1
|
Vakhrusheva O, Zhao F, Markowitsch SD, Slade KS, Brandt MP, Tsaur I, Cinatl J, Michaelis M, Efferth T, Blaheta RA, Haferkamp A, Juengel E. Artesunate Inhibits Metastatic Potential in Cisplatin-Resistant Bladder Cancer Cells by Altering Integrins. Cells 2025; 14:570. [PMID: 40277897 PMCID: PMC12026051 DOI: 10.3390/cells14080570] [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: 02/20/2025] [Revised: 03/28/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
The survival of patients with locally advanced and metastatic bladder cancer (BCa) is persistently low. Hence, new treatment options are urgently needed. Artesunate (ART) a derivative of artemisinin, used in Traditional Chinese Medicine, shows anti-tumor activity extending over a broad spectrum of human cancers. As we have previously shown, ART inhibits growth in cisplatin-sensitive (parental) and cisplatin-resistant BCa cells. However, how ART acts on the metastatic potential of BCa remained unclear. To clarify, we applied ART to parental and cisplatin-resistant RT4, RT112, T24, and TCCSup BCa cell lines. We examined tumor cell adhesion to vascular endothelium and immobilized collagen and evaluated chemotactic activity, migration, and invasive activity of the BCa cells. Adhesion receptors, integrin α and β subtypes, integrin-linked kinase (ILK), and focal adhesion kinase (FAK) were investigated. The functional relevance of integrin expression altered by ART was determined by blocking studies. ART significantly reduced tumor cell adhesion to vascular endothelium and immobilized collagen in parental as well as in cisplatin-resistant BCa cells. Depending on cell type, ART suppressed tumor cell motility and diminished integrin expression (surface and total). Functional blocking of integrins altered by ART reduced cell adhesion and invasion of the BCa cells. Thus, the metastatic potential of parental and cisplatin-resistant BCa cells was significantly inhibited by ART, making it a promising treatment option for patients with advanced or therapy-resistant BCa.
Collapse
Affiliation(s)
- Olesya Vakhrusheva
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler-Strasse 3, 72076 Tübingen, Germany
| | - Fuguang Zhao
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
| | - Sascha Dennis Markowitsch
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
| | - Kimberly Sue Slade
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
| | - Maximilian Peter Brandt
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
| | - Igor Tsaur
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
- Department of Urology, University Hospital Tübingen, Hoppe-Seyler-Strasse 3, 72076 Tübingen, Germany
| | - Jindrich Cinatl
- Interdisciplinary Laboratory for Paediatric Tumour and Virus Research, Dr. Petra Joh Research Institute, 60529 Frankfurt am Main, Germany; (J.C.J.); (M.M.)
| | - Martin Michaelis
- Interdisciplinary Laboratory for Paediatric Tumour and Virus Research, Dr. Petra Joh Research Institute, 60529 Frankfurt am Main, Germany; (J.C.J.); (M.M.)
- School of Natural Sciences, University of Kent, Canterbury CT2 7NJ, UK
| | - Thomas Efferth
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Staudinger Weg 5, 55128 Mainz, Germany;
| | - Roman Alexander Blaheta
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
| | - Axel Haferkamp
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
| | - Eva Juengel
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (O.V.); (F.Z.); (S.D.M.); (K.S.S.); (M.P.B.); (I.T.); (R.A.B.); (A.H.)
| |
Collapse
|
2
|
Mungo C, Sorgi K, Misiko B, Cheserem C, Rahangdale L, Githongo G, Ogollah C, Omoto J, Plesa M, Zamboni W. Phase I study on the pharmacokinetics of intravaginal, self-administered artesunate vaginal pessaries among women in Kenya. PLoS One 2025; 20:e0316334. [PMID: 40203246 PMCID: PMC11981651 DOI: 10.1371/journal.pone.0316334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/03/2024] [Indexed: 04/11/2025] Open
Abstract
OBJECTIVES The primary objective of this study is to investigate the pharmacokinetics of Artesunate (AS) and its active metabolite, dihydroartemisinin (DHA) following intravaginal use at the dosing and frequency intended for cervical precancer treatment. A secondary objective is to assess safety among study participants. METHODS We are conducting a single-arm, phase I trial with a sample size of 12 female volunteers. Participants will self-administer artesunate vaginal pessaries in the study clinic daily for 5 consecutive days. Participants will have their blood drawn prior to receiving the first dose of artesunate on day one of the study and then will receive 8 blood draws on study day five, prior to artesunate administration and at 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 6 hours, and 8 hours after pessary administration. Pharmacokinetic parameters of artesunate and DHA will be calculated by way of quantitative analysis of with determination of maximum concentration (Cmax), time to Cmax (Tmax), area under the serum concentration versus time curve (AUC), apparent clearance, and elimination half-life (t1/2).
Collapse
Affiliation(s)
- Chemtai Mungo
- Department of Obstetrics and Gynecology, University of North Carolina Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Katherine Sorgi
- Department of Obstetrics and Gynecology, University of North Carolina Chapel Hill, Chapel Hill, North Carolina, United States of America
| | | | | | - Lisa Rahangdale
- Department of Obstetrics and Gynecology, University of North Carolina Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, United States of America
| | | | | | - Jackton Omoto
- Department of Obstetrics and Gynecology, Maseno University School of Medicine, Kisumu, Kenya
| | - Mihaela Plesa
- Frantz Medical Development Ltd, Mentor, Ohio, United States of America
| | - William Zamboni
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
3
|
Mungo C, Sorgi K, Misiko B, Cheserem C, Rahangdale L, Githongo G, Ogollah C, Omoto J, Plesa M, Zamboni W. Phase I study on the pharmacokinetics of intravaginal, self-administered artesunate vaginal pessaries among women in Kenya. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.07.08.24309596. [PMID: 39148845 PMCID: PMC11326355 DOI: 10.1101/2024.07.08.24309596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Cervical cancer remains a significant global health issue, especially in low- and middle-income countries (LMICs), where access to prevention and treatment is limited and women are at a higher risk of cervical cancer. Artesunate, a widely available drug used to treat malaria, has shown promise in treating human papillomavirus (HPV)-associated anogenital lesions including high-grade cervical precancer, in a recent Phase I studies in the United States. Data on the pharmacokinetics of artesunate following intravaginal use, and its implications on malaria resistance, are lacking. Objectives The primary objective of this study is to investigate the pharmacokinetics of Artesunate (AS) and its active metabolite, dihydroartemisinin (DHA) following intravaginal use at the dosing and frequency intended for cervical precancer treatment. A secondary objective is to assess safety among study participants. Methods We are conducting a single-arm, phase I trial with a sample size of 12 female volunteers. Participants will self-administer artesunate vaginal pessaries in the study clinic daily for 5 consecutive days. Participants will have their blood drawn prior to receiving the first dose of artesunate on day one of the study and then will receive 8 blood draws on study day five, prior to artesunate administration and at 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 6 hours, and 8 hours after pessary administration. Pharmacokinetic parameters of artesunate and DHA will be calculated by way of quantitative analysis of with determination of maximum concentration (Cmax), time to Cmax (Tmax), area under the serum concentration versus time curve (AUC), apparent clearance, and elimination half-life (t1/2).
Collapse
Affiliation(s)
- Chemtai Mungo
- Department of Obstetrics and Gynecology, University of North Carolina Chapel Hill, 321 S Columbia St, Chapel Hill, North Carolina, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, 450 West Dr, Chapel Hill, North Carolina, 27599, USA
| | - Katherine Sorgi
- Department of Obstetrics and Gynecology, University of North Carolina Chapel Hill, 321 S Columbia St, Chapel Hill, North Carolina, 27599, USA
| | - Brenda Misiko
- Kenya Medical Research Institute, Busia Rd, Kisumu, Kenya
| | | | - Lisa Rahangdale
- Department of Obstetrics and Gynecology, University of North Carolina Chapel Hill, 321 S Columbia St, Chapel Hill, North Carolina, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, 450 West Dr, Chapel Hill, North Carolina, 27599, USA
| | | | | | - Jackton Omoto
- Department of Obstetrics and Gynecology, Maseno University School of Medicine, P.O, Kisumu, Kenya
| | - Mihaela Plesa
- Frantz Medical Development Ltd, 7740 Metric Dr, Mentor, OH, 44060
| | - William Zamboni
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, 450 West Dr, Chapel Hill, North Carolina, 27599, USA
| |
Collapse
|
4
|
Yan HX, Zhang YZ, Niu YQ, Wang YW, Liu LH, Tang YP, Huang JM, Leung ELH. Investigating the interaction between calcium signaling and ferroptosis for novel cancer treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 137:156377. [PMID: 39798340 DOI: 10.1016/j.phymed.2025.156377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/02/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Drug resistance in cancer is steadily rising, making the development of new therapeutic targets increasingly critical for improving treatment outcomes. PURPOSE The mutual regulation of ions is essential for cell growth. Based on this concept, ion interference strategies offer a highly effective approach for cancer treatment. Calcium ions (Ca2+), as major second messengers, are closely associated with ion exchange and homeostasis. Disruptions in this balance can lead to cell death. However, while iron ions are also crucial, the connection between Ca2+and iron-induced cell death (ferroptosis) has not been well established. Therefore, this study suggests that Ca2+ may play a role in the induction of ferroptosis, presenting a novel and efficient target for cancer therapy. STUDY DESIGN PubMed, Google Scholar, and Web of Science databases were systematically searched for articles published in the past 15 years on the mechanisms of calcium ion-induced ferroptosis in cancer and related drugs. RESULTS The analysis highlights how Ca2+regulate ferroptosis. The mechanisms by which Ca2+influence ferroptosis are summarized based on existing literature, and relevant drugs that act on Ca2+/ferroptosis axis are outlined. CONCLUSION Ca2+ regulate ferroptosis primarily through the modulation of reactive oxygen species (ROS) and glutathione (GSH) levels, a mechanism that applies to a wide range of cancer cells as well as paracancerous and normal cells in cancer treatment. Furthermore, plant-derived active compounds exhibit potent anticancer properties and often act on the Ca2+/ferroptosis axis. These natural compounds could play a significant role in the development of new cancer treatment strategies.
Collapse
Affiliation(s)
- Hao-Xin Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), PR China
| | - Yi-Zhong Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), PR China
| | - Yu-Qing Niu
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), PR China
| | - Yu-Wei Wang
- Key Laboratory of Shanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shanxi University of Chinese Medicine, Xianyang 712046, Shanxi Province, PR China
| | - Li-Hua Liu
- Economics and Management Yanbian University, Yanji, PR China
| | - Yu-Ping Tang
- Key Laboratory of Shanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shanxi University of Chinese Medicine, Xianyang 712046, Shanxi Province, PR China.
| | - Ju-Min Huang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), PR China.
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), PR China.
| |
Collapse
|
5
|
Mungo C, Sorgi K, Hoch C, Tang J, Rahangdale L, Omoto J. WITHDRAWN: Intravaginal artesunate pessaries for treatment of cervical intraepithelial neoplasia 2/3 among HIV-positive and HIV-negative women in Kenya: Study protocol for a pilot trial. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.27.24309586. [PMID: 39228723 PMCID: PMC11370548 DOI: 10.1101/2024.06.27.24309586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The authors have withdrawn this manuscript because the article was posted prior to consultation from all relevant collaborators. If you have any questions, please contact the corresponding author.
Collapse
|
6
|
Tiwari MK, Goslinski T. Searching for the Holy Grail - Highly Potent Bridged Endoperoxides for Targeted Cancer Therapy. Bioorg Chem 2024; 153:107893. [PMID: 39454496 DOI: 10.1016/j.bioorg.2024.107893] [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: 07/31/2024] [Revised: 09/08/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024]
Abstract
The International Agency for Research on Cancer (IARC) recently estimated the global cancer burden in 2050. The statistics are startling, with a 77% hike and 35 million new cancer cases per year. The present discoveries have recommended plant-derived bridged endoperoxides or artemisinin-based semisynthetic analogues as safe, well-tolerated and powerful substitutes that could be effectively utilized as a warhead to fight against global enemies like cancer. In addition, artemisinin-based drug repositioning crucially can reduce overriding drug development expenditures and establish accessibility of approved drugs with low risk to patients. Hence, the present review article provides a comprehensive account of the recent chemical and synthetic advancement of diverse cytotoxic artemisinin derivatives such as C(10)-O, C, N, S linked artemisinin analogues, artemisinin-derived metal complexes, artemisinin-derived hybrids/conjugates with other pharmaceutically active substances, and artemisinin-derived dimers, trimers and tetramers perceived during the last three decades (1997-2024). Moreover, the current preclinical and clinical anticancer application prospects of artemisinin derivatives with other defined drugs and their utilization in combination therapy and also nanoformulation approaches for targeted drug delivery have been discussed.
Collapse
Affiliation(s)
- Mohit K Tiwari
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, ul. Rokietnicka 3, 60-806, Poznań, Poland.
| | - Tomasz Goslinski
- Chair and Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, ul. Rokietnicka 3, 60-806, Poznań, Poland
| |
Collapse
|
7
|
Kang DW, Kim JH, Kim KM, Cho SJ, Choi GW, Cho HY. Inter-Species Pharmacokinetic Modeling and Scaling for Drug Repurposing of Pyronaridine and Artesunate. Int J Mol Sci 2024; 25:6998. [PMID: 39000107 PMCID: PMC11241507 DOI: 10.3390/ijms25136998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Even though several new targets (mostly viral infection) for drug repurposing of pyronaridine and artesunate have recently emerged in vitro and in vivo, inter-species pharmacokinetic (PK) data that can extend nonclinical efficacy to humans has not been reported over 30 years of usage. Since extrapolation of animal PK data to those of humans is essential to predict clinical outcomes for drug repurposing, this study aimed to investigate inter-species PK differences in three animal species (hamster, rat, and dog) and to support clinical translation of a fixed-dose combination of pyronaridine and artesunate. PK parameters (e.g., steady-state volume of distribution (Vss), clearance (CL), area under the concentration-time curve (AUC), mean residence time (MRT), etc.) of pyronaridine, artesunate, and dihydroartemisinin (an active metabolite of artesunate) were determined by non-compartmental analysis. In addition, one- or two-compartment PK modeling was performed to support inter-species scaling. The PK models appropriately described the blood concentrations of pyronaridine, artesunate, and dihydroartemisinin in all animal species, and the estimated PK parameters in three species were integrated for inter-species allometric scaling to predict human PKs. The simple allometric equation (Y = a × Wb) well explained the relationship between PK parameters and the actual body weight of animal species. The results from the study could be used as a basis for drug repurposing and support determining the effective dosage regimen for new indications based on in vitro/in vivo efficacy data and predicted human PKs in initial clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | - Hea-Young Cho
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea; (D.W.K.); (J.H.K.); (K.M.K.); (S.-j.C.); (G.-W.C.)
| |
Collapse
|
8
|
Chuang YT, Yen CY, Chien TM, Chang FR, Tsai YH, Wu KC, Tang JY, Chang HW. Ferroptosis-Regulated Natural Products and miRNAs and Their Potential Targeting to Ferroptosis and Exosome Biogenesis. Int J Mol Sci 2024; 25:6083. [PMID: 38892270 PMCID: PMC11173094 DOI: 10.3390/ijms25116083] [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: 04/28/2024] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Ferroptosis, which comprises iron-dependent cell death, is crucial in cancer and non-cancer treatments. Exosomes, the extracellular vesicles, may deliver biomolecules to regulate disease progression. The interplay between ferroptosis and exosomes may modulate cancer development but is rarely investigated in natural product treatments and their modulating miRNAs. This review focuses on the ferroptosis-modulating effects of natural products and miRNAs concerning their participation in ferroptosis and exosome biogenesis (secretion and assembly)-related targets in cancer and non-cancer cells. Natural products and miRNAs with ferroptosis-modulating effects were retrieved and organized. Next, a literature search established the connection of a panel of ferroptosis-modulating genes to these ferroptosis-associated natural products. Moreover, ferroptosis-associated miRNAs were inputted into the miRNA database (miRDB) to bioinformatically search the potential targets for the modulation of ferroptosis and exosome biogenesis. Finally, the literature search provided a connection between ferroptosis-modulating miRNAs and natural products. Consequently, the connections from ferroptosis-miRNA-exosome biogenesis to natural product-based anticancer treatments are well-organized. This review sheds light on the research directions for integrating miRNAs and exosome biogenesis into the ferroptosis-modulating therapeutic effects of natural products on cancer and non-cancer diseases.
Collapse
Affiliation(s)
- Ya-Ting Chuang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
| | - Tsu-Ming Chien
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan;
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Urology, Kaohsiung Gangshan Hospital, Kaohsiung Medical University, Kaohsiung 820111, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Yi-Hong Tsai
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung 907101, Taiwan;
| | - Kuo-Chuan Wu
- Department of Computer Science and Information Engineering, National Pingtung University, Pingtung 900391, Taiwan;
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
9
|
Cui J, Cai X, Qian R, Wu L, Qi X, Cao J, Shen S. Tween 80 Micelles Loaded with Fe 3O 4 Nanoparticles and Artemisinin for Combined Oxygen-Independent Ferroptosis Therapy of Cancer. Pharmaceutics 2024; 16:639. [PMID: 38794301 PMCID: PMC11124998 DOI: 10.3390/pharmaceutics16050639] [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: 04/11/2024] [Revised: 04/28/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Artemisinin has an endoperoxide bridge structure, which can be cleaved by ferrous ions to generate various carbonyl radicals in an oxygen-independent manner, highlighting its potential for treating hypoxic tumors. In our study, we fabricated Tween 80 micelles loaded with Fe3O4 nanoparticles and artemisinin for cancer therapy. The synthesized Fe3O4 nanoparticles and drug-loaded micelles have particle sizes of about 5 nm and 80 nm, respectively, both exhibiting excellent dispersibility and stability. After uptake by MCF-7 cells, drug-loaded micelles release Fe2+ and ART into the cytoplasm, effectively inducing the generation of reactive oxygen species (ROS) in hypoxic conditions, thereby enhancing toxicity against cancer cells. In vitro and in vivo studies have demonstrated that ART and Fe3O4 nanoparticles are encapsulated in Tween 80 to form micelles, which effectively prevent premature release during circulation in the body. Although free ART and Fe3O4 nanoparticles can inhibit tumor growth, TW80-Fe3O4-ART micelles demonstrate a more pronounced inhibitory effect, with a tumor suppression rate of up to 85%. A novel strategy based on artemisinin and ferroptosis is thus offered, holding a favorable prospect for hypoxic cancer therapy.
Collapse
Affiliation(s)
- Junming Cui
- Department of Pharmacy, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; (J.C.); (X.C.); (R.Q.)
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang 212013, China; (X.Q.); (J.C.)
| | - Xinxi Cai
- Department of Pharmacy, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; (J.C.); (X.C.); (R.Q.)
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang 212013, China; (X.Q.); (J.C.)
| | - Rui Qian
- Department of Pharmacy, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; (J.C.); (X.C.); (R.Q.)
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang 212013, China; (X.Q.); (J.C.)
| | - Lin Wu
- Department of Pharmacy, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; (J.C.); (X.C.); (R.Q.)
| | - Xueyong Qi
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang 212013, China; (X.Q.); (J.C.)
| | - Jin Cao
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang 212013, China; (X.Q.); (J.C.)
| | - Song Shen
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang 212013, China; (X.Q.); (J.C.)
| |
Collapse
|
10
|
Ding W, Su Y, Mo J, Sun D, Cao C, Zhang X, Wang Y. Novel artemisinin derivative P31 inhibits VEGF-induced corneal neovascularization through AKT and ERK1/2 pathways. Heliyon 2024; 10:e29984. [PMID: 38699723 PMCID: PMC11063438 DOI: 10.1016/j.heliyon.2024.e29984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
Corneal neovascularization (CoNV)is a major cause of blindness in many ocular diseases. Substantial evidence indicates that vascular endothelial growth factor (VEGF) plays an important role in the pathogenesis of corneal neovascularization. Previous evidence showed that artemisinin may inhibit angiogenesis through down regulation of the VEGF receptors. We designed and synthesized artemisinin derivatives, and validated their inhibitory effect on neovascularization in cell and animal models, and explored the mechanisms by which they exert an inhibitory effect on CoNV. Among these derivatives, P31 demonstrated significant anti-angiogenic effects in vivo and in vitro. Besides, P31 inhibited VEGF-induced HUVECs angiogenesis and neovascularization in rabbit model via AKT and ERK pathways. Moreover, P31 alleviated angiogenic and inflammatory responses in suture rabbit cornea. In conclusion, as a novel artemisinin derivative, P31 attenuates corneal neovascularization and has a promising application in ocular diseases.
Collapse
Affiliation(s)
- Wen Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Engineering Research Center for Ophthalmic Drug Creation and Evaluation, Guangzhou, 510060, China
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Yingxue Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Engineering Research Center for Ophthalmic Drug Creation and Evaluation, Guangzhou, 510060, China
| | - Jianshan Mo
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Danyuan Sun
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Chen Cao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Engineering Research Center for Ophthalmic Drug Creation and Evaluation, Guangzhou, 510060, China
| | - Xiaolei Zhang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Yandong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Engineering Research Center for Ophthalmic Drug Creation and Evaluation, Guangzhou, 510060, China
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| |
Collapse
|
11
|
Jochims F, Strohm R, von Montfort C, Wenzel CK, Klahm N, Kondadi AK, Stahl W, Reichert AS, Brenneisen P. The Antimalarial Drug Artesunate Mediates Selective Cytotoxicity by Upregulating HO-1 in Melanoma Cells. Biomedicines 2023; 11:2393. [PMID: 37760834 PMCID: PMC10525565 DOI: 10.3390/biomedicines11092393] [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: 08/03/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Despite great efforts to develop new therapeutic strategies to combat melanoma, the prognosis remains rather poor. Artesunate (ART) is an antimalarial drug displaying anti-cancer effects in vitro and in vivo. In this in vitro study, we investigated the selectivity of ART on melanoma cells. Furthermore, we aimed to further elucidate the mechanism of the drug with a focus on the role of iron, the induction of oxidative stress and the implication of the enzyme heme oxygenase 1 (HO-1). ART treatment decreased the cell viability of A375 melanoma cells while it did not affect the viability of normal human dermal fibroblasts, used as a model for normal (healthy) cells. ART's toxicity was shown to be dependent on intracellular iron and the drug induced high levels of oxidative stress as well as upregulation of HO-1. Melanoma cells deficient in HO-1 or treated with a HO-1 inhibitor were less sensitive towards ART. Taken together, our study demonstrates that ART induces oxidative stress resulting in the upregulation of HO-1 in melanoma cells, which subsequently triggers the effect of ART's own toxicity. This new finding that HO-1 is involved in ART-mediated toxicity may open up new perspectives in cancer therapy.
Collapse
Affiliation(s)
- Finn Jochims
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (R.S.); (C.-K.W.); (N.K.); (A.K.K.); (W.S.); (A.S.R.)
| | | | | | | | | | | | | | | | - Peter Brenneisen
- Institute of Biochemistry and Molecular Biology I, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (R.S.); (C.-K.W.); (N.K.); (A.K.K.); (W.S.); (A.S.R.)
| |
Collapse
|
12
|
Zeng ZW, Chen D, Chen L, He B, Li Y. A comprehensive overview of Artemisinin and its derivatives as anticancer agents. Eur J Med Chem 2023; 247:115000. [PMID: 36538859 DOI: 10.1016/j.ejmech.2022.115000] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/20/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022]
Abstract
Artemisinin is the crucial ingredient of artemisia annua, a traditional Chinese medicine used for the therapy of malaria in China for hundreds of years. In recent years, the anticancer properties of artemisinin and its derivatives have also been reported. This review has summarized the research and development of artemisinin and its derivatives as anticancer agents, which included both natural and synthetic monomers as well as their dimers. In addition, it highlights the antitumor effects of artemisinin and its derivatives after site-modification or after transformation to a nano-delivery system. Moreover, we have further explored their potential mechanisms of action and also discussed the clinical trials of ARTs used to treat cancer, which will facilitate in further development of novel anticancer drugs based on the scaffold of artemisinin.
Collapse
Affiliation(s)
- Zi-Wei Zeng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, School of Basic Medical Science, Guizhou Medical University, Guiyang, 550004, China
| | - Di Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, School of Basic Medical Science, Guizhou Medical University, Guiyang, 550004, China
| | - Lei Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, School of Basic Medical Science, Guizhou Medical University, Guiyang, 550004, China
| | - Bin He
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, School of Basic Medical Science, Guizhou Medical University, Guiyang, 550004, China.
| | - Yan Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmacy, School of Basic Medical Science, Guizhou Medical University, Guiyang, 550004, China.
| |
Collapse
|
13
|
Wei W, Cao B, Xu D, Liu Y, Zhang X, Wang Y. Development and validation of a prognostic prediction model for iron metabolism-related genes in patients with pancreatic adenocarcinoma. Front Genet 2023; 13:1058062. [PMID: 36685915 PMCID: PMC9846079 DOI: 10.3389/fgene.2022.1058062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/30/2022] [Indexed: 01/05/2023] Open
Abstract
Background: Pancreatic adenocarcinoma (PAAD) is one of the most aggressive tumors of the digestive tract, with low surgical resection rate and insensitivity to radiotherapy and chemotherapy. Existing evidence suggests that regulation of ferroptosis can induce PAAD cell death, inhibit tumor growth, and may synergistically improve the sensitivity of other antitumor drugs. However, there is little of systematic research on iron metabolism-related genes in PAAD. In this study, a risk-score system of PAAD iron metabolism-related genes was designed and tested, and verified to be robust. Materials and Methods: The TCGA database was used to download 177 PAAD patients' message RNA (mRNA) expression profiles and clinical characteristics. By identifying dysregulated iron metabolism-related genes between PAAD related tissues and adjacent normal tissues, univariate Cox proportional hazards regression and LASSO regression algorithm were used to establish prognostic risk-score system and construct nomogram to estimate the 1-, 2-, 3-year survival in PAAD patients. Finally, selected genes were validated by quantitative PCR (q-PCR). Results: A 9-gene related to iron metabolism risk-score system of PAAD was constructed and validated. The clinicopathological characteristics of age, histologic grade, pathologic stage, T stage, residual tumor, and primary therapy outcome were all worse in patients with a higher risk-score. Further, immunohistochemistry results of SLC2A1, MBOAT2, XDH, CTSE, MOCOS, and ATP6V0A4 confirmed that patients with higher expression are more malignant. Then, a nomogram with 9-gene risk score system as a separate clinical factor was utilized to foretell the 1-, 2-, 3-year overall survival rate of PAAD patients. Results of q-PCR showed that 8 of the 9 genes screened were significantly up-regulated in at least one PAAD cell line, and one gene was significantly down-regulated in three PAAD cell lines. Conclusion: To conclude, we generated a nine-gene system linked to iron metabolism as an independent indicator for predicting PAAD prognosis, therefore presenting a possible prognostic biomarker and potential treatment targets for PAAD.
Collapse
Affiliation(s)
- Wenhan Wei
- Department of Gastroenterology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,China State Key Laboratory of CAD&CG, Zhejiang University, Hangzhou, China
| | - Bin Cao
- Department of Pharmacy, First Affiliated Hospital, Huzhou University, Huzhou, China
| | - Dongchao Xu
- Department of Gastroenterology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,Hangzhou Institute of Digestive Diseases, Hangzhou, China,Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| | - Yusheng Liu
- China State Key Laboratory of CAD&CG, Zhejiang University, Hangzhou, China
| | - Xiaofeng Zhang
- Department of Gastroenterology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,Hangzhou Institute of Digestive Diseases, Hangzhou, China,Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China,*Correspondence: Xiaofeng Zhang, ; Yu Wang,
| | - Yu Wang
- Department of Gastroenterology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,Hangzhou Institute of Digestive Diseases, Hangzhou, China,Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China,*Correspondence: Xiaofeng Zhang, ; Yu Wang,
| |
Collapse
|
14
|
Greene CJ, Attwood K, Sharma NJ, Balderman B, Deng R, Muhitch JB, Smith GJ, Gross KW, Xu B, Kauffman EC. Iron accumulation typifies renal cell carcinoma tumorigenesis but abates with pathological progression, sarcomatoid dedifferentiation, and metastasis. Front Oncol 2022; 12:923043. [PMID: 35992801 PMCID: PMC9389085 DOI: 10.3389/fonc.2022.923043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
Iron is a potent catalyst of oxidative stress and cellular proliferation implicated in renal cell carcinoma (RCC) tumorigenesis, yet it also drives ferroptosis that suppresses cancer progression and represents a novel therapeutic target for advanced RCC. The von Hippel Lindau (VHL)/hypoxia-inducible factor-α (HIF-α) axis is a major regulator of cellular iron, and its inactivation underlying most clear cell (cc) RCC tumors introduces both iron dependency and ferroptosis susceptibility. Despite the central role for iron in VHL/HIF-α signaling and ferroptosis, RCC iron levels and their dynamics during RCC initiation/progression are poorly defined. Here, we conducted a large-scale investigation into the incidence and prognostic significance of total tissue iron in ccRCC and non-ccRCC patient primary tumor cancer cells, tumor microenvironment (TME), metastases and non-neoplastic kidneys. Prussian Blue staining was performed to detect non-heme iron accumulation in over 1600 needle-core sections across multiple tissue microarrays. We found that RCC had significantly higher iron staining scores compared with other solid cancers and, on average, >40 times higher than adjacent renal epithelium. RCC cell iron levels correlated positively with TME iron levels and inversely with RCC levels of the main iron uptake protein, transferrin receptor 1 (TfR1/TFRC/CD71). Intriguingly, RCC iron levels, including in the TME, decreased significantly with pathologic (size/stage/grade) progression, sarcomatoid dedifferentiation, and metastasis, particularly among patients with ccRCC, despite increasing TfR1 levels, consistent with an increasingly iron-deficient tumor state. Opposite to tumor iron changes, adjacent renal epithelial iron increased significantly with RCC/ccRCC progression, sarcomatoid dedifferentiation, and metastasis. Lower tumor iron and higher renal epithelial iron each predicted significantly shorter ccRCC patient metastasis-free survival. In conclusion, iron accumulation typifies RCC tumors but declines toward a relative iron-deficient tumor state during progression to metastasis, despite precisely opposite dynamics in adjacent renal epithelium. These findings raise questions regarding the historically presumed selective advantage for high iron during all phases of cancer evolution, suggesting instead distinct tissue-specific roles during RCC carcinogenesis and early tumorigenesis versus later progression. Future study is warranted to determine how the relative iron deficiency of advanced RCC contributes to ferroptosis resistance and/or introduces a heightened susceptibility to iron deprivation that might be therapeutically exploitable.
Collapse
Affiliation(s)
- Christopher J. Greene
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
- Department of Biological Sciences, University at Buffalo, Buffalo, NY, United States
| | - Kristopher Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Nitika J. Sharma
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Benjamin Balderman
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Rongia Deng
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Jason B. Muhitch
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Gary J. Smith
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Kenneth W. Gross
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Bo Xu
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Eric C. Kauffman
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
- Department of Cancer Genetics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
- *Correspondence: Eric C. Kauffman,
| |
Collapse
|
15
|
The Potential Mechanisms by which Artemisinin and Its Derivatives Induce Ferroptosis in the Treatment of Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1458143. [PMID: 35028002 PMCID: PMC8752222 DOI: 10.1155/2022/1458143] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/02/2021] [Accepted: 11/23/2021] [Indexed: 02/06/2023]
Abstract
Artemisinin (ART) is a bioactive molecule derived from the Chinese medicinal plant Artemisia annua (Asteraceae). ART and artemisinin derivatives (ARTs) have been effectively used for antimalaria treatment. The structure of ART is composed of a sesquiterpene lactone, including a peroxide internal bridge that is essential for its activity. In addition to their well-known antimalarial effects, ARTs have been shown recently to resist a wide range of tumors. The antineoplastic mechanisms of ART mainly include cell cycle inhibition, inhibition of tumor angiogenesis, DNA damage, and ferroptosis. In particular, ferroptosis is a novel nonapoptotic type of programmed cell death. However, the antitumor mechanisms of ARTs by regulating ferroptosis remain unclear. Through this review, we focus on the potential antitumor function of ARTs by acting on ferroptosis, including the regulation of iron metabolism, generation of reactive oxygen species (ROS), and activation of endoplasmic reticulum stress (ERS). This article systematically reviews the recent progress in ferroptosis research and provides a basis for ARTs as an anticancer drug in clinical practice.
Collapse
|
16
|
Ma Z, Woon CYN, Liu CG, Cheng JT, You M, Sethi G, Wong ALA, Ho PCL, Zhang D, Ong P, Wang L, Goh BC. Repurposing Artemisinin and its Derivatives as Anticancer Drugs: A Chance or Challenge? Front Pharmacol 2022; 12:828856. [PMID: 35035355 PMCID: PMC8758560 DOI: 10.3389/fphar.2021.828856] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 12/13/2021] [Indexed: 11/30/2022] Open
Abstract
Cancer has become a global health problem, accounting for one out of six deaths. Despite the recent advances in cancer therapy, there is still an ever-growing need for readily accessible new therapies. The process of drug discovery and development is arduous and takes many years, and while it is ongoing, the time for the current lead compounds to reach clinical trial phase is very long. Drug repurposing has recently gained significant attention as it expedites the process of discovering new entities for anticancer therapy. One such potential candidate is the antimalarial drug, artemisinin that has shown anticancer activities in vitro and in vivo. In this review, major molecular and cellular mechanisms underlying the anticancer effect of artemisinin and its derivatives are summarised. Furthermore, major mechanisms of action and some key signaling pathways of this group of compounds have been reviewed to explore potential targets that contribute to the proliferation and metastasis of tumor cells. Despite its established profile in malaria treatment, pharmacokinetic properties, anticancer potency, and current formulations that hinder the clinical translation of artemisinin as an anticancer agent, have been discussed. Finally, potential solutions or new strategies are identified to overcome the bottlenecks in repurposing artemisinin-type compounds as anticancer drugs.
Collapse
Affiliation(s)
- Zhaowu Ma
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Clariis Yi-Ning Woon
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Chen-Guang Liu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Jun-Ting Cheng
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Mingliang You
- Hangzhou Cancer Institute, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, China.,Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Andrea Li-Ann Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Paul Chi-Lui Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Daping Zhang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Peishi Ong
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Boon-Cher Goh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| |
Collapse
|
17
|
Nazem S, Mirzaei A, Tavakoli-Yaraki M, Najafi T, Salami S, Bashash D, Shahsavari Z. Evaluation of the expression level of apoptosis and ferroptosis cell death pathways genes in patients with osteosarcoma. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
18
|
Gu C, She X, Zhou C, Su T, He S, Meng C, Gu Q, Luo D, Zheng Z, Qiu Q. Dihydroartemisinin ameliorates retinal vascular dysfunction in diabetes mellitus via the FASN/Kmal-mTOR/SREBP1 feedback loop. Pharmacol Res 2021; 174:105871. [PMID: 34619345 DOI: 10.1016/j.phrs.2021.105871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/21/2021] [Accepted: 09/02/2021] [Indexed: 02/07/2023]
Abstract
Microvascular dysfunction is the primary aetiology of visual impairment caused by diabetic retinopathy (DR). Dihydroartemisinin (DHA), the active metabolite of the antimalarials artemisinins, exhibits antiangiogenic properties in numerous diseases. Here, we investigated the function and mechanisms of DHA as a vasculoprotective agent in DR. DHA exerted its protective effect on vascular injuries in diabetic mice and inhibited cell proliferation and tube formation in human retinal microvascular endothelial cells by decreasing the level of fatty acid synthase (FASN), enhancing the malonylation of mechanistic target of rapamycin (mTOR) at lysine 1218 (K1218) and attenuating the activation of mTOR complex 1 (mTORC1). Impressively, a chemosynthetic small interfering RNA against FASN and mutagenesis of K1218 of mTOR showed therapeutic potential in suppressing cell proliferation and tube formation induced by high glucose. Notably, suppression of mTORC1 kinase activity further inhibited FASN by reducing p70S6K phosphorylation to subsequently reduce the expression of sterol regulatory element binding protein 1, which interacted directly with the FASN promoter at nucleotide positions -64 and -55. In conclusion, our study elucidated the promising effects of FASN and malonylation on vascular injuries of DR and indicated the great potential of DHA as a therapeutic approach.
Collapse
Affiliation(s)
- Chufeng Gu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Xinping She
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Chuandi Zhou
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Tong Su
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Shuai He
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Chunren Meng
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Qing Gu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Dawei Luo
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Zhi Zheng
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China.
| | - Qinghua Qiu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China; Department of Ophthalmology, Shigatse People's Hospital, Shigatse, Xizang, PR China.
| |
Collapse
|
19
|
Targeting Drug Chemo-Resistance in Cancer Using Natural Products. Biomedicines 2021; 9:biomedicines9101353. [PMID: 34680470 PMCID: PMC8533186 DOI: 10.3390/biomedicines9101353] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer is one of the leading causes of death globally. The development of drug resistance is the main contributor to cancer-related mortality. Cancer cells exploit multiple mechanisms to reduce the therapeutic effects of anticancer drugs, thereby causing chemotherapy failure. Natural products are accessible, inexpensive, and less toxic sources of chemotherapeutic agents. Additionally, they have multiple mechanisms of action to inhibit various targets involved in the development of drug resistance. In this review, we have summarized the basic research and clinical applications of natural products as possible inhibitors for drug resistance in cancer. The molecular targets and the mechanisms of action of each natural product are also explained. Diverse drug resistance biomarkers were sensitive to natural products. P-glycoprotein and breast cancer resistance protein can be targeted by a large number of natural products. On the other hand, protein kinase C and topoisomerases were less sensitive to most of the studied natural products. The studies discussed in this review will provide a solid ground for scientists to explore the possible use of natural products in combination anticancer therapies to overcome drug resistance by targeting multiple drug resistance mechanisms.
Collapse
|
20
|
Xu S, Wang Z, Ye J, Mei S, Zhang J. Identification of Iron Metabolism-Related Genes as Prognostic Indicators for Lower-Grade Glioma. Front Oncol 2021; 11:729103. [PMID: 34568059 PMCID: PMC8458946 DOI: 10.3389/fonc.2021.729103] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
Lower-grade glioma (LGG) is characterized by genetic and transcriptional heterogeneity, and a dismal prognosis. Iron metabolism is considered central for glioma tumorigenesis, tumor progression and tumor microenvironment, although key iron metabolism-related genes are unclear. Here we developed and validated an iron metabolism-related gene signature LGG prognosis. RNA-sequence and clinicopathological data from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA) were downloaded. Prognostic iron metabolism-related genes were screened and used to construct a risk-score model via differential gene expression analysis, univariate Cox analysis, and the Least Absolute Shrinkage and Selection Operator (LASSO)-regression algorithm. All LGG patients were stratified into high- and low-risk groups, based on the risk score. The prognostic significance of the risk-score model in the TCGA and CGGA cohorts was evaluated with Kaplan-Meier (KM) survival and receiver operating characteristic (ROC) curve analysis. Risk- score distributions in subgroups were stratified by age, gender, the World Health Organization (WHO) grade, isocitrate dehydrogenase 1 (IDH1) mutation status, the O6-methylguanine-DNA methyl-transferase (MGMT) promoter-methylation status, and the 1p/19q co-deletion status. Furthermore, a nomogram model with a risk score was developed, and its predictive performance was validated with the TCGA and CGGA cohorts. Additionally, the gene set enrichment analysis (GSEA) identified signaling pathways and pathological processes enriched in the high-risk group. Finally, immune infiltration and immune checkpoint analysis were utilized to investigate the tumor microenvironment characteristics related to the risk score. We identified a prognostic 15-gene iron metabolism-related signature and constructed a risk-score model. High risk scores were associated with an age of > 40, wild-type IDH1, a WHO grade of III, an unmethylated MGMT promoter, and 1p/19q non-codeletion. ROC analysis indicated that the risk-score model accurately predicted 1-, 3-, and 5-year overall survival rates of LGG patients in the both TCGA and CGGA cohorts. KM analysis showed that the high-risk group had a much lower overall survival than the low-risk group (P < 0.0001). The nomogram model showed a strong ability to predict the overall survival of LGG patients in the TCGA and CGGA cohorts. GSEA analysis indicated that inflammatory responses, tumor-associated pathways, and pathological processes were enriched in high-risk group. Moreover, a high risk score correlated with the infiltration immune cells (dendritic cells, macrophages, CD4+ T cells, and B cells) and expression of immune checkpoint (PD1, PDL1, TIM3, and CD48). Our prognostic model was based on iron metabolism-related genes in LGG, can potentially aid in LGG prognosis, and provides potential targets against gliomas.
Collapse
Affiliation(s)
- Shenbin Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- Department of Gastroenterology Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zefeng Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Juan Ye
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Shuhao Mei
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Brain Research Institute, Zhejiang University, Hangzhou, China
- Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
21
|
Geng B, Zhu Y, Yuan Y, Bai J, Dou Z, Sui A, Luo W. Artesunate Suppresses Choroidal Melanoma Vasculogenic Mimicry Formation and Angiogenesis via the Wnt/CaMKII Signaling Axis. Front Oncol 2021; 11:714646. [PMID: 34476217 PMCID: PMC8406848 DOI: 10.3389/fonc.2021.714646] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/26/2021] [Indexed: 01/14/2023] Open
Abstract
Angiogenesis and vasculogenic mimicry (VM) are considered to be the main processes to ensure tumor blood supply during the proliferation and metastasis of choroidal melanoma (CM). The traditional antimalarial drug artesunate (ART) has some potential anti-CM effects; however, the underlying mechanisms remain unclarified. Recent studies have shown that the Wnt5a/calmodulin-dependent kinase II (CaMKII) signaling pathway has a close correlation with angiogenesis and VM formation. This study demonstrated that ART eliminated VM formation by inhibiting the aforementioned signaling pathway in CM cells. The microvessel sprouting of the mouse aortic rings and the microvessel density of chicken chorioallantoic membrane (CAM) decreased significantly after ART treatment. VM formation assay and periodic acid schiff (PAS) staining revealed that ART inhibited VM formation in CM. Moreover, ART downregulated the expression levels of the angiogenesis-related proteins vascular endothelial growth factor receptor (VEGFR) 2, platelet-derived growth factor receptor (PDGFR) and vascular endothelial growth factor (VEGF) A, and VM-related proteins ephrin type-A receptor (EphA) 2 and vascular endothelial (VE)-cadherin. The expression of hypoxia-inducible factor (HIF)-1α, Wnt5a, and phosphorylated CaMKII was also downregulated after ART treatment. In addition, we further demonstrated that ART inhibited the proliferation, migration, and invasion of OCM-1 and C918 cells. Collectively, our results suggested that ART inhibited angiogenesis and VM formation of choroidal melanoma likely by regulating the Wnt5a/CaMKII signaling pathway. These findings further supported the feasibility of ART for cancer therapy.
Collapse
Affiliation(s)
- Bochao Geng
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuanzhang Zhu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yingying Yuan
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jingyi Bai
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhizhi Dou
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Aihua Sui
- Central Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenjuan Luo
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
22
|
Ozkan E, Bakar-Ates F. Ferroptosis: A Trusted Ally in Combating Drug Resistance in Cancer. Curr Med Chem 2021; 29:41-55. [PMID: 34375173 DOI: 10.2174/0929867328666210810115812] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/11/2021] [Accepted: 07/14/2021] [Indexed: 11/22/2022]
Abstract
Ferroptosis, which is an iron-dependent, non-apoptotic cell death mechanism, has recently been proposed as a novel approach in cancer treatment. Bearing distinctive features and its exclusive mechanism have put forward the potential therapeutic benefit of triggering this newly discovered form of cell death. Numerous studies have indicated that apoptotic pathways are often deactivated in resistant cells, leading to a failure in therapy. Hence, alternative strategies to promote cell death are required. Mounting evidence suggests that drug-resistant cancer cells are particularly sensitive to ferroptosis. Given that cancer cells consume a higher amount of iron than healthy ones, ferroptosis not only stands as an excellent alternative to trigger cell death and reverse drug-resistance, but also provides selectivity in therapy. This review focuses specifically on overcoming drug-resistance in cancer through activating ferroptotic pathways and brings together the relevant chemotherapeutics-based and nanotherapeutics-based studies to offer a perspective for researchers regarding the potential use of this mechanism in developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Erva Ozkan
- Ankara University, Faculty of Pharmacy, Department of Biochemistry, Ankara, Turkey
| | - Filiz Bakar-Ates
- Ankara University, Faculty of Pharmacy, Department of Biochemistry, Ankara, Turkey
| |
Collapse
|
23
|
Drug Repurposing to Identify a Synergistic High-Order Drug Combination to Treat Sunitinib-Resistant Renal Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13163978. [PMID: 34439134 PMCID: PMC8391235 DOI: 10.3390/cancers13163978] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary In this study, drug combination screening was used to design a multidrug combination consisting of repurposed drugs to treat sunitinib-resistant clear cell renal cell carcinoma. In the frame of this project, the multidrug combination has been optimized and validated and an insight into the mechanism of action is given. The multidrug combinations significantly altered the transcription of genes related to apoptosis and metabolic pathways. Further analysis of the metabolism revealed strong upregulation of the presence of sphingolipids after multidrug combination treatment. Final evaluation for translation of the multidrug combination in ex vivo organoid-like cultures demonstrated significant anti-cancer efficacy. Abstract Repurposed drugs have been evaluated for the management of clear cell renal cell carcinoma (ccRCC), but only a few have influenced the overall survival of patients with advanced disease. To combine repurposed non-oncology with oncological drugs, we applied our validated phenotypic method, which consisted of a reduced experimental part and data modeling. A synergistic optimized multidrug combination (ODC) was identified to significantly reduce the energy levels in cancer remaining inactive in non-cancerous cells. The ODC consisted of Rapta-C, erlotinib, metformin and parthenolide and low doses. Molecular and functional analysis of ODC revealed a loss of adhesiveness and induction of apoptosis. Gene-expression network analysis displayed significant alterations in the cellular metabolism, confirmed by LC-MS based metabolomic analysis, highlighting significant changes in the lipid classes. We used heterotypic in vitro 3D co-cultures and ex vivo organoids to validate the activity of the ODC, maintaining an efficacy of over 70%. Our results show that repurposed drugs can be combined to target cancer cells selectively with prominent activity. The strong impact on cell adherence and metabolism indicates a favorable mechanism of action of the ODC to treat ccRCC.
Collapse
|
24
|
Repurposing of Antimicrobial Agents for Cancer Therapy: What Do We Know? Cancers (Basel) 2021; 13:cancers13133193. [PMID: 34206772 PMCID: PMC8269327 DOI: 10.3390/cancers13133193] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023] Open
Abstract
The substantial costs of clinical trials, the lengthy timelines of new drug discovery and development, along the high attrition rates underscore the need for alternative strategies for finding quickly suitable therapeutics agents. Given that most approved drugs possess more than one target tightly linked to other diseases, it encourages promptly testing these drugs in patients. Over the past decades, this has led to considerable attention for drug repurposing, which relies on identifying new uses for approved or investigational drugs outside the scope of the original medical indication. The known safety of approved drugs minimizes the possibility of failure for adverse toxicology, making them attractive de-risked compounds for new applications with potentially lower overall development costs and shorter development timelines. This latter case is an exciting opportunity, specifically in oncology, due to increased resistance towards the current therapies. Indeed, a large body of evidence shows that a wealth of non-cancer drugs has beneficial effects against cancer. Interestingly, 335 drugs are currently being evaluated in different clinical trials for their potential activities against various cancers (Redo database). This review aims to provide an extensive discussion about the anti-cancer activities exerted by antimicrobial agents and presents information about their mechanism(s) of action and stage of development/evaluation.
Collapse
|
25
|
Meng Y, Ma N, Lyu H, Wong YK, Zhang X, Zhu Y, Gao P, Sun P, Song Y, Lin L, Wang J. Recent pharmacological advances in the repurposing of artemisinin drugs. Med Res Rev 2021; 41:3156-3181. [PMID: 34148245 DOI: 10.1002/med.21837] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/27/2021] [Accepted: 05/21/2021] [Indexed: 12/18/2022]
Abstract
Artemisinins are a family of sesquiterpene lactones originally derived from the sweet wormwood (Artemisia annua). Beyond their well-characterized role as frontline antimalarial drugs, artemisinins have also received increased attention for other potential pharmaceutical effects, which include antiviral, antiparsitic, antifungal, anti-inflammatory, and anticancer activities. With concerted efforts in further preclinical and clinical studies, artemisinin-based drugs have the potential to be viable treatments for a great variety of human diseases. Here, we provide a comprehensive update on recent reports of pharmacological actions and applications of artemisinins outside of their better-known antimalarial role and highlight their potential therapeutic viability for various diseases.
Collapse
Affiliation(s)
- Yuqing Meng
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Nan Ma
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haining Lyu
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yin Kwan Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xing Zhang
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yongping Zhu
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Peng Gao
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Peng Sun
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yali Song
- Center for Reproductive Medicine, Dongguan Maternal And Child Health Care Hospital, Southern Medical University, Dongguan, China
| | - Lizhu Lin
- Oncology Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jigang Wang
- Artemisinin Research Center and the Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,Oncology Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| |
Collapse
|
26
|
Li W, Zhao L, Li Y, Zhai Z. Artesunate attenuates proliferation of epithelial cells by downregulating the NF-κB and AKT signaling pathways in benign mammary gland hyperplasia rats. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:848. [PMID: 34164482 PMCID: PMC8184455 DOI: 10.21037/atm-21-1447] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background The aim of this study was to investigate the effects of artesunate (ART) on breast epithelial cell proliferation in vitro and in vivo. Methods Immortalized human non-cancer mammary epithelial (MCF-10A) cells were used to determine the effect of ART on estrogen-induced mammary hyperplasia cells. We investigated the effect of ART on the synthesis of cyclooxygenase-2 (COX-2) and proliferating cell nuclear antigen (PCNA) in MCF-10A by treating MCF-10A 36 h with different concentrations of ART (0, 100, 200, 400 µm, n=12/group). We then investigated the effect of ART on estrogen induced COX-2, PCNA, nuclear factor-kappa B (NF-κB), and pNF-κB synthesis by treating MCF-10A with both estrogen and ART (0, 50, 100, 200 µm, n=12/group). A mammary hyperplasia model (MGH) was established in rats. All rats (n=12) were divided into 4 groups [group A: negative control (NC) + Art −; group B: NC + Art +; group C: MGH + Art −; group D: MGH + Art +] by the random number table method and the effects of ART on estradiol-induced mammary hyperplasia, fibrosis, and phosphorylation of AKT and NF-κB were studied by histopathological staining, Masson trichrome staining, immunohistochemistry (IHC), and western blotting. Results The proliferation and inflammation of mammary epithelial cells were blocked by ART (P<0.05). The phosphorylation of NF-κB induced by estradiol in MCF-10A was attenuated by ART (P<0.05). In the rat MGH, ART reduced cell proliferation and fibrosis (P<0.05) and inhibited the phosphorylation of AKT and NF-κB (P<0.05). Conclusions The drug ART inhibits estrogen-induced breast hyperplasia by blocking AKT and NFkB phosphorylation.
Collapse
Affiliation(s)
- Wei Li
- Department of Breast, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Lina Zhao
- Department of Breast, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Yiliang Li
- Department of Breast, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Zhen Zhai
- Department of Breast, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
27
|
Huang X, Ou C, Shu Y, Wang Y, Gong S, Luo R, Chen S, Wu Q, Gong C. A self-sustained nanoplatform reverses TRAIL-resistance of pancreatic cancer through coactivating of exogenous and endogenous apoptotic pathway. Biomaterials 2021; 272:120795. [PMID: 33836292 DOI: 10.1016/j.biomaterials.2021.120795] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/27/2021] [Accepted: 03/29/2021] [Indexed: 02/05/2023]
Abstract
Since the 5-year survival rate of pancreatic cancer is only 10.0%, new therapies are urgently needed. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis specifically on tumor cells, nevertheless its clinical application was seriously restricted by resistance and short in vivo half-life. Herein, a novel multifunctional R6ST protein equipped with cell penetrating peptides R6, intrinsic apoptosis inducing tetrapeptide AVPI and soluble TRAIL was designed and constructed. Then, it was recruited to prepare self-sustained nanoplatform (SSN) to reverse TRAIL-resistance of pancreatic cancer through simultaneously promoting extrinsic and intrinsic apoptotic pathway, as well to elongate circulation time. Once administrated, high tumor accumulation and cellular uptake of SSN were achieved through prolonged circulation time, targeting ability of soluble TRAIL to death receptors and positive-charged R6, and further enhanced through reversed upregulation of death receptors on TRAIL-resistant tumor cells by the cumulated artesunate released in cytoplasm as a positive feedback loop. Furthermore, this loop simultaneously promoted extrinsic apoptosis of TRAIL fragment via the upregulated death receptors on TRAIL-resistant pancreatic cancer cells and intrinsic apoptosis of AVPI tetrapeptide via the efficient accumulation and uptake of R6ST on SSN. Hence, SSN exhibited synergistic antitumor effect and provided a new strategy for TRAIL-resistant pancreatic cancer therapy.
Collapse
Affiliation(s)
- Xianzhou Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chunqing Ou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yaqian Shu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Songlin Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shouchun Chen
- Chengdu Huachuang Biotechnology Co. Ltd., Chengdu, 610041, China
| | - Qinjie Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Changyang Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
28
|
Mirzaei S, Mohammadi AT, Gholami MH, Hashemi F, Zarrabi A, Zabolian A, Hushmandi K, Makvandi P, Samec M, Liskova A, Kubatka P, Nabavi N, Aref AR, Ashrafizadeh M, Khan H, Najafi M. Nrf2 signaling pathway in cisplatin chemotherapy: Potential involvement in organ protection and chemoresistance. Pharmacol Res 2021; 167:105575. [PMID: 33771701 DOI: 10.1016/j.phrs.2021.105575] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/20/2021] [Accepted: 03/21/2021] [Indexed: 12/14/2022]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a vital transcription factor and its induction is of significant importance for protecting against oxidative damage. Increased levels of Reactive Oxygen Species (ROS) stimulate Nrf2 signaling, enhancing the activity of antioxidant enzymes such as catalase, superoxide dismutase and glutathione peroxidase. These enzymes are associated with retarding oxidative stress. On the other hand, Nrf2 activation in cancer cells is responsible for the development of chemoresistance due to disrupting oxidative mediated-cell death by reducing ROS levels. Cisplatin (CP), cis-diamminedichloroplatinum(II), is a potent anti-tumor agent extensively used in cancer therapy, but its frequent application leads to the development of chemoresistance as well. In the present study, association of Nrf2 signaling with chemoresistance to CP and protection against its deleterious effects is discussed. Anti-tumor compounds, mainly phytochemicals, retard chemoresistance by suppressing Nrf2 signaling. Upstream mediators such as microRNAs can regulate Nrf2 expression during CP chemotherapy regimens. Protection against side effects of CP is mediated via activating Nrf2 signaling and its downstream targets activating antioxidant defense system. Protective agents that activate Nrf2 signaling, can ameliorate CP-mediated ototoxicity, nephrotoxicity and neurotoxicity. Reducing ROS levels and preventing cell death are the most important factors involved in alleviating CP toxicity upon Nrf2 activation. As pre-clinical experiments advocate the role of Nrf2 in chemoprotection and CP resistance, translating these findings to the clinic can provide a significant progress in treatment of cancer patients.
Collapse
Affiliation(s)
- Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Aliasghar Tabatabaei Mohammadi
- Asu Vanda Gene Research Company, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Science Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Pooyan Makvandi
- Centre for Materials Interface, Istituto Italiano di Tecnologia, viale Rinaldo Piaggio 34, 56025 Pisa, Pontedera, Italy
| | - Marek Samec
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Alena Liskova
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Noushin Nabavi
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6 Canada
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Translational Sciences, Xsphera Biosciences Inc., Boston, MA, USA
| | - Milad Ashrafizadeh
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey; Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan.
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanashah University of Medical Sciences, Kermanshah 6715847141, Iran; Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
29
|
Li Y, Zhou X, Liu J, Yuan X, He Q. Therapeutic Potentials and Mechanisms of Artemisinin and its Derivatives for Tumorigenesis and Metastasis. Anticancer Agents Med Chem 2021; 20:520-535. [PMID: 31958040 DOI: 10.2174/1871520620666200120100252] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/10/2019] [Accepted: 10/24/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Tumor recurrence and metastasis are still leading causes of cancer mortality worldwide. The influence of traditional treatment strategies against metastatic tumors may still be limited. To search for novel and powerful agents against tumors has become a major research focus. In this study, Artemisinin (ARM), a natural compound isolated from herbs, Artemisia annua L., proceeding from drug repurposing methods, attracts more attention due to its good efficacy and tolerance in antimalarial practices, as well as newly confirmed anticancer activity. METHODS We have searched and reviewed the literatures about ARM and its derivatives (ARMs) for cancer using keywords "artemisinin" until May 2019. RESULTS In preclinical studies, ARMs can induce cell cycle arrest and cell death by apoptosis etc., to inhibit the progression of tumors, and suppress EMT and angiogenesis to inhibit the metastasis of tumors. Notably, the complex relationships of ARMs and autophagy are worth exploring. Inspired by the limitations of its antimalarial applications and the mechanical studies of artemisinin and cancer, people are also committed to develop safer and more potent ARM-based modified compounds (ARMs) or combination therapy, such as artemisinin dimers/ trimers, artemisinin-derived hybrids. Some clinical trials support artemisinins as promising candidates for cancer therapy. CONCLUSION ARMs show potent therapeutic potentials against carcinoma including metastatic tumors. Novel compounds derived from artemisinin and relevant combination therapies are supposed to be promising treatment strategies for tumors, as the important future research directions.
Collapse
Affiliation(s)
- Yue Li
- Department of Clinical Laboratories, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Xiaoyan Zhou
- Department of Clinical Laboratories, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Jiali Liu
- Department of Clinical Laboratories, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Xiaohong Yuan
- Department of Clinical Laboratories, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Qian He
- Department of Clinical Laboratories, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| |
Collapse
|
30
|
Artesunate synergizes with sorafenib to induce ferroptosis in hepatocellular carcinoma. Acta Pharmacol Sin 2021; 42:301-310. [PMID: 32699265 DOI: 10.1038/s41401-020-0478-3] [Citation(s) in RCA: 192] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023]
Abstract
Sorafenib is the first-line medication for advanced hepatocellular carcinoma (HCC), but it can only extend limited survival. It is imperative to find a combination strategy to increase sorafenib efficacy. Artesunate is such a preferred candidate, because artesunate is clinically well-tolerated and more importantly both drugs can induce ferroptosis through different mechanisms. In this study we investigated the combined effect of sorafenib and artesunate in inducing ferroptosis of HCC and elucidated the involved molecular mechanisms. We showed that artesunate greatly enhanced the anticancer effects of low dose of sorafenib against Huh7, SNU-449, and SNU-182 HCC cell lines in vitro and against Huh7 cell xenograft model in Balb/c nude mice. The combination index method confirmed that the combined effect of sorafenib and artesunate was synergistic. Compared with the treatment with artesunate or sorafenib alone, combined treatment induced significantly exacerbated lipid peroxidation and ferroptosis, which was blocked by N-acetyl cysteine and ferroptosis inhibitors liproxstatin-1 and deferoxamine mesylate, but not by inhibitors of other types of cell death (z-VAD, necrostatin-1 and belnacasan). In Huh7 cells, we demonstrated that the combined treatment induced oxidative stress and lysosome-mediated ferritinophagy, two essential aspects of ferroptosis. Sorafenib at low dose mainly caused oxidative stress through mitochondrial impairments and SLC7A11-invovled glutathione depletion. Artesunate-induced lysosome activation synergized with sorafenib-mediated pro-oxidative effects by promoting sequential reactions including lysosomal cathepsin B/L activation, ferritin degradation, lipid peroxidation, and consequent ferroptosis. Taken together, artesunate could be repurposed to sensitize sorafenib in HCC treatment. The combined treatment can be easily translated into clinical applications.
Collapse
|
31
|
Zhao QF, Ji J, Cai Q, Wang C, Shi M, Zhou CF, Zhu ZG, Zhang J. Low expression of transferrin receptor 2 predict poor prognosis in gastric cancer patients. Kaohsiung J Med Sci 2020; 36:1014-1020. [PMID: 32705773 DOI: 10.1002/kjm2.12278] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/18/2020] [Accepted: 06/29/2020] [Indexed: 11/06/2022] Open
Abstract
This study was to detect the expression level of transferrin receptor 2 (TfR2) in gastric cancer and to analyze its value in predicting the prognosis of gastric cancer patients. Real-time PCR (RT-qPCR) was applied to detect the mRNA expression of TfR2 in gastric cancer tissues and paired adjacent nontumorous tissues. Immunohistochemistry (IHC) and western blotting were used to determine the protein expression of TfR2 in gastric cancer, and the relationship with the prognosis of gastric cancer patients was analyzed. The results were: (1) The mRNA and protein levels of TfR2 in gastric cancer tissues were remarkably lower than those in adjacent nontumorous gastric tissue (P < .05). (2) Clinicopathological analysis showed that the expression level of TfR2 was significantly correlated with TNM stage of patients (P = .047). (3) The results of univariate survival analysis showed that histological grade, T stage, N stage, M stage, Lauren's classification, and TfR2 expression level influenced the overall survival of gastric cancer patients. Multivariate survival analysis showed that low TfR2 expression (HR = 1.671, 95%CI: 1.006-2.774, P = .047), poor differentiation (HR = 2.123, 95%CI: 1.188-3.795, P = .011), and M1 stage (HR = 8.541, 95%CI: 3.416-21.353, P < .001) were independent risk factors affecting the overall survival of gastric cancer patients. In conclusion, TfR2 exhibited low expression in gastric cancer tissue, and is an independent prognostic factor of gastric cancer patients.
Collapse
Affiliation(s)
- Qian-Fu Zhao
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Ji
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qu Cai
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Wang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Shi
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen-Fei Zhou
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng-Gang Zhu
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
32
|
Markowitsch SD, Schupp P, Lauckner J, Vakhrusheva O, Slade KS, Mager R, Efferth T, Haferkamp A, Juengel E. Artesunate Inhibits Growth of Sunitinib-Resistant Renal Cell Carcinoma Cells through Cell Cycle Arrest and Induction of Ferroptosis. Cancers (Basel) 2020; 12:cancers12113150. [PMID: 33121039 PMCID: PMC7692972 DOI: 10.3390/cancers12113150] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/22/2020] [Accepted: 10/24/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Renal cell carcinoma (RCC) is the most common kidney malignancy. Due to development of therapy resistance, efficacy of conventional drugs such as sunitinib is limited. Artesunate (ART), a drug originating from Traditional Chinese Medicine, has exhibited anti-tumor effects in several non-urologic tumors. ART inhibited growth, reduced metastatic properties, and curtailed metabolism in sunitinib-sensitive and sunitinib–resistant RCC cells. In three of four tested cell lines, ART’s growth inhibitory effects were accompanied by cell cycle arrest and modulation of cell cycle regulating proteins. In a fourth cell line, KTCTL-26, ART evoked ferroptosis, an iron-dependent cell death, and exhibited stronger anti-tumor effects than in the other cell lines. The regulatory protein, p53, was only detectable in the KTCTL-26 cells, possibly making p53 a predictive marker of cancer that may respond better to ART. ART, therefore, may hold promise as an additive therapy option for selected patients with advanced or therapy-resistant RCC. Abstract Although innovative therapeutic concepts have led to better treatment of advanced renal cell carcinoma (RCC), efficacy is still limited due to the tumor developing resistance to applied drugs. Artesunate (ART) has demonstrated anti-tumor effects in different tumor entities. This study was designed to investigate the impact of ART (1–100 µM) on the sunitinib-resistant RCC cell lines, Caki-1, 786-O, KTCTL26, and A-498. Therapy-sensitive (parental) and untreated cells served as controls. ART’s impact on tumor cell growth, proliferation, clonogenic growth, apoptosis, necrosis, ferroptosis, and metabolic activity was evaluated. Cell cycle distribution, the expression of cell cycle regulating proteins, p53, and the occurrence of reactive oxygen species (ROS) were investigated. ART significantly increased cytotoxicity and inhibited proliferation and clonogenic growth in both parental and sunitinib-resistant RCC cells. In Caki-1, 786-O, and A-498 cell lines growth inhibition was associated with G0/G1 phase arrest and distinct modulation of cell cycle regulating proteins. KTCTL-26 cells were mainly affected by ART through ROS generation, ferroptosis, and decreased metabolism. p53 exclusively appeared in the KTCTL-26 cells, indicating that p53 might be predictive for ART-dependent ferroptosis. Thus, ART may hold promise for treating selected patients with advanced and even therapy-resistant RCC.
Collapse
Affiliation(s)
- Sascha D. Markowitsch
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (S.D.M.); (P.S.); (J.L.); (O.V.); (K.S.S.); (R.M.); (A.H.)
| | - Patricia Schupp
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (S.D.M.); (P.S.); (J.L.); (O.V.); (K.S.S.); (R.M.); (A.H.)
| | - Julia Lauckner
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (S.D.M.); (P.S.); (J.L.); (O.V.); (K.S.S.); (R.M.); (A.H.)
| | - Olesya Vakhrusheva
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (S.D.M.); (P.S.); (J.L.); (O.V.); (K.S.S.); (R.M.); (A.H.)
| | - Kimberly S. Slade
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (S.D.M.); (P.S.); (J.L.); (O.V.); (K.S.S.); (R.M.); (A.H.)
| | - René Mager
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (S.D.M.); (P.S.); (J.L.); (O.V.); (K.S.S.); (R.M.); (A.H.)
| | - Thomas Efferth
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Staudingerweg 5, 55128 Mainz, Germany;
| | - Axel Haferkamp
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (S.D.M.); (P.S.); (J.L.); (O.V.); (K.S.S.); (R.M.); (A.H.)
| | - Eva Juengel
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (S.D.M.); (P.S.); (J.L.); (O.V.); (K.S.S.); (R.M.); (A.H.)
- Correspondence: ; Tel.: +49-631-175-433; Fax: +49-6131-174-410
| |
Collapse
|
33
|
Augustin Y, Staines HM, Krishna S. Artemisinins as a novel anti-cancer therapy: Targeting a global cancer pandemic through drug repurposing. Pharmacol Ther 2020; 216:107706. [PMID: 33075360 PMCID: PMC7564301 DOI: 10.1016/j.pharmthera.2020.107706] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/09/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023]
Abstract
Artemisinins are a unique class of antimalarial drugs with significant potential for drug repurposing for a wide range of diseases including cancer. Cancer is a leading cause of death globally and the majority of cancer related deaths occur in Low and Middle Income Countries (LMICs) where conventional treatment options are often limited by financial cost. Drug repurposing can significantly shorten new therapeutic discovery pathways, ensuring greater accessibility and affordability globally. Artemisinins have an excellent safety and tolerability profile as well as being affordable for deployment in Low and Middle Class Income Countries at around USD1 per daily dose. Robust, well designed clinical trials of artemisinin drug repurposing are indicated for a variety of different cancers and treatment settings.
Collapse
Affiliation(s)
- Yolanda Augustin
- Institute of Infection & Immunity, St George's University of London, United Kingdom
| | - Henry M Staines
- Institute of Infection & Immunity, St George's University of London, United Kingdom
| | - Sanjeev Krishna
- Institute of Infection & Immunity, St George's University of London, United Kingdom.
| |
Collapse
|
34
|
Xu C, Zhang H, Mu L, Yang X. Artemisinins as Anticancer Drugs: Novel Therapeutic Approaches, Molecular Mechanisms, and Clinical Trials. Front Pharmacol 2020; 11:529881. [PMID: 33117153 PMCID: PMC7573816 DOI: 10.3389/fphar.2020.529881] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 09/16/2020] [Indexed: 12/26/2022] Open
Abstract
Artemisinin and its derivatives have shown broad-spectrum antitumor activities in vitro and in vivo. Furthermore, outcomes from a limited number of clinical trials provide encouraging evidence for their excellent antitumor activities. However, some problems such as poor solubility, toxicity and controversial mechanisms of action hamper their use as effective antitumor agents in the clinic. In order to accelerate the use of ARTs in the clinic, researchers have recently developed novel therapeutic approaches including developing novel derivatives, manufacturing novel nano-formulations, and combining ARTs with other drugs for cancer therapy. The related mechanisms of action were explored. This review describes ARTs used to induce non-apoptotic cell death containing oncosis, autophagy, and ferroptosis. Moreover, it highlights the ARTs-caused effects on cancer metabolism, immunosuppression and cancer stem cells and discusses clinical trials of ARTs used to treat cancer. The review provides additional insight into the molecular mechanism of action of ARTs and their considerable clinical potential.
Collapse
Affiliation(s)
- Cangcang Xu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, China
| | - Huihui Zhang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, China
| | - Lingli Mu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, China
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, China
| |
Collapse
|
35
|
Armando RG, Gómez DLM, Gomez DE. New drugs are not enough‑drug repositioning in oncology: An update. Int J Oncol 2020; 56:651-684. [PMID: 32124955 PMCID: PMC7010222 DOI: 10.3892/ijo.2020.4966] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 12/16/2019] [Indexed: 11/24/2022] Open
Abstract
Drug repositioning refers to the concept of discovering novel clinical benefits of drugs that are already known for use treating other diseases. The advantages of this are that several important drug characteristics are already established (including efficacy, pharmacokinetics, pharmacodynamics and toxicity), making the process of research for a putative drug quicker and less costly. Drug repositioning in oncology has received extensive focus. The present review summarizes the most prominent examples of drug repositioning for the treatment of cancer, taking into consideration their primary use, proposed anticancer mechanisms and current development status.
Collapse
Affiliation(s)
- Romina Gabriela Armando
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| | - Diego Luis Mengual Gómez
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| | - Daniel Eduardo Gomez
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| |
Collapse
|
36
|
Yang Y, Wu N, Wu Y, Chen H, Qiu J, Qian X, Zeng J, Chiu K, Gao Q, Zhuang J. Artesunate induces mitochondria-mediated apoptosis of human retinoblastoma cells by upregulating Kruppel-like factor 6. Cell Death Dis 2019; 10:862. [PMID: 31723124 PMCID: PMC6853908 DOI: 10.1038/s41419-019-2084-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 09/29/2019] [Accepted: 10/17/2019] [Indexed: 01/28/2023]
Abstract
Retinoblastoma (RB) is the most common primary intraocular malignancy in children. Intravitreal chemotherapy achieves favorable clinical outcomes in controlling RB vitreous seeds, which are a common reason for treatment failure. Thus, a novel, effective and safe intravitreal chemotherapeutic drug is urgently required. The malaria drug artesunate (ART) recently demonstrated remarkable anticancer effects with mild side effects. The purpose of this study is to investigate the anti-RB efficacy, the underlying mechanism and the intraocular safety of ART. Herein, we verified that ART inhibits RB cell viability and induces cell apoptosis in a dose- and time-dependent manner. Microarray analysis revealed that Kruppel-like factor 6 (KLF6) was upregulated after ART treatment, and this was further confirmed by real-time PCR and western blot assays. Silencing of KLF6 expression significantly reversed ART-induced RB cell growth inhibition and apoptosis. Furthermore, ART activated mitochondria-mediated apoptosis of RB cells, while silencing KLF6 expression significantly inhibited this effect. In murine xenotransplantation models of RB, we further confirmed that ART inhibits RB tumor growth, induces tumor cell apoptosis and upregulates KLF6 expression. In addition, KLF6 silencing attenuates ART-mediated inhibition of tumor growth in vivo. Furthermore, we proved that intravitreal injection of ART in Sprague-Dawley (SD) rats is safe, with no obvious retinal function damage or structural disorders observed by electrophysiology (ERG), fundal photographs, fundus fluorescein angiography (FFA) or optical coherence tomography (OCT) examinations. Collectively, our study revealed that ART induces mitochondrial apoptosis of RB cells via upregulating KLF6, and our results may extend the application of ART to the clinic as an effective and safe intravitreal chemotherapeutic drug to treat RB, especially RB with vitreous seeds.
Collapse
Affiliation(s)
- Ying Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Nandan Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Yihui Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Haoting Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Jin Qiu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Xiaobing Qian
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Jieting Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Kin Chiu
- Department of Ophthalmology, The University of Hong Kong, Hong Kong SAR, P. R. China
| | - Qianying Gao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, P. R. China. .,Department of Ophthalmology, The 2nd Affiliate Hospital, Wenzhou Medical University, Wenzhou, 325000, P. R. China.
| | - Jing Zhuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, P. R. China.
| |
Collapse
|
37
|
Luo H, Vong CT, Chen H, Gao Y, Lyu P, Qiu L, Zhao M, Liu Q, Cheng Z, Zou J, Yao P, Gao C, Wei J, Ung COL, Wang S, Zhong Z, Wang Y. Naturally occurring anti-cancer compounds: shining from Chinese herbal medicine. Chin Med 2019; 14:48. [PMID: 31719837 PMCID: PMC6836491 DOI: 10.1186/s13020-019-0270-9] [Citation(s) in RCA: 318] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/23/2019] [Indexed: 12/24/2022] Open
Abstract
Numerous natural products originated from Chinese herbal medicine exhibit anti-cancer activities, including anti-proliferative, pro-apoptotic, anti-metastatic, anti-angiogenic effects, as well as regulate autophagy, reverse multidrug resistance, balance immunity, and enhance chemotherapy in vitro and in vivo. To provide new insights into the critical path ahead, we systemically reviewed the most recent advances (reported since 2011) on the key compounds with anti-cancer effects derived from Chinese herbal medicine (curcumin, epigallocatechin gallate, berberine, artemisinin, ginsenoside Rg3, ursolic acid, silibinin, emodin, triptolide, cucurbitacin B, tanshinone I, oridonin, shikonin, gambogic acid, artesunate, wogonin, β-elemene, and cepharanthine) in scientific databases (PubMed, Web of Science, Medline, Scopus, and Clinical Trials). With a broader perspective, we focused on their recently discovered and/or investigated pharmacological effects, novel mechanism of action, relevant clinical studies, and their innovative applications in combined therapy and immunomodulation. In addition, the present review has extended to describe other promising compounds including dihydroartemisinin, ginsenoside Rh2, compound K, cucurbitacins D, E, I, tanshinone IIA and cryptotanshinone in view of their potentials in cancer therapy. Up to now, the evidence about the immunomodulatory effects and clinical trials of natural anti-cancer compounds from Chinese herbal medicine is very limited, and further research is needed to monitor their immunoregulatory effects and explore their mechanisms of action as modulators of immune checkpoints.
Collapse
Affiliation(s)
- Hua Luo
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Chi Teng Vong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Hanbin Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yan Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peng Lyu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Ling Qiu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Mingming Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Qiao Liu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zehua Cheng
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jian Zou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peifen Yao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Caifang Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jinchao Wei
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Carolina Oi Lam Ung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Shengpeng Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zhangfeng Zhong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yitao Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| |
Collapse
|
38
|
Ding X, Yue W, Chen H. Effect of artesunate on apoptosis and autophagy in tamoxifen resistant breast cancer cells (TAM-R). Transl Cancer Res 2019; 8:1863-1872. [PMID: 35116937 PMCID: PMC8797964 DOI: 10.21037/tcr.2019.08.41] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/12/2019] [Indexed: 11/24/2022]
Abstract
Background The antitumor effect of artesunate (ART) is well-recognized. To investigate the effect of ART on tamoxifen-resistant breast cancer cells (TAM-R) proliferation, apoptosis, and autophagy with TAM-R cells of breast cancer as objects of study, and to investigate whether ART could re-sensitize TAM-R cells to TAM therapy. Methods Experiments were performed using TAM-R cell lines. Cell Death Detection ELISA kit was used to detect the level of apoptosis. Western blot and immunofluorescent staining analysis were conducted to detect autophagy and apoptosis related proteins in TAM-R cells. Results After treated with ART, the proliferation activity of TAM-R cells was inhibited in a concentration-dependent manner. Increased apoptosis activity was detected in TAM-R cells when treated with ART. Compared with 10−6 M TAM monotherapy group, treatment group with ART and TAM in combination caused significant reduction in the levels of Bcl-2, XIAP, and Survivin proteins, and elevation of caspase-7. However, increased p53 proteins was not detected after ART treatment. No significant change was observed in autophagy proteins LC-3 and Beclin-1 among control, ART, TAM, and ART combined with TAM groups. Conclusions The intervention of ART could not inhibit protective autophagy in TAM-R cells, however, possess potential in inducing apoptosis. In addition, as ART inhibit TAM-R cells growth in a dose-dependent manner, co-administration of 1 or 3 µM of ART with 10−6 M TAM might not be enough to re-sensitize TAM-R cells to TAM therapy.
Collapse
Affiliation(s)
- Xiaoqing Ding
- Department of Hematology, Dongfang Hospital Affiliated to Beijing University of Traditional Chinese Medicine, Beijing 100078, China
| | - Wei Yue
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA 22908, USA
| | - Haiyan Chen
- Department of Hematology, Dongfang Hospital Affiliated to Beijing University of Traditional Chinese Medicine, Beijing 100078, China
| |
Collapse
|
39
|
Liang W, Liu J, Wu H, Qiao X, Lu X, Liu Y, Zhu H, Ma L. Artemisinin induced reversal of EMT affects the molecular biological activity of ovarian cancer SKOV3 cell lines. Oncol Lett 2019; 18:3407-3414. [PMID: 31452821 PMCID: PMC6676620 DOI: 10.3892/ol.2019.10608] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 06/13/2019] [Indexed: 12/24/2022] Open
Abstract
Accumulating evidence suggests that celecoxib and artemisinin could mediate ovarian cancer development and metastasis. The present study investigated the effects of celecoxib and artemisinin on the epithelial-mesenchymal transition (EMT) characteristics of the human ovarian epithelial adenocarcinoma cell line, SKOV3. SKOV3 cells were incubated with celecoxib (10 µM) for different periods of time to establish an EMT cell model. Subsequently, artemisinin (20, 40 and 80 µM) was used to establish a cell model of the reverse process, mesenchymal-epithelial transition (MET). Cell proliferation, metastasis, invasiveness and the expression of vimentin and E-cadherin were measured using Cell Counting Kit-8, wound healing assay, western blotting, flow cytometry and immunofluorescence. The EMT cell model exhibited enhanced proliferative capacity, increased migration, increased vimentin expression and decreased E-cadherin expression. By contrast, artemisinin decreased proliferative capacity, decreased migration, decreased vimentin expression and increased E-cadherin expression of EMT model cells, indicating that MET was induced. These results demonstrated that artemisinin may reverse celecoxib-induced epithelial-mesenchymal transition in SKOV3 cells.
Collapse
Affiliation(s)
- Weichen Liang
- Department of Gynecologic Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Jian Liu
- Department of Gynecologic Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Huazhang Wu
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Xuxu Qiao
- Graduate Department, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Xiang Lu
- Department of Gynecologic Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Yonghong Liu
- Graduate Department, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Hong Zhu
- Department of Gynecologic Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Ling Ma
- Department of Gynecologic Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| |
Collapse
|
40
|
Cytotoxic Effects of Artemisia annua L. and Pure Artemisinin on the D-17 Canine Osteosarcoma Cell Line. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1615758. [PMID: 31354901 PMCID: PMC6637696 DOI: 10.1155/2019/1615758] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/21/2019] [Accepted: 06/13/2019] [Indexed: 12/20/2022]
Abstract
Artemisia annua has been used for centuries in Traditional Chinese Medicine. Although used as an antimalarial drug, its active compound artemisinin and the semisynthetic derivatives have also been investigated for their anticancer properties, with interesting and promising results. The aims of this research were to evaluate (i) the cytotoxicity and the antiproliferative effect of pure artemisinin and a hydroalcoholic extract obtained from A. annua on the D-17 canine osteosarcoma cell line and (ii) the intracellular iron concentration and its correlation with the cytotoxic effects. Both artemisinin and hydroalcoholic extract induced a cytotoxic effect in a dose-dependent manner. Pure artemisinin caused an increase of cells in the S phase, whereas the hydroalcoholic extract induced an evident increase in the G2/M phase. A significant decrease of iron concentration was measured in D-17 cells treated with pure artemisinin and hydroalcoholic extract compared to untreated cells. In conclusion, although preliminary, the data obtained in this study are indicative of a more potent cytotoxic activity of the hydroalcoholic extract than pure artemisinin, indicating a possible synergistic effect of the phytocomplex and a mechanism of action involving iron and possibly ferroptosis. Considering the similarities between human and canine osteosarcomas, progress in deepening knowledge and improving therapeutic protocols will probably be relevant for both species, in a model of reciprocal translational medicine.
Collapse
|
41
|
Dong D, Zhang G, Yang J, Zhao B, Wang S, Wang L, Zhang G, Shang P. The role of iron metabolism in cancer therapy focusing on tumor-associated macrophages. J Cell Physiol 2018; 234:8028-8039. [PMID: 30362549 DOI: 10.1002/jcp.27569] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/17/2018] [Indexed: 12/11/2022]
Abstract
Iron is an essential micronutrient in mammalian cells for basic processes such as DNA synthesis, cell cycle progression, and mitochondrial activity. Macrophages play a vital role in iron metabolism, which is tightly linked to their phagocytosis of senescent and death erythrocytes. It is now recognized that the polarization process of macrophages determines the expression profile of genes associated with iron metabolism. Although iron metabolism is strictly controlled by physiology, cancer has recently been connected with disordered iron metabolism. Moreover, in the environment of cancer, tumor-associated macrophages (TAMs) exhibit an iron release phenotype, which stimulates tumor cell survival and growth. Usually, the abundance of TAMs in the tumor is implicated in poor disease prognosis. Therefore, important attention has been drawn toward the development of tumor immunotherapies targeting these TAMs focussing on iron metabolism and reprogramming polarized phenotypes. Although further systematic research is still required, these efforts are almost certainly valuable in the search for new and effective cancer treatments.
Collapse
Affiliation(s)
- Dandan Dong
- School of Life Sciences, Northwestern Polytechnical University, Xi'an Shanxi, China.,Key Laboratory for Space Biosciences and Biotechnology, Xi'an Shanxi, China
| | - Gejing Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an Shanxi, China.,Key Laboratory for Space Biosciences and Biotechnology, Xi'an Shanxi, China
| | - Jiancheng Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an Shanxi, China.,Key Laboratory for Space Biosciences and Biotechnology, Xi'an Shanxi, China
| | - Bin Zhao
- School of Life Sciences, Northwestern Polytechnical University, Xi'an Shanxi, China.,Key Laboratory for Space Biosciences and Biotechnology, Xi'an Shanxi, China
| | - Shenghang Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an Shanxi, China.,Key Laboratory for Space Biosciences and Biotechnology, Xi'an Shanxi, China
| | - Luyao Wang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong, China
| | - Ge Zhang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University (HKBU), Hong Kong, China
| | - Peng Shang
- Research & Development Institute in Shenzhen, Northwestern Polytechnical University, Shenzhen, China.,Key Laboratory for Space Biosciences and Biotechnology, Xi'an Shanxi, China
| |
Collapse
|
42
|
Son JY, Yoon S, Tae IH, Park YJ, De U, Jeon Y, Park YJ, Rhyu IJ, Lee BM, Chung K, Lim JE, Lee SJ, Lee HW, Kwak JH, Kim HS, Choi HY. Novel therapeutic roles of MC-4 in combination with everolimus against advanced renal cell carcinoma by dual targeting of Akt/pyruvate kinase muscle isozyme M2 and mechanistic target of rapamycin complex 1 pathways. Cancer Med 2018; 7:5083-5095. [PMID: 30160042 PMCID: PMC6198234 DOI: 10.1002/cam4.1748] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/08/2018] [Accepted: 08/05/2018] [Indexed: 12/11/2022] Open
Abstract
Current clinical trials of new anticancer therapies against metastatic renal cell carcinoma (RCC), including molecular-targeted therapies, have not shown promise. The purpose of this study was to preclinically assess the antitumor effects of MC-4, a partially purified material of Artemisia annua L., as a monotherapy or in combination with the known mechanistic target of rapamycin complex 1 (mTORC1) inhibitor, everolimus, against Caki-1 (Von Hippel-Lindau (VHL)+/+) and 786-O (VHL-/-) human RCC cells. MC-4 monotherapy significantly increased tumor growth inhibition and autophagic cell death in RCC cells in vitro and in vivo. Everolimus led to compensatory Akt activation by inhibiting only mTORC1 signaling pathway. In contrast to everolimus, MC-4 enhanced phosphatase and tensin homolog expression and reduced its downstream effector, Akt/pyruvate kinase muscle isozyme M2 (PKM2), leading to decreased expression of glucose transporter 1, which is associated with cancer cell metabolism. The synergistic antitumor and anti-metastatic effects induced by co-administration of MC-4 and everolimus involve cell growth inhibition and autophagic cell death via dual targeting of phosphatidylinositol 3-kinase (PI3K)/Akt/PKM2 and mTORC1. These findings suggest that MC-4 is a novel Akt/PKM2 inhibitor that can overcome the limitation of existing mTOR inhibitors and can be considered a novel strategy to treat patients with rapidly progressing advanced RCC.
Collapse
Affiliation(s)
- Ji Yeon Son
- School of PharmacySungkyunkwan UniversitySuwonKorea
| | - Sungpil Yoon
- School of PharmacySungkyunkwan UniversitySuwonKorea
| | - In Hwan Tae
- School of PharmacySungkyunkwan UniversitySuwonKorea
| | - Yu Jin Park
- School of PharmacySungkyunkwan UniversitySuwonKorea
| | - Umasankar De
- School of PharmacySungkyunkwan UniversitySuwonKorea
| | | | | | - Im Joo Rhyu
- Department of AnatomyKorea University College of MedicineSeoulKorea
| | - Byung Mu Lee
- School of PharmacySungkyunkwan UniversitySuwonKorea
| | | | - Joung Eun Lim
- Departments of UrologySamsung Medical CenterSungkyunkwan University School of MedicineSeoulKorea
| | - Se Jeong Lee
- Department of AnatomyKorea University College of MedicineSeoulKorea
| | - Hye Won Lee
- Departments of UrologySamsung Medical CenterSungkyunkwan University School of MedicineSeoulKorea
- Department of Anatomy and Cell BiologySungkyunkwan University School of MedicineSuwonKorea
- Single Cell Network Research CenterSungkyunkwan University School of MedicineSuwonKorea
| | | | | | - Han Yong Choi
- Department of UrologyKangbuk Samsung HospitalSeoulKorea
| |
Collapse
|
43
|
Hamaidi I, Coquard C, Danilin S, Dormoy V, Béraud C, Rothhut S, Barthelmebs M, Benkirane-Jessel N, Lindner V, Lang H, Massfelder T. The Lim1 oncogene as a new therapeutic target for metastatic human renal cell carcinoma. Oncogene 2018; 38:60-72. [DOI: 10.1038/s41388-018-0413-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 06/12/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022]
|
44
|
Prevention of carcinogenesis and metastasis by Artemisinin-type drugs. Cancer Lett 2018; 429:11-18. [DOI: 10.1016/j.canlet.2018.05.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 05/02/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022]
|
45
|
Conte M, Lupette J, Seddiki K, Meï C, Dolch LJ, Gros V, Barette C, Rébeillé F, Jouhet J, Maréchal E. Screening for Biologically Annotated Drugs That Trigger Triacylglycerol Accumulation in the Diatom Phaeodactylum. PLANT PHYSIOLOGY 2018; 177:532-552. [PMID: 29535162 PMCID: PMC6001342 DOI: 10.1104/pp.17.01804] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/06/2018] [Indexed: 05/03/2023]
Abstract
Microalgae are a promising feedstock for the production of triacylglycerol (TAG) for a variety of potential applications, ranging from food and human health to biofuels and green chemistry. However, obtaining high TAG yields is challenging. A phenotypic assay for the accumulation of oil droplets was developed to screen a library of 1,200 drugs, annotated with pharmacology information, to select compounds that trigger TAG accumulation in the diatom Phaeodactylum tricornutum Using this screen, we identified 34 molecules acting in a dose-dependent manner. Previously characterized targets of these compounds include cell division and cell signaling effectors, membrane receptors and transporters, and sterol metabolism. Among the five compounds possibly acting on sterol metabolism, we focused our study on ethynylestradiol, a synthetic form of estrogen that is used in contraceptive pills and known for its ecological impact as an endocrine disruptor. Ethynylestradiol impaired the production of very-long-chain polyunsaturated fatty acids, destabilized the galactolipid versus phospholipid balance, and triggered the recycling of fatty acids from membrane lipids to TAG. The P. tricornutum transcriptomic response to treatment with ethynylestradiol was consistent with the reallocation of carbon from sterols to acetyl-coenzyme A and TAG. The mode of action and catabolism of ethynylestradiol are unknown but might involve several up-regulated cytochrome P450 proteins. A fatty acid elongase, Δ6-ELO-B1, might be involved in the impairment of very-long-chain polyunsaturated fatty acids and fatty acid turnover. This phenotypic screen opens new perspectives for the exploration of novel bioactive molecules, potential target genes, and pathways controlling TAG biosynthesis. It also unraveled the sensitivity of diatoms to endocrine disruptors, highlighting an impact of anthropogenic pollution on phytoplankton.
Collapse
Affiliation(s)
- Melissa Conte
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Josselin Lupette
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Khawla Seddiki
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Coline Meï
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Lina-Juana Dolch
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Valérie Gros
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Caroline Barette
- Laboratoire de Biologie à Grande Echelle, Commissariat à l'Energie Atomique, INSERM, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Fabrice Rébeillé
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Juliette Jouhet
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Eric Maréchal
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| |
Collapse
|
46
|
Jiang F, Zhou JY, Zhang D, Liu MH, Chen YG. Artesunate induces apoptosis and autophagy in HCT116 colon cancer cells, and autophagy inhibition enhances the artesunate‑induced apoptosis. Int J Mol Med 2018; 42:1295-1304. [PMID: 29901098 PMCID: PMC6089754 DOI: 10.3892/ijmm.2018.3712] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/30/2018] [Indexed: 12/16/2022] Open
Abstract
The present study assessed the antitumor effect of artesunate (ART) in vitro and in vivo, as well as its underlying mechanism of action in HCT116 colon cancer cells. An MTT assay, DAPI staining, flow cytometry, western blotting, immunohistochemistry, transmission electron microscopy and TUNEL assay were performed to study the molecular mechanism underlying the antitumor effects of ART in HCT116 colon cancer cells. ART was observed to inhibit proliferation by inducing the apoptosis of HCT116 cells both in vitro and in vivo. Flow cytometry analysis demonstrated that treatment with 2 and 4 µg/ml ART for 48 h induced early apoptosis in 22.7 and 33.8% of cells, respectively. In the xenograft tumors of BALB/c nude mice, TUNEL-positive cells increased in the ART group compared with that in the normal saline group. Furthermore,the associated mitochondrial cleaved-caspase 3, poly-ADP ribose polymerase (PARP), caspase 9 and Bcl-2-associated X protein levels increased while B-cell lymphoma-2 (Bcl-2) decreased both in the cell and animal ART-treated group. ART-treated cells also exhibited autophagy induction, as evidenced by increased protein expression levels of light chain 3 (LC3) and beclin-1, and the presence of autophagosomes. Notably, pharmacological blockade of autophagy activation using hydroxychloroquine markedly enhanced ART-induced apoptosis and increased the protein levels of cleaved caspase 3 and PARP, while decreasing the levels of LC3 and beclin-1. These findings suggested that the ART-induced autophagy may have a cytoprotective effect by suppressing apoptosis. In conclusion, ART may be a potentially clinically useful anticancer drug for human colon cancer. In addition, co-treatment with ART and an autophagy inhibitor may be an effective anticancer therapy.
Collapse
Affiliation(s)
- Feng Jiang
- Department of Colorectal Surgery, The Affiliated Hospital of Nanjing University of TCM, Nanjing, Jiangsu 210029, P.R. China
| | - Jin-Yong Zhou
- Department of Central Laboratory, The Affiliated Hospital of Nanjing University of TCM, Nanjing, Jiangsu 210029, P.R. China
| | - Dan Zhang
- Department of Colorectal Surgery, The Affiliated Hospital of Nanjing University of TCM, Nanjing, Jiangsu 210029, P.R. China
| | - Ming-Hao Liu
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Yu-Gen Chen
- Department of Colorectal Surgery, The Affiliated Hospital of Nanjing University of TCM, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
47
|
Coleman DT, Gray AL, Stephens CA, Scott ML, Cardelli JA. Repurposed drug screen identifies cardiac glycosides as inhibitors of TGF-β-induced cancer-associated fibroblast differentiation. Oncotarget 2017; 7:32200-9. [PMID: 27058757 PMCID: PMC5078007 DOI: 10.18632/oncotarget.8609] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/14/2016] [Indexed: 12/22/2022] Open
Abstract
The tumor microenvironment, primarily composed of myofibroblasts, directly influences the progression of solid tumors. Through secretion of growth factors, extracellular matrix deposition, and contractile mechanotransduction, myofibroblasts, or cancer-associated fibroblasts (CAFs), support angiogenesis and cancer cell invasion and metastasis. The differentiation of fibroblasts to CAFs is primarily induced by TGF-β from cancer cells. To discover agents capable of blocking CAF differentiation, we developed a high content immunofluorescence-based assay to screen repurposed chemical libraries utilizing fibronectin expression as an initial CAF marker. Screening of the Prestwick chemical library and NIH Clinical Collection repurposed drug library, totaling over 1700 compounds, identified cardiac glycosides as particularly potent CAF blocking agents. Cardiac glycosides are traditionally used to regulate intracellular calcium by inhibiting the Na+/K+ ATPase to control cardiac contractility. Herein, we report that multiple cardiac glycoside compounds, including digoxin, are able to inhibit TGF-β-induced fibronectin expression at low nanomolar concentrations without undesirable cell toxicity. We found this inhibition to hold true for multiple fibroblast cell lines. Using real-time qPCR, we determined that digoxin prevented induction of multiple CAF markers. Furthermore, we report that digoxin is able to prevent TGF-β-induced fibroblast contraction of extracellular matrix, a major phenotypic consequence of CAF differentiation. Assessing the mechanism of inhibition, we found digoxin reduced SMAD promoter activity downstream of TGF-β, and we provide data that the effect is through inhibition of its known target, the Na+/K+ ATPase. These findings support a critical role for calcium signaling during CAF differentiation and highlight a novel, repurposable modality for cancer therapy.
Collapse
Affiliation(s)
- David T Coleman
- Louisiana State University Health Sciences Center, Feist-Weiller Cancer Center, Shreveport, LA, USA
| | - Alana L Gray
- Louisiana State University Health Sciences Center, Feist-Weiller Cancer Center, Shreveport, LA, USA
| | - Charles A Stephens
- Louisiana State University Health Sciences Center, Feist-Weiller Cancer Center, Shreveport, LA, USA
| | - Matthew L Scott
- Louisiana State University Health Sciences Center, Feist-Weiller Cancer Center, Shreveport, LA, USA
| | - James A Cardelli
- Louisiana State University Health Sciences Center, Feist-Weiller Cancer Center, Shreveport, LA, USA
| |
Collapse
|
48
|
Liu R, Yu X, Su C, Shi Y, Zhao L. Nanoparticle Delivery of Artesunate Enhances the Anti-tumor Efficiency by Activating Mitochondria-Mediated Cell Apoptosis. NANOSCALE RESEARCH LETTERS 2017; 12:403. [PMID: 28610396 PMCID: PMC5468175 DOI: 10.1186/s11671-017-2169-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 05/29/2017] [Indexed: 05/18/2023]
Abstract
Artemisinin and its derivatives were considered to exert a broad spectrum of anti-cancer activities, and they induced significant anti-cancer effects in tumor cells. Artemisinin and its derivatives could be absorbed quickly, and they were widely distributed, selectively killing tumor cells. Since low concentrations of artesunate primarily depended on oncosis to induce cell death in tumor cells, its anti-tumor effects were undesirable and limited. To obtain better anti-tumor effects, in this study, we took advantage of a new nanotechnology to design novel artesunate-loaded bovine serum albumin nanoparticles to achieve the mitochondrial accumulation of artesunate and induce mitochondrial-mediated apoptosis. The results showed that when compared with free artesunate's reliance on oncotic death, artesunate-loaded bovine serum albumin nanoparticles showed higher cytotoxicity and their significant apoptotic effects were induced through the distribution of artesunate in the mitochondria. This finding indicated that artesunate-loaded bovine serum albumin nanoparticles damaged the mitochondrial integrity and activated mitochondrial-mediated cell apoptosis by upregulating apoptosis-related proteins and facilitating the rapid release of cytochrome C.
Collapse
Affiliation(s)
- Rui Liu
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000 Liaoning People’s Republic of China
| | - Xiwei Yu
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000 Liaoning People’s Republic of China
| | - Chang Su
- School of Veterinary Medicine, Jinzhou Medical University, Jinzhou, 121000 Liaoning People’s Republic of China
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000 Liaoning People’s Republic of China
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000 Liaoning People’s Republic of China
| |
Collapse
|
49
|
Zhao C, Liu Q, Wang K. Artesunate attenuates ACLT-induced osteoarthritis by suppressing osteoclastogenesis and aberrant angiogenesis. Biomed Pharmacother 2017; 96:410-416. [DOI: 10.1016/j.biopha.2017.10.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 09/18/2017] [Accepted: 10/02/2017] [Indexed: 01/22/2023] Open
|
50
|
Zyad A, Tilaoui M, Jaafari A, Oukerrou MA, Mouse HA. More insights into the pharmacological effects of artemisinin. Phytother Res 2017; 32:216-229. [PMID: 29193409 DOI: 10.1002/ptr.5958] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/26/2017] [Accepted: 09/28/2017] [Indexed: 12/23/2022]
Abstract
Artemisinin is one of the most widely prescribed drugs against malaria and has recently received increased attention because of its other potential biological effects. The aim of this review is to summarize recent discoveries of the pharmaceutical effects of artemisinin in basic science along with its mechanistic action, as well as the intriguing results of recent clinical studies, with a focus on its antitumor activity. Scientific evidence indicates that artemisinin exerts its biological activity by generating reactive oxygen species that damage the DNA, mitochondrial depolarization, and cell death. In the present article review, scientific evidence suggests that artemisinin is a potential therapeutic agent for various diseases. Thus, this review is expected to encourage interested scientists to conduct further preclinical and clinical studies to evaluate these biological activities.
Collapse
Affiliation(s)
- Abdelmajid Zyad
- Laboratory of Biological Engineering, Team of Natural Substances and Cellular and Molecular Immuno-pharmacology, Immuno-biology of Cancer Cells, Sultan Moulay Slimane University, Faculty of Science and Technology, Beni-Mellal, Morocco
| | - Mounir Tilaoui
- Laboratory of Biological Engineering, Team of Natural Substances and Cellular and Molecular Immuno-pharmacology, Immuno-biology of Cancer Cells, Sultan Moulay Slimane University, Faculty of Science and Technology, Beni-Mellal, Morocco
| | - Abdeslam Jaafari
- Laboratory of Biological Engineering, Team of Natural Substances and Cellular and Molecular Immuno-pharmacology, Immuno-biology of Cancer Cells, Sultan Moulay Slimane University, Faculty of Science and Technology, Beni-Mellal, Morocco
| | - Moulay Ali Oukerrou
- Laboratory of Biological Engineering, Team of Natural Substances and Cellular and Molecular Immuno-pharmacology, Immuno-biology of Cancer Cells, Sultan Moulay Slimane University, Faculty of Science and Technology, Beni-Mellal, Morocco
| | - Hassan Ait Mouse
- Laboratory of Biological Engineering, Team of Natural Substances and Cellular and Molecular Immuno-pharmacology, Immuno-biology of Cancer Cells, Sultan Moulay Slimane University, Faculty of Science and Technology, Beni-Mellal, Morocco
| |
Collapse
|