserstraße 12, Karlsruhe, Germany
| | - Joachim Podlech
- Institute of Organic Chemistry, Karlsruhe Institute of Technology (KIT), Kaiserstraße 12, Karlsruhe, Germany
| |
Collapse
|
4
|
Wang ZP, You W, Peng J, Xu B, Yang X, Tang W, He Y, Yang A, Yu C, Nian W. Synthesis and structural modification of the natural product Ivesinol to discover novel autophagy activators. Eur J Med Chem 2025; 284:117180. [PMID: 39724726 DOI: 10.1016/j.ejmech.2024.117180] [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: 11/04/2024] [Revised: 12/07/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024]
Abstract
Autophagy is a lysosome-dependent cellular degradation pathway that responds to a variety of environmental and cellular stresses, which is defective in aging and age-related diseases, therefore, targeting autophagy with small-molecule activators has potential therapeutic benefits. In this study, we successfully completed the first total synthesis of Ivesinol, an identified antibacterial natural product, and efficiently constructed a library of its analogs. To measure the effect of Ivesinol analogs on autophagic activity, we performed cell imaging-based screening approach, and observed that several Ivesinol analogs exhibited potent autophagy-regulating activity. Specifically, the derivative B2 significantly activated autophagy activity in concentration- and time-dependent manners, and even outperformed the commonly used activator Torin1 in activating autophagy in MCF-7 cells at 0.5 μM. Bioinformatics analysis showed that B2 treatment significantly impacted ubiquitin mediated proteolysis and AMPK signaling pathway, with functionally related gene sets displaying strong correlations. Based on these findings, we proposed that B2 activates autophagy by mechanisms involved in downregulation of key HSP70 family members, activation of the UPR, and ultimately leading to autophagy. In conclusion, we suggest that B2 could be a promising and valuable autophagy activator with significant potential for further development.
Collapse
Affiliation(s)
- Zhi-Peng Wang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Wenxin You
- School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Jie Peng
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Biao Xu
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaohong Yang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Wanyan Tang
- Department of Oncology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China
| | - Aimin Yang
- School of Life Sciences, Chongqing University, Chongqing, 401331, China.
| | - Chao Yu
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China.
| | - Weiqi Nian
- Department of Oncology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| |
Collapse
|
5
|
Zheng B, Wang YX, Wu ZY, Li XW, Qin LQ, Chen NY, Su GF, Su JC, Pan CX. Design, Synthesis and Bioactive Evaluation of Topo I/ c-MYC Dual Inhibitors to Inhibit Oral Cancer via Regulating the PI3K/AKT/NF-κB Signaling Pathway. Molecules 2025; 30:894. [PMID: 40005202 PMCID: PMC11858653 DOI: 10.3390/molecules30040894] [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: 12/24/2024] [Revised: 01/11/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
The significantly rising incidence of oral cancer worldwide urgently requires the identification of novel, effective molecular targets to inhibit the progression of malignancy. DNA topoisomerase I (Topo I) is a well-established target for cancer treatment, and many studies have shown that different cancer cell genes could be targeted more selectively with one type of Topo I inhibitor. In this report, a new scaffold pyridothieno[3,2-c]isoquinoline 11,11-dioxide was designed via the combination of the key fragment or bioisoster of Topo I inhibitor azaindenoisoquinolines and G-quadruplex binder quindoline. Thirty-two target derivatives were synthesized, among which compounds 7be, with potent Topo I inhibition, exhibited effective antiproliferative activity against Cal27, one of the oral cancer cell lines highly expressing Topo I protein. Further studies indicated that 7be could also inhibit the activation of PI3K/AKT/NF-κB pathway and downregulate the level of c-MYC, repress the colony formation and the migration of Cal27 cells and trigger apoptosis and autophagy. Molecular docking indicated that 7be could interact with the complex of Topo I and DNA via a mode similar to the indenoisoquinolines. The results of the Cal27 xenograft model confirmed that 7be exhibited promising anticancer efficacy in vivo, with tumor growth inhibition (TGI) of 64.7% at 20 mg/kg.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Cheng-Xue Pan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yu Cai Road, Guilin 541004, China; (B.Z.); (Y.-X.W.); (Z.-Y.W.); (X.-W.L.); (L.-Q.Q.); (N.-Y.C.); (G.-F.S.); (J.-C.S.)
| |
Collapse
|
6
|
Wei X, Zhang X, Peng Y, Wu J, Mo H, An Z, Deng X, Peng Y, Liu L, Jiang W, Chen J, Hu Z, Wang Z, Zhuo L. Identification of a novel 10-hydroxyevodiamine prodrug as a potent topoisomerase inhibitor with improved aqueous solubility for treatment of hepatocellular carcinoma. Eur J Med Chem 2024; 279:116807. [PMID: 39243453 DOI: 10.1016/j.ejmech.2024.116807] [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: 05/10/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/09/2024]
Abstract
Natural product evodiamine (Evo) and its synthetic derivatives represent an attractive dual Topo 1/2 inhibitors with broad-spectrum antitumor efficacy. However, the clinical applications of these compounds have been impeded by their poor aqueous solubility. Herein, a series of water-soluble 10-substituted-N(14)-phenylevodiamine derivatives were designed and synthesized. The most potent compound 45 featuring a quaternary ammonium salt fragment achieved robust aqueous solubility and nanomolar potency against a panel of human hepatoma cell lines Huh7, HepG2, SK-Hep-1, SMMC-7721, and SMMC-7721/DOX (doxorubicin-resistant cell). Further studies revealed that 45 could inhibit Topo 1 and Topo 2, induce apoptosis, arrest the cell cycle at the G2/M stage and inhibit the migration and invasion. Compound 45 exhibited potent antitumor activity (TGI = 51.1 %, 10 mg/kg) in the Huh7 xenograft model with acceptable safety profile. In addition, a 21-day long-term dose toxicity study confirmed that the maximum tolerated dose of compound 45 was 20 mg/kg. Overall, this study presented a promising Evo-derived candidate for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xiuzhen Wei
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xi Zhang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yan Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Junbo Wu
- Department of Colorectal Surgery, Hengyang Central Hospital, Hengyang, Hunan, 421001, China
| | - Hanxuan Mo
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhigang An
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xinyu Deng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Ying Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Linyi Liu
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Weifan Jiang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jinjin Chen
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zecheng Hu
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Zhen Wang
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, 810008, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, 410008, China; MOE Key Lab of Rare Pediatric Diseases, School of Life Sciences, Central South University, Changsha, 410000, China.
| | - Linsheng Zhuo
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
7
|
Wang B, Wu S, Jia S, Ruan X, Sheng C, Zhou Q. Discovery of Indolo[3,2- c]isoquinoline Derivatives as Novel Top1/2 Dual Inhibitors with Orally Efficacious Antitumor Activity and Low Toxicity. J Med Chem 2024; 67:14155-14174. [PMID: 39106476 DOI: 10.1021/acs.jmedchem.4c00982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2024]
Abstract
Topoisomerase (Top) inhibitors used in clinical cancer treatments are limited because of their toxicity and severe side effects. Noteworthily, Top1/2 dual inhibitors overcome the compensatory effect between Top1 and 2 inhibitors to exhibit stronger antitumor efficacies. In this study, a series of indolo[3,2-c]isoquinoline derivatives were designed as Top1/2 dual inhibitors possessing apparent antiproliferative activities. Mechanistic studies indicated that the optimal compounds 23 and 31 with increasing reactive oxygen species levels damage DNA, inducing both cancer cell apoptosis and cycle arrest. Importantly, the results of the toxicity studies showed that compounds 23 and 31 possessed good oral safety profiles. In xenograft models, compound 23 exhibited remarkable antitumor potency, which was superior to the clinical Top inhibitors irinotecan and etoposide. Overall, this work highlights the therapeutic potential and safety profile of compound 23 as a Top1/2 dual inhibitor in tumor therapy and provides valuable lead compounds for further development of Top inhibitors.
Collapse
Affiliation(s)
- Bichuan Wang
- State Key Laboratory of Natural Medicines, Department of Organic Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Shanchao Wu
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Shuolei Jia
- State Key Laboratory of Natural Medicines, Department of Organic Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Xiuqin Ruan
- State Key Laboratory of Natural Medicines, Department of Organic Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Chunquan Sheng
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai 200433, China
| | - Qingfa Zhou
- State Key Laboratory of Natural Medicines, Department of Organic Chemistry, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
8
|
Zeng H, Zhang S, Nie H, Li J, Yang J, Zhuang Y, Huang Y, Zeng M. Identification of FTY720 and COH29 as novel topoisomerase I catalytic inhibitors by experimental and computational studies. Bioorg Chem 2024; 147:107412. [PMID: 38696845 DOI: 10.1016/j.bioorg.2024.107412] [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: 03/16/2024] [Revised: 04/17/2024] [Accepted: 04/27/2024] [Indexed: 05/04/2024]
Abstract
The development of novel topoisomerase I (TOP1) inhibitors is crucial for overcoming the drawbacks and limitations of current TOP1 poisons. Here, we identified two potential TOP1 inhibitors, namely, FTY720 (a sphingosine 1-phosphate antagonist) and COH29 (a ribonucleotide reductase inhibitor), through experimental screening of known active compounds. Biological experiments verified that FTY720 and COH29 were nonintercalative TOP1 catalytic inhibitors that did not induce the formation of DNA-TOP1 covalent complexes. Molecular docking revealed that FTY720 and COH29 interacted favorably with TOP1. Molecular dynamics simulations revealed that FTY720 and COH29 could affect the catalytic domain of TOP1, thus resulting in altered DNA-binding cavity size. The alanine scanning and interaction entropy identified Arg536 as a hotspot residue. In addition, the bioinformatics analysis predicted that FTY720 and COH29 could be effective in treating malignant breast tumors. Biological experiments verified their antitumor activities using MCF-7 breast cancer cells. Their combinatory effects with TOP1 poisons were also investigated. Further, FTY720 and COH29 were found to cause less DNA damage compared with TOP1 poisons. The findings provide reliable lead compounds for the development of novel TOP1 catalytic inhibitors and offer new insights into the potential clinical applications of FTY720 and COH29 in targeting TOP1.
Collapse
Affiliation(s)
- Huang Zeng
- Institute of Hakka Medicinal Bio-resources, Medical College, Jiaying University, Meizhou 514031, China.
| | - Shengyuan Zhang
- Institute of Hakka Medicinal Bio-resources, Medical College, Jiaying University, Meizhou 514031, China
| | - Hua Nie
- Institute of Hakka Medicinal Bio-resources, Medical College, Jiaying University, Meizhou 514031, China
| | - Junhao Li
- Department of Physics and Astronomy, Uppsala University, Lägerhyddsvägen 1, SE-75121 Uppsala, Sweden
| | - Jiunlong Yang
- Institute of Hakka Medicinal Bio-resources, Medical College, Jiaying University, Meizhou 514031, China
| | - Yuanbei Zhuang
- Institute of Hakka Medicinal Bio-resources, Medical College, Jiaying University, Meizhou 514031, China
| | - Yingjie Huang
- Institute of Hakka Medicinal Bio-resources, Medical College, Jiaying University, Meizhou 514031, China
| | - Miao Zeng
- Institute of Hakka Medicinal Bio-resources, Medical College, Jiaying University, Meizhou 514031, China
| |
Collapse
|
9
|
Çevik UA, Kaya B, Celik I, Rudrapal M, Rakshit G, Karayel A, Levent S, Osmaniye D, Sağlık Özkan BN, Baysal M, Atlı Ekliog̈lu Ö, Özkay Y, Kaplancıklı ZA. New Benzimidazole-Triazole Derivatives as Topoisomerase I Inhibitors: Design, Synthesis, Anticancer Screening, and Molecular Modeling Studies. ACS OMEGA 2024; 9:13359-13372. [PMID: 38524479 PMCID: PMC10955584 DOI: 10.1021/acsomega.3c10345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/26/2024]
Abstract
In this study, we designed, synthesized, and evaluated a series of 1,2,4-triazole benzimidazoles for their cytotoxic effects against the A549, C6, and NIH3T3 cell lines. Additionally, these compounds were assessed for their inhibitory activity against DNA topoisomerase I, aiming to develop novel anticancer agents. The synthesized final compounds 4a-h were characterized using 1H NMR, 13C NMR, and HRMS. Among them, compounds 4b and 4h emerged as the most potent agents against the A549 cell line, exhibiting an IC50 value of 7.34 ± 0.21 μM and 4.56 ± 0.18 μM, respectively. These results were compared to standard drugs, doxorubicin (IC50 = 12.420 ± 0.5 μM) and Hoechst 33342 (IC50 = 0.422 ± 0.02 μM). Notably, all tested compounds displayed higher cytotoxicity toward A549 cells than C6 cells. Compounds 4b and 4h demonstrated significant inhibitory activity against topoisomerase I, highlighting their potential as lead compounds in anticancer therapy. Subsequent in silico molecular docking studies were conducted to elucidate the potential binding interactions of compounds 4b and 4h with the target enzyme topoisomerase I. Molecular dynamics studies also assessed and validated the binding affinity and stability. These studies confirmed the promising binding affinity of these compounds, reinforcing their status as lead candidates. According to DFT, compound 4b having the lower energy gap value (ΔE = 3.598 eV) is more chemically reactive than the others, which is consistent with significant inhibitory activity against topoisomerase I. Furthermore, in silico ADME profiles for compounds 4b and 4h were evaluated using SwissADME, providing insights into their pharmacokinetic properties.
Collapse
Affiliation(s)
- Ulviye Acar Çevik
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eski̧ehir 26470, Turkey
| | - Betül Kaya
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Zonguldak Bülent Ecevit University, Zonguldak 67100, Turkey
| | - Ismail Celik
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Kayseri 38039, Turkey
| | - Mithun Rudrapal
- Department
of Pharmaceutical Sciences, School of Biotechnology and Pharmaceutical
Sciences, Vignan’s Foundation for
Science, Technology & Research (Deemed to Be University), Guntur 522213, India
| | - Gourav Rakshit
- Department
of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Ranchi 835215, India
| | - Arzu Karayel
- Department
of Physics, Faculty of Arts and Science, Hitit University, Çorum 19030, Turkey
| | - Serkan Levent
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eski̧ehir 26470, Turkey
| | - Derya Osmaniye
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eski̧ehir 26470, Turkey
| | | | - Merve Baysal
- Department
of Pharmaceutical Toxicology, Faculty of Pharmacy, Anadolu University, Eski̧ehir 26470, Turkey
| | - Özlem Atlı Ekliog̈lu
- Department
of Pharmaceutical Toxicology, Faculty of Pharmacy, Anadolu University, Eski̧ehir 26470, Turkey
| | - Yusuf Özkay
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eski̧ehir 26470, Turkey
| | - Zafer Asım Kaplancıklı
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eski̧ehir 26470, Turkey
| |
Collapse
|
10
|
Yang Y, Cao Y, Yu J, Yu X, Guo Y, Wang F, Ren Q, Li C. Design and synthesis of novel 3-amino-5-phenylpyrazole derivatives as tubulin polymerization inhibitors targeting the colchicine-binding site. Eur J Med Chem 2024; 267:116177. [PMID: 38280356 DOI: 10.1016/j.ejmech.2024.116177] [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: 11/20/2023] [Revised: 01/04/2024] [Accepted: 01/23/2024] [Indexed: 01/29/2024]
Abstract
As the basic unit of microtubules, tubulin is one of the most important targets in the study of anticarcinogens. A novel series of 3-amino-5-phenylpyrazole derivatives were designed and synthesized, and evaluates for their biological activities. Among them, a majority of compounds exerted excellent inhibitory activities against five cancer cell lines in vitro. Especially, compound 5b showed a strong antiproliferative activity against MCF-7 cells, with IC50 value of 38.37 nM. Further research indicated that compound 5b can inhibit the polymerization of tubulin targeting the tubulin colchicine-binding sites. Furthermore, 5b could arrest MCF-7 cells at the G2/M phase and induce MCF-7 cells apoptotic in a dose-dependent and time-dependent manners, and regulate the level of related proteins expression. Besides, compound 5b could inhibit the cancer cell migration and angiogenesis. In addition, 5b could inhibit tumor growth in MCF-7 xenograft model without obvious toxicity. All these results indicating that 5b could be a promising antitumor agent targeting tubulin colchicine-binding site and it was worth further study.
Collapse
Affiliation(s)
- Yang Yang
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, PR China; Department of Trauma Center, Affiliated Hospital of Nantong University, No.20 Xisi Road, Chongchuan District, Nantong City, Jiangsu Province, 226001, PR China
| | - Yan Cao
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Jingwen Yu
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Xinyu Yu
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Yali Guo
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Fei Wang
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Qingjia Ren
- Tibetan Medicine Research Institute, Tibetan Traditional Medical College, Tibet, 850000, PR China.
| | - Caolong Li
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, PR China.
| |
Collapse
|
11
|
Sun P, Xu N, Zhang X, Gao YF, Zhang JY, Zhang QM, Liang C, Yu RL, Xia YM, Gao WW. Rational design and synthesis of triazene-amonafide derivatives as novel potential antitumor agents causing oxidative damage towards DNA through intercalation mode. Bioorg Chem 2024; 144:107141. [PMID: 38244381 DOI: 10.1016/j.bioorg.2024.107141] [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: 12/14/2023] [Revised: 01/14/2024] [Accepted: 01/16/2024] [Indexed: 01/22/2024]
Abstract
In this work, we rationally designed and synthesized two novel triazene-amonafide derivatives 2-(2-(diisopropylamino)ethyl)-5-(3,3-dimethyltriaz-1-en-1-yl)-1H-benzo[de]isoquinoline-1,3(2H)-dione (D-11) and 5-(3,3-diethyltriaz-1-en-1-yl)-2-(2-(diisopropylamino)ethyl)-1H-benzo[de]isoquinoline-1,3(2H)-dione (D-12) as potential antitumor agents. The DNA damage induced by the intercalation mode of D-11 (D-12) towards DNA was electrochemically detected through the construction of efficient biosensors. The consecutive processes of reversible redox of naphthylimide ring and irreversible oxidation of triazene moiety were elucidated on the surface of glassy carbon electrode (GCE) by CV, SWV, and DPV methods. Electrochemical biosensors were obtained through the immobilization of ctDNA, G-quadruplexes, poly(dG), and poly(dA), respectively, on the clean surface of GCE. After the incubation of biosensors with D-11 or D-12, the peaks of dGuo and dAdo decreased prominently, and the peak of 8-oxoGua appeared at +0.50 V, suggesting that the interaction between D-11 (D-12) and DNA could result in the oxidative damage of guanine. Unexpected, the as-prepared DNA biosensor possessed satisfactory anti-interference property and good practicability in real samples. UV-vis and fluorescence spectra, and gel electrophoresis assays were employed to further confirm the intercalation mode of D-11 (D-12) towards DNA base pairs. Moreover, D-11 was proved to exhibit stronger anti-proliferation activity than mitionafide and amonafide against both A549 and HeLa cell lines.
Collapse
Affiliation(s)
- Ping Sun
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, People's Republic of China
| | - Na Xu
- Yantai Fushan District People's Hospital, Yantai 265599, People's Republic of China
| | - Xue Zhang
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, People's Republic of China
| | - Yun-Fei Gao
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, People's Republic of China
| | - Jia-Yin Zhang
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, People's Republic of China
| | - Qi-Ming Zhang
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, People's Republic of China
| | - Chen Liang
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, People's Republic of China
| | - Ri-Lei Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Ya-Mu Xia
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, People's Republic of China.
| | - Wei-Wei Gao
- State Key Laboratory Base of Eco-chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, People's Republic of China.
| |
Collapse
|
12
|
Feng Y, An Q, Zhao Z, Wu M, Yang C, Liang W, Xu X, Jiang T, Zhang G. Beta-elemene: A phytochemical with promise as a drug candidate for tumor therapy and adjuvant tumor therapy. Biomed Pharmacother 2024; 172:116266. [PMID: 38350368 DOI: 10.1016/j.biopha.2024.116266] [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: 11/22/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND β-Elemene (IUPAC name: (1 S,2 S,4 R)-1-ethenyl-1-methyl-2,4-bis(prop-1-en-2-yl) cyclohexane), is a natural compound found in turmeric root. Studies have demonstrated its diverse biological functions, including its anti-tumor properties, which have been extensively investigated. However, these have not yet been reviewed. The aim of this review was to provide a comprehensive summary of β-elemene research, with respect to disease treatment. METHODS β-Elemene-related articles were found in PubMed, ScienceDirect, and Google Scholar databases to systematically summarize its structure, pharmacokinetics, metabolism, and pharmacological activity. We also searched the Traditional Chinese Medicine System Pharmacology database for therapeutic targets of β-elemene. We further combined these targets with the relevant literature for KEGG and GO analyses. RESULTS Studies on the molecular mechanisms underlying β-elemene activity indicate that it regulates multiple pathways, including STAT3, MAPKs, Cyclin-dependent kinase 1/cyclin B, Notch, PI3K/AKT, reactive oxygen species, METTL3, PTEN, p53, FAK, MMP, TGF-β/Smad signaling. Through these molecular pathways, β-elemene has been implicated in tumor cell proliferation, apoptosis, migration, and invasion and improving the immune microenvironment. Additionally, β-elemene increases chemotherapeutic drug sensitivity and reverses resistance by inhibiting DNA damage repair and regulating pathways including CTR1, pak1, ERK1/2, ABC transporter protein, Prx-1 and ERCC-1. Nonetheless, owing to its lipophilicity and low bioavailability, additional structural modifications could improve the efficacy of this drug. CONCLUSION β-Elemene exhibits low toxicity with good safety, inhibiting various tumor types via diverse mechanisms in vivo and in vitro. When combined with chemotherapeutic drugs, it enhances efficacy, reduces toxicity, and improves tumor killing. Thus, β-elemene has vast potential for research and development.
Collapse
Affiliation(s)
- Yewen Feng
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Qingwen An
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Zhengqi Zhao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Mengting Wu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Chuqi Yang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - WeiYu Liang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Xuefei Xu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China
| | - Tao Jiang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China.
| | - Guangji Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Zhejiang 310053, China; Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Zhejiang 310053, China; Traditional Chinese Medicine "Preventing Disease" Wisdom Health Project Research Center of Zhejiang, Zhejiang 310053, China.
| |
Collapse
|
13
|
Guo YL, Yu JW, Cao Y, Cheng KX, Dong-Zhi SNM, Zhang YF, Ren QJ, Yin Y, Li CL. Design, synthesis, and biological evaluation of harmine derivatives as topoisomerase I inhibitors for cancer treatment. Eur J Med Chem 2024; 265:116061. [PMID: 38154256 DOI: 10.1016/j.ejmech.2023.116061] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/27/2023] [Accepted: 12/14/2023] [Indexed: 12/30/2023]
Abstract
A series of β-carboline derivatives were designed and synthesized by introducing the chalcone moiety into the harmine. The synthesized derivatives were evaluated their anti-proliferative activities against six human cancer cell lines (MCF-7, MDA-MB-231, HepG2, HT29, A549, and PC-3) and one normal cell line (L02). Among them, compound G11 exhibited the potent anti-proliferative activity against MCF-7 cell line, with an IC50 value of 0.34 μM. Further biological studies revealed that compound G11 inhibited colony formation of MCF-7 cells, suppressed MCF-7 cell migration by downregulating migration-associated protein MMP-2. In addition, it could induce apoptosis of MCF-7 cells by downregulating Bcl-2 and upregulating Cleaved-PARP, Bax, and phosphorylated Bim proteins. Furthermore, compound G11 can act as a Topo I inhibitor, affecting DNA synthesis and transcription, thereby inhibiting cancer cell proliferation. Moreover, compound G11 inhibited tumor growth in 4T1 syngeneic transplant mice with an inhibition rate of 43.19 % at a dose of 10 mg/kg, and 63.87 % at 20 mg/kg, without causing significant toxicity to the mice or their organs, achieving the goal of reduced toxicity and increased efficacy. All these results indicate of G11 has enormous potential as an anti-tumor agent and merits further investigation.
Collapse
Affiliation(s)
- Ya-Li Guo
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Jing-Wen Yu
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Yan Cao
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Ke-Xin Cheng
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Suo-Nan-Mu Dong-Zhi
- Tibetan Medicine Research Institute, Tibetan Traditional Medical College, Tibet, 850000, PR China
| | - Yan-Fei Zhang
- Tibetan Medicine Research Institute, Tibetan Traditional Medical College, Tibet, 850000, PR China
| | - Qing-Jia Ren
- Tibetan Medicine Research Institute, Tibetan Traditional Medical College, Tibet, 850000, PR China
| | - Yong Yin
- Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Cao-Long Li
- Department of Chemistry, School of Science, China Pharmaceutical University, Nanjing, 211198, PR China.
| |
Collapse
|
14
|
Zhao R, Zhu J, Jiang X, Bai R. Click chemistry-aided drug discovery: A retrospective and prospective outlook. Eur J Med Chem 2024; 264:116037. [PMID: 38101038 DOI: 10.1016/j.ejmech.2023.116037] [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: 10/22/2023] [Revised: 11/20/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
Click chemistry has emerged as a valuable tool for rapid compound synthesis, presenting notable advantages and convenience in the exploration of potential drug candidates. In particular, in situ click chemistry capitalizes on enzymes as reaction templates, leveraging their favorable conformation to selectively link individual building blocks and generate novel hits. This review comprehensively outlines and introduces the extensive use of click chemistry in compound library construction, and hit and lead discovery, supported by specific research examples. Additionally, it discusses the limitations and precautions associated with the application of click chemistry in drug discovery. Our intention for this review is to contribute to the development of a modular synthetic approach for the rapid identification of drug candidates.
Collapse
Affiliation(s)
- Rui Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Junlong Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Xiaoying Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Renren Bai
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China.
| |
Collapse
|
15
|
Pavunkumar V, Harikrishnan K, Mohanakrishnan AK. Synthesis of Cytotoxic Quino[4,3- b]carbazole Frameworks through an Intramolecular Diels-Alder Reaction. J Org Chem 2024; 89:191-215. [PMID: 38133929 DOI: 10.1021/acs.joc.3c01909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Abstract
An intramolecular Diels-Alder reaction of positionally isomeric indole-2/3-phenylvinyl-N-alkynylated (N-phenylsulfonyl)amines has been successfully exploited for the synthesis of quino[4,3-b]carbazole and its analogues. This reaction proceeds through a [4 + 2] cycloaddition followed by elimination and deprotection of phenylsulfonyl units to afford the quinocarbazoles in moderate to good yields. The reaction features a broad substrate scope and remarkable functional group forbearance. A preliminary in vitro cytotoxicity evaluation of representative quino[4,3-b]carbazoles was performed against NCI-H460 human cancer cell culture. Among the quino[4,3-b]carbazoles evaluated, five of the fluorine-containing quinocarbazoles displayed nano molar range (0.8-2.0 nm) GI50 values. The UV-vis and fluorescence spectral studies of representative quinocarbazoles were also performed. Like ellipticine, four of the quinocarbazoles displayed dual emissions confirming the existence of p-quinonoid like tautomeric forms in a polar protic solvent.
Collapse
Affiliation(s)
- Vinayagam Pavunkumar
- Department of Organic Chemistry, School of Chemical Science, University of Madras, Guindy Campus, Chennai 600 025, Tamil Nadu, India
| | - Kesavan Harikrishnan
- Department of Organic Chemistry, School of Chemical Science, University of Madras, Guindy Campus, Chennai 600 025, Tamil Nadu, India
| | - Arasambattu K Mohanakrishnan
- Department of Organic Chemistry, School of Chemical Science, University of Madras, Guindy Campus, Chennai 600 025, Tamil Nadu, India
| |
Collapse
|
16
|
Wu HY, Cao Z, Li SQ, Fu YW, Li JM, Li XH, He CM, Chen JY. Visible-Light-Mediated Annulation/Thiolation of 2-Isocyanobiaryls with Disulfides to Organoylthiophenanthridines Derivatives. J Org Chem 2023. [PMID: 38044560 DOI: 10.1021/acs.joc.3c02152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
A visible-light-induced annulation/thiolation of 2-isocyanobiaryls with dialkyl(aryl)disulfides has been established, delivering a sustainable and atom-economic route to 6-organoylthiophenanthridines with wild functional group tolerance and good to excellent yields under oxidant-, base-, and transition-metal-free conditions.
Collapse
Affiliation(s)
- Hong-Yu Wu
- Hunan Provincial Key Laboratory of Materials Protection for Electric Power and Transportation, Changsha University of Science and Technology, Changsha, Hunan 410114, China
| | - Zhong Cao
- Hunan Provincial Key Laboratory of Materials Protection for Electric Power and Transportation, Changsha University of Science and Technology, Changsha, Hunan 410114, China
| | - Shi-Qi Li
- College of Chemistry and Chemical Engineering, Yangtze Normal University, Chongqing 408000, China
| | - Yu-Wei Fu
- College of Chemistry and Chemical Engineering, Yangtze Normal University, Chongqing 408000, China
| | - Jia-Min Li
- College of Chemistry and Chemical Engineering, Yangtze Normal University, Chongqing 408000, China
| | - Xing-Hong Li
- College of Chemistry and Chemical Engineering, Yangtze Normal University, Chongqing 408000, China
| | - Chun-Mei He
- College of Chemistry and Chemical Engineering, Yangtze Normal University, Chongqing 408000, China
| | - Jin-Yang Chen
- College of Chemistry and Chemical Engineering, Yangtze Normal University, Chongqing 408000, China
| |
Collapse
|
17
|
Krishnan A, Kamaraj S. Direct Access to Quinone-Fused 5-Substituted-1,4-Benzodiazepine Scaffolds from Azidoquinones with/without [1,2]-Azide-Nitrogen Migration: Mechanistic Insights. J Org Chem 2023; 88:16315-16329. [PMID: 37966974 DOI: 10.1021/acs.joc.3c01810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
Seven-membered nitrogen heterocycles have a strong influence in drug discovery due to their inherent 3D character, which allows the ability to explore a vast conformational space with a biological target. Notably, the privileged 1,4-benzodiazepine scaffold is dominant in treating the central nervous system due to its binding affinity with the GABAA receptor. Herein, we report a protocol for the transformation of azidoquinones to p-quinone fused 5-substituted-1,4-benzodiazepines (p-QBZDs) from InCl3-catalyzed intermolecular tandem cycloannulation of azidoquinones with amines and aldehydes. Detailed mechanistic studies reveal that the EDA complex between azidoquinones and InCl3 is crucial in determining the reaction pathway. In the absence of EDA complex formation, the reaction proceeds via the intermediacy of 2,3-bridged-2H-azirine followed by regiospecific addition of an amine to C═N/ring opening/cyclization to deliver p-QBZD with 1,2-azide-nitrogen migration. In the case of EDA complex formation, the reaction proceeds through regioselective aza-Michael addition/nitrene insertion with aldehyde and subsequent cyclization to deliver p-QBZD and p-quinone fused imidazole as a secondary product without 1,2-azide-nitrogen migration. This protocol provides straightforward access to redox-active quinone embedded 5-substituted-1,4-benzodiazepines from azidoquinones with diverse substrate scopes that would find potential applications in medicinal chemistry and drug discovery.
Collapse
Affiliation(s)
- Ashokkumar Krishnan
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Sriraghavan Kamaraj
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| |
Collapse
|
18
|
Zhong L, Wang Q, Wang Y, Cheng Y, Xiong Y, Peng H, Zhou Z, He Y, Dai Y. Facile and stereospecific synthesis of diverse β- N-glycosyl sulfonamide scaffolds via palladium catalysis. Chem Commun (Camb) 2023; 59:12907-12910. [PMID: 37823213 DOI: 10.1039/d3cc04063a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Glycosylation is an important strategy to improve the druggability of lead compounds. Here, we present a palladium-catalysed stereospecific N-glycosylation of sulfonamides. This approach stands out with wide substrate scope, high functional group tolerance, and easy scalability, furnishing a broad spectrum of densely functionalized β-N-glycosyl sulfonamides with good efficiency and exceptional regio-/stereoselectivity. Diverse drug-like glycosulfonamido scaffolds have been constructed via a late-stage diversification strategy and various facile synthetic transformations of the products. Collectively, the established protocol provides a valuable tool for efficiently preparing glycosyl sulfonamides to facilitate drug discovery.
Collapse
Affiliation(s)
- Lei Zhong
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, P. R. China.
| | - Qunliang Wang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, P. R. China.
| | - Yujuan Wang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, P. R. China.
| | - Yiyang Cheng
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, P. R. China.
| | - Yimeng Xiong
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, P. R. China.
| | - Haibo Peng
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, P. R. China
| | - Zhen Zhou
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, P. R. China.
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, P. R. China.
| | - Yuanwei Dai
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, P. R. China.
| |
Collapse
|
19
|
Li Z, Zhu X, Wu Z, Sun T, Tong Y. Recent Advances in Cyanotoxin Synthesis and Applications: A Comprehensive Review. Microorganisms 2023; 11:2636. [PMID: 38004647 PMCID: PMC10673588 DOI: 10.3390/microorganisms11112636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
Over the past few decades, nearly 300 known cyanotoxins and more than 2000 cyanobacterial secondary metabolites have been reported from the environment. Traditional studies have focused on the toxic cyanotoxins produced by harmful cyanobacteria, which pose a risk to both human beings and wildlife, causing acute and chronic poisoning, resulting in diarrhea, nerve paralysis, and proliferation of cancer cells. Actually, the biotechnological potential of cyanotoxins is underestimated, as increasing studies have demonstrated their roles as valuable products, including allelopathic agents, insecticides and biomedicines. To promote a comprehensive understanding of cyanotoxins, a critical review is in demand. This review aims to discuss the classifications; biosynthetic pathways, especially heterogenous production; and potential applications of cyanotoxins. In detail, we first discuss the representative cyanotoxins and their toxic effects, followed by an exploration of three representative biosynthetic pathways (non-ribosomal peptide synthetases, polyketide synthetases, and their combinations). In particular, advances toward the heterologous biosynthesis of cyanotoxins in vitro and in vivo are summarized and compared. Finally, we indicate the potential applications and solutions to bottlenecks for cyanotoxins. We believe that this review will promote a comprehensive understanding, synthetic biology studies, and potential applications of cyanotoxins in the future.
Collapse
Affiliation(s)
- Zipeng Li
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300072, China; (Z.L.); (Z.W.)
| | - Xiaofei Zhu
- Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology, Tianjin University, Tianjin 300072, China;
| | - Zhengyu Wu
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300072, China; (Z.L.); (Z.W.)
| | - Tao Sun
- Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology, Tianjin University, Tianjin 300072, China;
- Center for Biosafety Research and Strategy, Tianjin University, Tianjin 300072, China
| | - Yindong Tong
- School of Environmental Science and Engineering, Tianjin University, Tianjin 300072, China; (Z.L.); (Z.W.)
- College of Ecology and Environment, Tibet University, Lhasa 850000, China
| |
Collapse
|
20
|
Koteva K, Xu M, Wang W, Fiebig-Comyn AA, Cook MA, Coombes BK, Wright GD. Synthetic Biology Facilitates Semisynthetic Development of Type V Glycopeptide Antibiotics Targeting Vancomycin-Resistant Enterococcus. J Med Chem 2023. [PMID: 37315221 DOI: 10.1021/acs.jmedchem.3c00633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The continued efficacy of glycopeptide antibiotics (GPAs) against Gram-positive bacteria is challenged by the emergence and spread of GPA-resistant pathogens, particularly vancomycin-resistant enterococci (VRE). The growing frequency of GPA resistance propels the need for innovative development of more effective antibiotics. Unlike canonical GPAs like vancomycin, Type V GPAs adopt a distinct mode of action by binding peptidoglycan and blocking the activity of autolysins essential for cell division, rendering them a promising class of antibiotics for further development. In this study, the Type V GPA, rimomycin A, was modified to generate 32 new analogues. Compound 17, derived from rimomycin A through N-terminal acylation and C-terminal amidation, exhibited improved anti-VRE activity and solubility. In a VRE-A neutropenic thigh infection mouse model, compound 17 significantly lowered the bacterial load by 3-4 orders of magnitude. This study sets the stage to develop next-generation GPAs in response to growing VRE infections.
Collapse
Affiliation(s)
- Kalinka Koteva
- David Braley Centre for Antibiotic Discovery, M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Min Xu
- David Braley Centre for Antibiotic Discovery, M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4K1, Canada
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, West 7th Avenue No. 32, 300308 Tianjin, China
- Haihe Laboratory of Synthetic Biology, West 15th Avenue No. 21, 300308 Tianjin, China
| | - Wenliang Wang
- David Braley Centre for Antibiotic Discovery, M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Aline A Fiebig-Comyn
- David Braley Centre for Antibiotic Discovery, M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Michael A Cook
- David Braley Centre for Antibiotic Discovery, M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Brian K Coombes
- David Braley Centre for Antibiotic Discovery, M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Gerard D Wright
- David Braley Centre for Antibiotic Discovery, M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| |
Collapse
|
21
|
Chen XM, Zhou JY, Liu SQ, Song LH, Wang HL, Wang Q, Liang SM, Lu L, Wei JH, Huang R, Zhang Y. Design, synthesis, and antitumor evaluation of morpholine substituted bisnaphthalimides as DNA targeting agents. Bioorg Med Chem Lett 2023; 85:129218. [PMID: 36894107 DOI: 10.1016/j.bmcl.2023.129218] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/25/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
A series of mono- and bisnaphthalimides derivatives containing 3-nitro and 4-morpholine moieties were designed, synthesized, and evaluated for their in vitro anticancer activities against four cancer cell lines. Some compounds exhibited relatively good antiproliferative activity on the cell lines tested, in comparison with mitonafide and amonafide. It is noteworthy that bisnaphthalimide A6 was identified as the most potent compound in anti-proliferation against MGC-803 cells, with an IC50 lowered to 0.09 μM, a far greater potency than that of mono-naphthalimide A7, mitonafide, and amonafide. A gel electrophoresis assay revealed that DNA and Topo I were the potential targets of compounds A6 and A7. The treatment of CNE-2 cells with compounds A6 and A7 resulted in an S phase cell cycle arrest, accompanied by the upregulation of the expression levels of the antioncogene p27 and the down-regulation of the expression levels of CDK2 and cyclin E. In addition, compounds A6 and A7-induced apoptosis was further confirmed by flow cytometry, ROS generation assay, and Hoechst 33,258 staining. In particular, in vivo antitumor assay results revealed that bisnaphthalimide A6 exhibited potent anticancer efficiency in an MGC-803 xenograft tumor model, in comparison with mitonafide, and had lower toxicity than mono-naphthalimide A7. In brief, the results suggested that bisnaphthalimide derivatives containing 3-nitro and 4-morpholine moieties might serve as DNA binding agents for the development of new antitumor agents.
Collapse
Affiliation(s)
- Xiao-Man Chen
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China
| | - Jian-Yu Zhou
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China
| | - Shuang-Qiang Liu
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China
| | - Long-Hao Song
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China
| | - Hui-Ling Wang
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China
| | - Qi Wang
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China
| | - Si-Min Liang
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China
| | - Lin Lu
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China
| | - Jian-Hua Wei
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China.
| | - Rizhen Huang
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China.
| | - Ye Zhang
- Guangxi Key Laboratory for Pharmaceutical Molecular Discovery and Druggability Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 5411199, China.
| |
Collapse
|
22
|
Shen Z, Tang Q, Jiao W, Shao H, Ma X. One-Pot Synthesis of 2- C-Branched Glycosyl Triazoles by Integrating 1,2-Cyclopropanated Sugar Ring-Opening Azidation and CuAAC Reaction. J Org Chem 2022; 87:16736-16742. [PMID: 36399138 DOI: 10.1021/acs.joc.2c02390] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A series of 2-C-branched glycosyl triazoles including triazole-tethered oligosaccharides and glycopeptides were synthesized in one pot from 1,2-cyclopropanated sugars or 2'-acetonyl-2-O-Ts-C-furanosides, NaN3, and alkynes using PEG-400 as a single solvent. Nucleophilic ring-opening azidation of 1,2-cyclopropanated sugars (or 2'-acetonyl group 1,2-migration-azidation of C-furanosides) obtained glycosyl azides, which upon reaction with alkynes under CuAAC conditions achieved glycosyl triazoles in good yields and high stereoselectivity without the need to change the solvent and isolate any intermediates.
Collapse
Affiliation(s)
- Zhongke Shen
- Natural Products Research Centre, Chengdu Institute of Biology, Chinese Academy of Sciences, 610041Chengdu, People's Republic of China.,University of Chinese Academy of Sciences, Beijing100049, People's Republic of China
| | - Qin Tang
- Natural Products Research Centre, Chengdu Institute of Biology, Chinese Academy of Sciences, 610041Chengdu, People's Republic of China
| | - Wei Jiao
- Natural Products Research Centre, Chengdu Institute of Biology, Chinese Academy of Sciences, 610041Chengdu, People's Republic of China
| | - Huawu Shao
- Natural Products Research Centre, Chengdu Institute of Biology, Chinese Academy of Sciences, 610041Chengdu, People's Republic of China
| | - Xiaofeng Ma
- Natural Products Research Centre, Chengdu Institute of Biology, Chinese Academy of Sciences, 610041Chengdu, People's Republic of China.,University of Chinese Academy of Sciences, Beijing100049, People's Republic of China
| |
Collapse
|