Skip to main content

OPINION article

Front. Pharmacol., 15 June 2023
Sec. Pharmacology of Anti-Cancer Drugs
This article is part of the Research Topic Drugs and methods that enhance the anti-cancer efficacy of artesunate View all 3 articles

Artemisitene: a promising natural drug candidate with various biological activities needs to confirm the interactional targets

  • 1Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
  • 2Institute of Immunology and Inflammatory Diseases, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
  • 3Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China

Introduction

The road of new drug research and development is full of challenges. To ensure the safety and effectiveness of drugs in patients, new drug research and development are a long screening process of discovering promising compounds and continuously eliminating compounds with obvious side effects. In the history of new drug research and development, natural products are a huge treasure trove and have made great contributions to the treatment of human diseases. For instance, the discovery and research of artemisinin (qinghaosu), an active component extracted from the plant Artemisia annua L. helps people defeat malaria. The famous Chinese scientist Youyou Tu shared the 2015 Nobel Prize in Physiology or Medicine for the discovery of artemisinin (Croft and Ward, 2015), which highlights the great development potential of natural products especially traditional Chinese medicine.

Artemisitene (ATT) is a natural compound initially isolated from the herb Artemisia annua L. and is an endoperoxide close to the famous antimalaria drug artemisinin (Acton and Klayman, 1985). It was reported that ATT could be obtained from artemisinin through a one-pot selenoxide elimination reaction (Paitayatat et al., 1997), and Avery et al. optimized the synthesis route of ATT (Avery et al., 2003). Over the past few decades, more and more studies show that ATT has a variety of biological activities in multiple human diseases. However, the unclear target protein is one of the most critical bottlenecks affecting the clinical application of ATT. Therefore, it is urgent and necessary to identify the target proteins of ATT in treating multiple human diseases.

Biological activities of ATT

As an analogue of artemisinin, ATT was shown to have anti-malaria activity (Acton et al., 1993; Paitayatat et al., 1997; Avery et al., 2003). In addition, Chen et al. revealed that ATT serves as a novel nuclear factor erythroid 2-related factor 2 (Nrf2) activator that could induce an antioxidant response in an Nrf2-dependent manner by alleviating Nrf2 ubiquitination and increasing its stability and suppress lung injury induced by bleomycin (Chen et al., 2016). Subsequently, Liu et al. reported that the cysteine (Cys) residues of the cytosolic Nrf2 repressor Kelch-like ECH-associated protein-1(Keap1) might be a potential target contributing to ATT-stimulated Nrf2 activation and is indispensable for stabilizing Nrf2 and facilitating Nrf2-mediated transcription of downstream genes (Liu et al., 2019). Recently, researchers found that ATT could block the production of reactive oxygen species (ROS, especially mitochondrial ROS) and prevent NLRP3 inflammasome assembly and activation, resulting in the decrease of IL-1β production (Hua et al., 2022). Besides, ATT could also suppress NLRC4 and AIM2 inflammasome-mediated IL-1β secretion and IL-6 production, and it has the ability to alleviate ulcerative colitis induced by sulfate sodium salt in mice, suggesting that ATT might regard as a drug candidate in the treatment of inflammatory disorders (Hua et al., 2022). Our recent research also demonstrated that ATT has therapeutic potential for rheumatoid arthritis, an inflammatory autoimmune disease (Chen et al., 2022). In the above study, ATT was shown to have the capacity to manage rheumatoid arthritis by inhibiting proliferation, migration, and invasion, as well as inducing apoptosis of rheumatoid arthritis-fibroblast-like synoviocytes through regulating METTL3/ICAM2/PI3K/AKT/p300 signaling pathway (Chen et al., 2022). It’s worth noting that most reports have focused on the anti-cancer activity of ATT. For instance, in 1993, Woerdenbag et, al. found that the anti-cancer activity of ATT required higher concentrations than the in vitro antimalarial activity (Woerdenbag et al., 1993). Years later, Efferth’s group reported that ATT exhibits a cytotoxic effect on multiple cell lines such as cervical carcinoma, leukemia, breast cancer, colon cancer, melanoma, brain cancer, lung cancer, ovarian cancer, and renal cancer, etc., and its anti-cancer activity is superior to some artemisinins (including artemisinin, arteanuine B, arteether, artemether, artesunate) and other compounds exist in Artemisia annua L. like scopoletin and 1,8-cineole (Efferth and Oesch, 2004; Efferth et al., 2011). Interestingly, they also demonstrated that ATT regulates iron-related genes to induce ferroptosis, a new form of cell death, contributing to an attractive strategy for cancer treatment (Ooko et al., 2015). Furthermore, our group further revealed that ATT exhibits anti-cancer activity by regulating NEDD4/c-Myc/topoisomerase pathway and inhibits tumor growth in xenotransplanted tumor models (Chen et al., 2018). These findings supported that ATT could be a promising natural drug candidate to treat multiple human diseases.

Discussion

The target is the junction and pivot of the interaction between pharmaceutical chemical components and the human biological system. The discovery of the target is of great significance to the optimization of the pharmaceutical chemical structure and the interpretation of the effect mechanism. Nowadays, the identification of drug targets is gradually becoming a bottleneck that hinders the development of pharmaceutical chemistry. What’s more, the mechanism of drug action is revealed after their clinical application. For a classic example, artemisinin is used for the treatment of malaria over the past few decades, but its interactional targets are uncertain. It’s exciting that researchers used an alkyne-tagged and biotin-linked artemisinin analogue to identify more than 100 artemisinin covalent binding target proteins, many of which participate in the biological processes essential for the parasite (Wang et al., 2015). This study supported a unifying model to elucidate the action mechanism of artemisinin in killing parasites. However, it is still necessary to further confirm the interactional relationship between artemisinin and target proteins using gold standard methods such as isothermal titration calorimetry, surface plasmon resonance, cellular thermal shift assay, and so on. Previous studies have demonstrated that ATT exhibits excellent biological activities in multiple human diseases and investigating the direct target proteins of ATT became one of the priority issues for promoting the clinical application of ATT. Therefore, it is urgent and necessary to identify the target proteins of ATT in treating multiple human diseases with available drug target screening and validation methods.

In summary, ATT has a variety of biological activities including anti-malaria, anti-anti-cancer, anti-lung injury, anti-ulcerative colitis, anti-rheumatoid arthritis, anti-oxidative stress-related diseases, etc., and could be regarded as a parent compound for structural modification and even a promising natural drug candidate for the treatments of multiple human diseases. Nevertheless, the unclear target protein is one of the most critical limitations affecting the development of ATT. The identification of ATT target proteins will help to study the structure optimization, structure-activity relationship, and molecular mechanism of ATT in treating human diseases, laying a foundation for its clinical application, and also providing a scientific basis for the application of related targets in clinical translational medicine.

Author contributions

XL and JC wrote the manuscript. JC also edited and submitted the manuscript. All authors contributed to the article and approved the submitted version.

Funding

This study was supported by the National Natural Science Foundation of China (No. 82103692), Shenzhen Science and Technology Program (No. JCYJ20210324110209026), and the Scientific Research Foundation of Peking University Shenzhen Hospital KYQD202100X (No. KYQD2023250).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Acton, N., Karle, J. M., and Miller, R. E. (1993). Synthesis and antimalarial activity of some 9-substituted artemisinin derivatives. J. Med. Chem. 36 (17), 2552–2557. doi:10.1021/jm00069a014

PubMed Abstract | CrossRef Full Text | Google Scholar

Acton, N., and Klayman, D. L. (1985). Artemisitene, a new sesquiterpene lactone endoperoxide from Artemisia annua. Planta Med. 51 (5), 441–442. doi:10.1055/s-2007-969543

PubMed Abstract | CrossRef Full Text | Google Scholar

Avery, M. A., Muraleedharan, K. M., Desai, P. V., Bandyopadhyaya, A. K., Furtado, M. M., and Tekwani, B. L. (2003). Structure-activity relationships of the antimalarial agent artemisinin. 8. design, synthesis, and CoMFA studies toward the development of artemisinin-based drugs against leishmaniasis and malaria. J. Med. Chem. 46 (20), 4244–4258. doi:10.1021/jm030181q

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, J., Li, W., Cui, K., Ji, K., Xu, S., and Xu, Y. (2018). Artemisitene suppresses tumorigenesis by inducing DNA damage through deregulating c-Myc-topoisomerase pathway. Oncogene 37 (37), 5079–5087. doi:10.1038/s41388-018-0331-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, J., Lin, X., He, J., Liu, D., He, L., Zhang, M., et al. (2022). Artemisitene suppresses rheumatoid arthritis progression via modulating METTL3-mediated N6-methyladenosine modification of ICAM2 mRNA in fibroblast-like synoviocytes. Clin. Transl. Med. 12 (12), e1148. doi:10.1002/ctm2.1148

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, W., Li, S., Li, J., Zhou, W., Wu, S., Xu, S., et al. (2016). Artemisitene activates the Nrf2-dependent antioxidant response and protects against bleomycin-induced lung injury. FASEB J. 30 (7), 2500–2510. doi:10.1096/fj.201500109R

PubMed Abstract | CrossRef Full Text | Google Scholar

Croft, S. L., and Ward, S. (2015). The Nobel Prize in Medicine 2015: Two drugs that changed global health. Sci. Transl. Med. 7 (316), 316ed14. doi:10.1126/scitranslmed.aad5868

PubMed Abstract | CrossRef Full Text | Google Scholar

Efferth, T., Herrmann, F., Tahrani, A., and Wink, M. (2011). Cytotoxic activity of secondary metabolites derived from Artemisia annua L. towards cancer cells in comparison to its designated active constituent artemisinin. Phytomedicine 18 (11), 959–969. doi:10.1016/j.phymed.2011.06.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Efferth, T., and Oesch, F. (2004). Oxidative stress response of tumor cells: Microarray-based comparison between artemisinins and anthracyclines. Biochem. Pharmacol. 68 (1), 3–10. doi:10.1016/j.bcp.2004.03.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Hua, L., Liang, S., Zhou, Y., Wu, X., Cai, H., Liu, Z., et al. (2022). Artemisinin-derived artemisitene blocks ROS-mediated NLRP3 inflammasome and alleviates ulcerative colitis. Int. Immunopharmacol. 113, 109431. doi:10.1016/j.intimp.2022.109431

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, S., Xu, S., Wei, R., Cui, Z., Wu, X., Wei, R., et al. (2019). Keap1 cystenine 151 as a potential target for artemisitene-induced Nrf2 activation. Biomed. Res. Int. 2019, 5198138. doi:10.1155/2019/5198138

PubMed Abstract | CrossRef Full Text | Google Scholar

Ooko, E., Saeed, M. E., Kadioglu, O., Sarvi, S., Colak, M., Elmasaoudi, K., et al. (2015). Artemisinin derivatives induce iron-dependent cell death (ferroptosis) in tumor cells. Phytomedicine 22 (11), 1045–1054. doi:10.1016/j.phymed.2015.08.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Paitayatat, S., Tarnchompoo, B., Thebtaranonth, Y., and Yuthavong, Y. (1997). Correlation of antimalarial activity of artemisinin derivatives with binding affinity with ferroprotoporphyrin IX. J. Med. Chem. 40 (5), 633–638. doi:10.1021/jm960767v

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, J., Zhang, C. J., Chia, W. N., Loh, C. C., Li, Z., Lee, Y. M., et al. (2015). Haem-activated promiscuous targeting of artemisinin in Plasmodium falciparum. Nat. Commun. 6, 10111. doi:10.1038/ncomms10111

PubMed Abstract | CrossRef Full Text | Google Scholar

Woerdenbag, H. J., Moskal, T. A., Pras, N., Malingre, T. M., el-Feraly, F. S., Kampinga, H. H., et al. (1993). Cytotoxicity of artemisinin-related endoperoxides to Ehrlich ascites tumor cells. J. Nat. Prod. 56 (6), 849–856. doi:10.1021/np50096a007

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: artemisitene, biological activities, rheumatoid arthritis, cancer, inflammatory disease

Citation: Lin X and Chen J (2023) Artemisitene: a promising natural drug candidate with various biological activities needs to confirm the interactional targets. Front. Pharmacol. 14:1221291. doi: 10.3389/fphar.2023.1221291

Received: 15 May 2023; Accepted: 09 June 2023;
Published: 15 June 2023.

Edited by:

Jianqiang Xu, Dalian University of Technology, China

Reviewed by:

Guozheng Huang, Anhui University of Technology, China

Copyright © 2023 Lin and Chen. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jian Chen, chenjian@pkuszh.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.