Natural Products and Bioprospecting    2024, Vol. 14 Issue (4) : 33-33     DOI: 10.1007/s13659-024-00455-x
ORIGINAL ARTICLES |
Metabolism characterization and toxicity of N-hydap, a marine candidate drug for lung cancer therapy by LC-MS method
Jindi Lu1, Weimin Liang1, Yiwei Hu2, Xi Zhang1, Ping Yu1, Meiqun Cai1, Danni Xie1, Qiong Zhou1, Xuefeng Zhou2, Yonghong Liu2, Junfeng Wang2, Jiayin Guo1, Lan Tang1
1. NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China;
2. CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology/Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
Download: PDF(5009 KB)   HTML ()  
Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks    
Abstract  N-Hydroxyapiosporamide (N-hydap), a marine product derived from a sponge-associated fungus, has shown promising inhibitory effects on small cell lung cancer (SCLC). However, there is limited understanding of its metabolic pathways and characteristics. This study explored the in vitro metabolic profiles of N-hydap in human recombinant cytochrome P450s (CYPs) and UDP-glucuronosyltransferases (UGTs), as well as human/rat/mice microsomes, and also the pharmacokinetic properties by HPLC-MS/MS. Additionally, the cocktail probe method was used to investigate the potential to create drug-drug interactions (DDIs). N-Hydap was metabolically unstable in various microsomes after 1 h, with about 50% and 70% of it being eliminated by CYPs and UGTs, respectively. UGT1A3 was the main enzyme involved in glucuronidation (over 80%), making glucuronide the primary metabolite. Despite low bioavailability (0.024%), N-hydap exhibited a higher distribution in the lungs (26.26%), accounting for its efficacy against SCLC. Administering N-hydap to mice at normal doses via gavage did not result in significant toxicity. Furthermore, N-hydap was found to affect the catalytic activity of drug metabolic enzymes (DMEs), particularly increasing the activity of UGT1A3, suggesting potential for DDIs. Understanding the metabolic pathways and properties of N-hydap should improve our knowledge of its drug efficacy, toxicity, and potential for DDIs.
Keywords N-Hydap      Metabolism      Pharmacokinetics      DMEs      Toxicity      DDIs     
Fund:This work was supported by the National Natural Science Foundation of China (Nos. 82274002, 42376124), Marine Economy Development Project of Guangdong Province (GDNRC[2021]52), Hainan Provincial Natural Science Foundation of China (823CXTD393), Key-Area Research and Development Program of Guangdong Province (2023B1111050008)
Corresponding Authors: Junfeng Wang,E-mail:wangjunfeng@scsio.ac.cn;Jiayin Guo,E-mail:g1227@smu.edu.cn;Lan Tang,E-mail:tl405@smu.edu.cn     E-mail: wangjunfeng@scsio.ac.cn;g1227@smu.edu.cn;tl405@smu.edu.cn
Issue Date: 01 August 2024
Service
E-mail this article
E-mail Alert
RSS
Articles by authors
Jindi Lu
Weimin Liang
Yiwei Hu
Xi Zhang
Ping Yu
Meiqun Cai
Danni Xie
Qiong Zhou
Xuefeng Zhou
Yonghong Liu
Junfeng Wang
Jiayin Guo
Lan Tang
Trendmd:   
Cite this article:   
Jindi Lu,Weimin Liang,Yiwei Hu, et al. Metabolism characterization and toxicity of N-hydap, a marine candidate drug for lung cancer therapy by LC-MS method[J]. Natural Products and Bioprospecting, 2024, 14(4): 33-33.
URL:  
http://npb.kib.ac.cn/EN/10.1007/s13659-024-00455-x     OR     http://npb.kib.ac.cn/EN/Y2024/V14/I4/33
[1] Rudin CM, Brambilla E, Faivre-Finn C, Sage J. Small-cell lung cancer. Nat Rev Dis Prim. 2021;7(1):3.
[2] Yang S, Zhang Z, Wang Q. Emerging therapies for small cell lung cancer. J Hematol Oncol. 2019;12(1):47.
[3] Liu Q, Luo X, Yi L, Zeng X, Tan C. First-line chemo-immunotherapy for extensive-stage small-cell lung cancer: a united states-based cost-effectiveness analysis. Front Oncol. 2021;11: 699781.
[4] Eckardt JR, von Pawel J, Pujol JL, Papai Z, Quoix E, Ardizzoni A, Poulin R, Preston AJ, Dane G, Ross G. Phase III study of oral compared with intravenous topotecan as second-line therapy in small-cell lung cancer. J Clin Oncol. 2007;25(15):2086-92.
[5] Owonikoko TK, Park K, Govindan R, Ready N, Reck M, Peters S, Dakhil SR, Navarro A, Rodriguez-Cid J, Schenker M, Lee JS, Gutierrez V, Percent I, Morgensztern D, Barrios CH, Greillier L, Baka S, Patel M, Lin WH, Selvaggi G, Baudelet C, Baden J, Pandya D, Doshi P, Kim HR. Nivolumab and ipilimumab as maintenance therapy in extensive-disease small-cell lung cancer: CheckMate 451. J Clin Oncol. 2021;39(12):1349-59.
[6] Chen J, Hu Y, Zhang J, Wang Q, Wu X, Huang W, Wang Q, Cai G, Wang H, Ou T, Feng W, Liu P, Liu Y, Wang J, Huang J, Wang J. Therapeutic targeting RORgamma with natural product N-hydroxyapiosporamide for small cell lung cancer by reprogramming neuroendocrine fate. Pharmacol Res. 2022;178: 106160.
[7] Han J, Liu C, Li L, Zhou H, Liu L, Bao L, Chen Q, Song F, Zhang L, Li E, Liu L, Pei Y, Jin C, Xue Y, Yin W, Ma Y, Liu H. Decalin-containing tetramic acids and 4-hydroxy-2-pyridones with antimicrobial and cytotoxic activity from the fungus Coniochaeta cephalothecoides collected in Tibetan Plateau (Medog). J Org Chem. 2017;82(21):11474-86.
[8] Wang J, Wei X, Qin X, Lin X, Zhou X, Liao S, Yang B, Liu J, Tu Z, Liu Y. Arthpyrones A-C, pyridone alkaloids from a sponge-derived fungus Arthrinium arundinis ZSDS1-F3. Org Lett. 2015;17(3):656-9.
[9] Yang Y, Liu Q, Shi X, Zheng Q, Chen L, Sun Y. Advances in plant-derived natural products for antitumor immunotherapy. Arch Pharm Res. 2021;44(11):987-1011.
[10] Khalifa SAM, Elias N, Farag MA, Chen L, Saeed A, Hegazy MF, Moustafa MS, Abd El-Wahed A, Al-Mousawi SM, Musharraf SG, Chang FR, Iwasaki A, Suenaga K, Alajlani M, Goransson U, El-Seedi HR. Marine natural products: a source of novel anticancer drugs. Mar Drugs. 2019;17(9):491.
[11] Zheng J, Wang J, Wang Q, Zou H, Wang H, Zhang Z, Chen J, Wang Q, Wang P, Zhao Y, Lu J, Zhang X, Xiang S, Wang H, Lei J, Chen HW, Liu P, Liu Y, Han F, Wang J. Targeting castration-resistant prostate cancer with a novel RORgamma antagonist elaiophylin. Acta Pharm Sin B. 2020;10(12):2313-22.
[12] Ge G, Zhu M. Preface for special issue on new analytical techniques and methods in drug metabolism and pharmacokinetics. J Pharm Anal. 2020;10(3):iii-iv.
[13] Saunders LJ, Fitzsimmons PN, Nichols JW, Gobas F. In vitro-in vivo extrapolation of hepatic and gastrointestinal biotransformation rates of hydrophobic chemicals in rainbow trout. Aquat Toxicol. 2020;228: 105629.
[14] Sharma SS, Sharma S, Bureik M. Screening of the whole human cytochrome P450 complement (CYPome) with enzyme bag cocktails. J Pharm Anal. 2020;10(3):271-6.
[15] Henderson GL, Harkey MR, Gershwin ME, Hackman RM, Stern JS, Stresser DM. Effects of ginseng components on c-DNA-expressed cytochrome P450 enzyme catalytic activity. Life Sci. 1999;65(15):PL209-14.
[16] Wang JJ, Guo JJ, Zhan J, Bu HZ, Lin JH. An in-vitro cocktail assay for assessing compound-mediated inhibition of six major cytochrome P450 enzymes. J Pharm Anal. 2014;4(4):270-8.
[17] Song JH, Sun DX, Chen B, Ji DH, Pu J, Xu J, Tian FD, Guo L. Inhibition of CYP3A4 and CYP2C9 by podophyllotoxin: implication for clinical drug-drug interactions. J Biosci. 2011;36(5):879-85.
[18] Zhang Y, Kuchimanchi M, Zhu M, Doshi S, Hoang T, Kasichayanula S. Assessment of pharmacokinetic interaction between rilotumumab and epirubicin, cisplatin and capecitabine (ECX) in a phase 3 study in gastric cancer. Br J Clin Pharmacol. 2017;83(5):1048-55.
[19] Yang SY, Yi JM, Chun J, Park S, Bui TT, Yun HY, Chae JW, Jeong MK. Evaluation of the potential herb-drug interaction between Bojungikki-tang and PD-L1 immunotherapy in a syngeneic mouse model. Front Pharmacol. 2023;14:1181263.
[20] Xie H, Wu J, Liu D, Liu M, Zhang H, Huang S, Xiong Y, Xia C. In vitro inhibition of UGT1A3, UGT1A4 by ursolic acid and oleanolic acid and drug-drug interaction risk prediction. Xenobiotica. 2017;47(9):785-92.
[21] Seibert E, Tracy TS. Different enzyme kinetic models. Methods Mol Biol. 2014;1113:23-35.
[22] Nakanishi T, Tamai I. Interaction of drug or food with drug transporters in intestine and liver. Curr Drug Metab. 2015;16(9):753-64.
[23] Takakusa H, Iwazaki N, Nishikawa M, Yoshida T, Obika S, Inoue T. Drug metabolism and pharmacokinetics of antisense oligonucleotide therapeutics: typical profiles, evaluation approaches, and points to consider compared with small molecule drugs. Nucleic Acid Ther. 2023;33(2):83-94.
[24] Tess D, Chang GC, Keefer C, Carlo A, Jones R, Di L. In vitro-in vivo extrapolation and scaling factors for clearance of human and preclinical species with liver microsomes and hepatocytes. AAPS J. 2023;25(3):40.
[25] Sodhi JK, Benet LZ. Successful and unsuccessful prediction of human hepatic clearance for lead optimization. J Med Chem. 2021;64(7):3546-59.
[26] Fang ZZ, He RR, Cao YF, Tanaka N, Jiang C, Krausz KW, Qi Y, Dong PP, Ai CZ, Sun XY, Hong M, Ge GB, Gonzalez FJ, Ma XC, Sun HZ. A model of in vitro UDP-glucuronosyltransferase inhibition by bile acids predicts possible metabolic disorders. J Lipid Res. 2013;54(12):3334-44.
[27] Thomas C, Pellicciari R, Pruzanski M, Auwerx J, Schoonjans K. Targeting bile-acid signalling for metabolic diseases. Nat Rev Drug Discov. 2008;7(8):678-93.
[28] Meech R, Hu DG, McKinnon RA, Mubarokah SN, Haines AZ, Nair PC, Rowland A, Mackenzie PI. The UDP-glycosyltransferase (UGT) superfamily: new members, new functions, and novel paradigms. Physiol Rev. 2019;99(2):1153-222.
[29] Ticho AL, Malhotra P, Dudeja PK, Gill RK, Alrefai WA. Intestinal absorption of bile acids in health and disease. Compr Physiol. 2019;10(1):21-56.
[30] Liu S, Hou L, Li C, Zhao Y, Yao X, Zhang X, Tian X. Contributions of UDP-glucuronosyltransferases to human hepatic and intestinal metabolism of ticagrelor and inhibition of UGTs and cytochrome P450 enzymes by ticagrelor and its glucuronidated metabolite. Front Pharmacol. 2021;12: 761814.
[31] Dong J, Wang NN, Yao ZJ, Zhang L, Cheng Y, Ouyang D, Lu AP, Cao DS. ADMETlab: a platform for systematic ADMET evaluation based on a comprehensively collected ADMET database. J Cheminform. 2018;10(1):29.
[32] Tian H, Ketkar R, Tao P. ADMETboost: a web server for accurate ADMET prediction. J Mol Model. 2022;28(12):408.
[33] Waghray D, Zhang Q. Inhibit or evade multidrug resistance P-glycoprotein in cancer treatment. J Med Chem. 2018;61(12):5108-21.
[34] Chen M, Hu S, Li Y, Gibson AA, Fu Q, Baker SD, Sparreboom A. Role of Oatp2b1 in drug absorption and drug-drug interactions. Drug Metab Dispos. 2020;48(5):419-25.
[35] Liu W, Shi J, Zhu L, Dong L, Luo F, Zhao M, Wang Y, Hu M, Lu L, Liu Z. Reductive metabolism of oxymatrine is catalyzed by microsomal CYP3A4. Drug Des Dev Ther. 2015;9:5771-83.
[36] Jeong H, Lee J, Kim S, Yeo YY, So H, Wu H, Song YS, Jang CY, Kim HD, Kim MJ, Chang M. Hepatic metabolism of sakuranetin and its modulating effects on cytochrome P450s and UDP-glucuronosyltransferases. Molecules. 2018;23(7):1542.
[1] Joana Ribeiro, Henrique Araújo-Silva, Mário Fernandes, Joilna Alves da Silva, Francisco das Chagas L. Pinto, Otília Deusdenia L. Pessoa, Hélcio Silva Santos, Jane Eire Silva Alencar de Menezes, Andreia C. Gomes. Petrosamine isolated from marine sponge Petrosia sp. demonstrates protection against neurotoxicity in vitro and in vivo[J]. Natural Products and Bioprospecting, 2024, 14(2): 4-4.
[2] Kengo Iwata, Farhana Ferdousi, Yoshinobu Arai, Hiroko Isoda. Modulation of mitochondrial activity by sugarcane (Saccharum officinarum L.) top extract and its bioactive polyphenols: a comprehensive transcriptomics analysis in C2C12 myotubes and HepG2 hepatocytes[J]. Natural Products and Bioprospecting, 2024, 14(1): 2-2.
[3] Shuyuan Mo, Ziming Zhao, Zi Ye, Zhihong Huang, Yaxin Zhang, Wanqi Yang, Jianping Wang, Zhengxi Hu, Yonghui Zhang. New secondary metabolites with cytotoxicity from fungus Penicillium roqueforti[J]. Natural Products and Bioprospecting, 2023, 13(3): 17-17.
[4] Li Hou, Cui-Xuan Mei, Chun-Mao Yuan, Gui-Hua Tang, Duo-Zhi Chen, Qing Zhao, Hong-Ping He, Ming-Ming Cao, Xiao-Jiang Hao. Five new limonoids isolated from Walsura robusta[J]. Natural Products and Bioprospecting, 2023, 13(2): 7-7.
[5] Ya-Li Hu, Xing-Ren Li, Gang Xu. Carascynol A, a hybrid of caryophyllane-type terpenoid and a C6 unit degraded by polyprenylated acylphloroglucinols from Hypericum ascyron[J]. Natural Products and Bioprospecting, 2022, 12(6): 38-38.
[6] Yue Zhao, Wen-Ke Gao, Xiang-Dong Wang, Li-Hua Zhang, Hai-Yang Yu, Hong-Hua Wu. Phytochemical and pharmacological studies on Solanum lyratum: a review[J]. Natural Products and Bioprospecting, 2022, 12(6): 39-39.
[7] Xin Zhang, Yun-Bao Ma, Xiao-Feng He, Tian-Ze Li, Chang-An Geng, Li-Hua Su, Shuang Tang, Zhen Gao, Ji-Jun Chen. Artemyrianosins A–J, cytotoxic germacrane-type sesquiterpene lactones from Artemisia myriantha[J]. Natural Products and Bioprospecting, 2022, 12(3): 16-16.
[8] Natividad Herrera Cano, Sebastian A. Andujar, Cristina Theoduloz, Daniel A. Wunderlin, Ana N. Santiago, Guillermo Schmeda-Hirschmann, Ricardo D. Enriz, Gabriela E. Feresin. Arylated analogues of cypronazole: fungicidal effect and activity on human fibroblasts. Docking analysis and molecular dynamics simulations[J]. Natural Products and Bioprospecting, 2022, 12(2): 9-9.
[9] Ruo-Song Zhang, Yang-Yang Liu, Pei-Feng Zhu, Qiong Jin, Zhi Dai, Xiao-Dong Luo. Furostanol Saponins from Asparagus cochinchinensis and Their Cytotoxicity[J]. Natural Products and Bioprospecting, 2021, 11(6): 651-658.
[10] Patrick O. Sakyi, Richard K. Amewu, Robert N. O. A. Devine, Emahi Ismaila, Whelton A. Miller, Samuel K. Kwofie. The Search for Putative Hits in Combating Leishmaniasis: The Contributions of Natural Products Over the Last Decade[J]. Natural Products and Bioprospecting, 2021, 11(5): 489-544.
[11] Soad Z. El-Emam. Sesamol Alleviates the Cytotoxic Effect of Cyclophosphamide on Normal Human Lung WI-38 Cells via Suppressing RAGE/NF-κB/Autophagy Signaling[J]. Natural Products and Bioprospecting, 2021, 11(3): 333-344.
[12] Christian Bailly. Anticancer Properties of Lobetyolin, an Essential Component of Radix Codonopsis (Dangshen)[J]. Natural Products and Bioprospecting, 2021, 11(2): 143-153.
[13] Chen Shi, Yue-Ling Peng, Juan He, Zheng-Hui Li, Ji-Kai Liu, Tao Feng. Structures, Chemical Conversions, and Cytotoxicity of Tricholopardins C and D, Two Tricholoma Triterpenoids from the Wild Mushroom Tricholoma pardinum[J]. Natural Products and Bioprospecting, 2021, 11(2): 235-241.
[14] Yi Chen, Song-Wei Li, Fang-Zhou Yin, Min Yang, Xia-Juan Huan, Ze-Hong Miao, Xiao-Ming Wang, Yue-Wei Guo. Lagerindicine, a New Pyrrole Alkaloid Isolated from the Flowers of Lagerstroemia indica Linnaeus[J]. Natural Products and Bioprospecting, 2021, 11(1): 73-79.
[15] Mohammad Sanad Abu-Darwish, Célia Cabral, Zulfigar Ali, Mei Wang, Shabana I. Khan, Melissa R. Jacob, Surendra K. Jain, Babu L. Tekwani, Fazila Zulfiqar, Ikhlas A. Khan, Hatem Taifour, Lígia Salgueiro, Thomas Efferth. Salvia ceratophylla L. from South of Jordan: new insights on chemical composition and biological activities[J]. Natural Products and Bioprospecting, 2020, 10(5): 307-316.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed