1
|
Power K, Leandri R, Federico G, De Vico G, Leonardi L. Ferritinophagy: a possible new iron-related metabolic target in canine osteoblastic osteosarcoma. Front Vet Sci 2025; 12:1546872. [PMID: 40196812 PMCID: PMC11973301 DOI: 10.3389/fvets.2025.1546872] [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: 12/17/2024] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Canine osteosarcomas (COS) are the most common bone tumors in dogs, characterized by high metastatic rates, poor prognosis, and poor responsiveness to routine therapies, which highlights the need for new treatment targets. In this context, the metabolism of neoplastic cells represents an increasingly studied element, as cancer cells depend on particular metabolic pathways that are also elements of vulnerability. Among these, tumor cells (TCs) show higher iron requirements to sustain proliferation (so-called iron addiction), which are achieved by increasing iron uptake and/or by activating ferritinophagy, a process mediated by the Nuclear receptor Co-Activator 4 (NCOA4) leading to iron mobilization from ferritin (Ft) deposits. Previous studies have shown that COS cells overexpress Transferrin Receptor 1 (TfR1) to increase iron uptake. In this study we evaluated the immunohistochemical expression of ferritinophagy-related proteins, namely Ferritin Heavy chain (FTH1) and NCOA4, and proliferating cell nuclear antigen (PCNA) in canine normal bone and canine osteoblastic osteosarcoma (COOS) samples. Normal samples revealed negative/weak immunoreactivity for FTH1, NCOA4 and PCNA in <10% of osteocytes. In COOS samples the majority of neoplastic cells showed immunoreactivity to FTH1, NCOA4 and PCNA. Our data suggest that the activation of ferritinophagy by COOS cells responds to the need for feed their "iron addiction." These data, though preliminary, further suggest that targeting iron metabolism represents a new potential strategy worthy of further study to be transferred into clinical practice.
Collapse
Affiliation(s)
- Karen Power
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Rebecca Leandri
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Giorgia Federico
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Gionata De Vico
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Leonardo Leonardi
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
2
|
Altuwaijri N, Atef E. Transferrin-Conjugated Nanostructured Lipid Carriers for Targeting Artemisone to Melanoma Cells. Int J Mol Sci 2024; 25:9119. [PMID: 39201805 PMCID: PMC11354828 DOI: 10.3390/ijms25169119] [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: 07/07/2024] [Revised: 07/27/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
We report a successful formulation of Artemisone (ATM) in transferrin (Tf)-conjugated nanostructured lipid carriers (NLCs), achieving nearly a five-times increase in cell toxicity. The escalating cost of new drug discoveries led to the repurposing of approved drugs for new indications. This study incorporated Artemisone, an antimalarial drug, into a nanostructured lipid carrier (NLC) and tested for possible anticancer effects. The aim was to develop NLCs, and transferrin-conjugated NLCs (NLC-Tf) encapsulating Artemisone to enhance its delivery and anticancer activity. NLC formulations were prepared using high-pressure homogenization followed by ultrasonication and were characterized by particle size, zeta potential, and PDI. The conjugation of (Tf) to (NLC) was confirmed using IR, and the anticancer activity was tested using MTS assay. All formulations were in the nanometer size range (140-167 nm) with different zeta potential values. IR spectroscopy confirmed the successful conjugation of transferrin to NLC. Upon testing the formulations on melanoma cell lines using MTS assay, there was a significant decrease in viability and an increase in the encapsulated ATM-Tf toxicity compared to positive control ATM. The NLCs presented a promising potential carrier for delivering ATM to melanoma cells, and further conjugation with Tf significantly improved the ATM cytotoxicity.
Collapse
Affiliation(s)
- Njoud Altuwaijri
- Pharmaceutical Sciences Department, MCPHS University, 179 Longwood Ave, Boston, MA 02115, USA
| | - Eman Atef
- Pharmacy College, West Coast University, 590 N Vermont Ave, Los Angeles, CA 90005, USA
| |
Collapse
|
3
|
Chen C, Chen YX, Zhang CJ. A Radical-Generating Probe to Release Free Fluorophores and Identify Artemisinin-Sensitive Cancer Cells. ACS Sens 2024; 9:2310-2316. [PMID: 38651676 DOI: 10.1021/acssensors.4c00010] [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] [Indexed: 04/25/2024]
Abstract
The smart light-up probes have been extensively developed to image various enzymes and other bioactive molecules. Upon activation, these probes result in light-up fluorophores that exist in a protein-bound or a free form. The difference between these two forms has not yet been reported. Here, we present a pair of smart light-up probes that generate a protein-bound fluorophore and a free fluorophore upon activation by heme. Probe 8 generated a radical-attached fluorophore that predominantly existed in the free form, while probe 10 generated an α,β-unsaturated ketone-attached fluorophore that showed extensive labeling of proteins. In live-cell imaging, probe 8 showed greater fluorescence intensity than probe 10 when low concentrations (0.1-5 μM) of the probes were used, but probe 8 was less fluorescent than probe 10 when the concentrations of the probes were high (10 μM). Finally, probe 8 was used to reflect the activation level of the endoperoxide bond in cancer cells and to effectively distinguish ART-sensitive cancer cells from ART-insensitive ones.
Collapse
Affiliation(s)
- Chen Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yi-Xin Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Chong-Jing Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
4
|
Yang J, Xia T, Zhou S, Liu S, Pan T, Li Y, Luo Z. Anticancer Effect of Dihydroartemisinin via Dual Control of ROS-induced Apoptosis and Protective Autophagy in Prostate Cancer 22Rv1 Cells. Curr Pharm Biotechnol 2024; 25:1321-1332. [PMID: 37605406 DOI: 10.2174/1389201024666230821155243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/24/2023] [Accepted: 07/07/2023] [Indexed: 08/23/2023]
Abstract
BACKGROUND Dihydroartemisinin (DHA), a natural agent, exhibits potent anticancer activity. However, its biological activity on prostate cancer (PCa) 22Rv1 cells has not been previously investigated. OBJECTIVES In this study, we demonstrate that DHA induces anticancer effects through the induction of apoptosis and autophagy. METHODS Cell viability and proliferation rate were assessed using the CCK-8 assay and cell clone formation assay. The generation of reactive oxygen species (ROS) was detected by flow cytometry. The molecular mechanism of DHA-induced apoptosis and autophagy was examined using Western blot and RT-qPCR. The formation of autophagosomes and the changes in autophagy flux were observed using transmission electron microscopy (TEM) and confocal microscopy. The effect of DHA combined with Chloroquine (CQ) was assessed using the EdU assay and flow cytometry. The expressions of ROS/AMPK/mTOR-related proteins were detected using Western blot. The interaction between Beclin-1 and Bcl-2 was examined using Co-IP. RESULTS DHA inhibited 22Rv1 cell proliferation and induced apoptosis. DHA exerted its antiprostate cancer effects by increasing ROS levels. DHA promoted autophagy progression in 22Rv1 cells. Inhibition of autophagy enhanced the pro-apoptotic effect of DHA. DHA-induced autophagy initiation depended on the ROS/AMPK/mTOR pathway. After DHA treatment, the impact of Beclin- 1 on Bcl-2 was weakened, and its binding with Vps34 was enhanced. CONCLUSION DHA induces apoptosis and autophagy in 22Rv1 cells. The underlying mechanism may involve the regulation of ROS/AMPK/mTOR signaling pathways and the interaction between Beclin-1 and Bcl-2 proteins. Additionally, the combination of DHA and CQ may enhance the efficacy of DHA in inhibiting tumor cell activity.
Collapse
Affiliation(s)
- Jiaxin Yang
- Laboratory of Medical Experiment Technology, Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Tong Xia
- Laboratory of Medical Experiment Technology, Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Sijie Zhou
- Laboratory of Medical Experiment Technology, Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Sihao Liu
- Laboratory of Medical Experiment Technology, Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Tingyu Pan
- Laboratory of Medical Experiment Technology, Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Ying Li
- Laboratory of Medical Experiment Technology, Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Ziguo Luo
- Laboratory of Medical Experiment Technology, Institute of Life Science, Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Yan JX, Li QH, Li TZ, Huang ZY, Ma YB, Chen JJ. Design and synthesis of guaianolide-germacranolide heterodimers as novel anticancer agents against hepatocellular carcinoma. Drug Dev Res 2023; 84:1285-1298. [PMID: 37345274 DOI: 10.1002/ddr.22087] [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: 02/21/2023] [Revised: 04/10/2023] [Accepted: 05/19/2023] [Indexed: 06/23/2023]
Abstract
Inspired by our previous finding that disesquiterpenoids showed more potent antihepatoma cytotoxicity than their corresponding parent monomers, natural product-like guaianolide-germacranolide heterodimers were designed and synthesized from guaianolide diene and germacranolides via a biomimetic Diels-Alder reaction to provide three antihepatoma active dimers with novel scaffolds. To explore the structure-activity relationship, 31 derivatives containing ester, carbamate, ether, urea, amide, and triazole functional groups at C-14' were synthesized and evaluated for their cytotoxic activities against HepG2, Huh7, and SK-Hep-1 cell lines. Among them, 25 compounds were more potent than sorafenib against HepG2 cells, 15 compounds were stronger than sorafenib against Huh7 cells, and 17 compounds were stronger than sorafenib against SK-Hep-1 cells. Compound 23 showed the most potent cytotoxicity against three hepatoma cell lines with IC50 values of 4.4 µM (HepG2), 3.7 µM (Huh7), and 3.1 µM (SK-Hep-1), which were 2.7-, 2.2-, and 2.8-fold more potent than sorafenib, respectively. The underlying mechanism study demonstrated that compound 23 could induce cell apoptosis, prevent cell migration and invasion, cause G2/M phase arrest in SK-Hep-1 cells. Network pharmacology analyses predicted PDGFRA was one of the potential targets of compound 23, and surface plasmon resonance (SPR) assay verified that 23 had strong affinity with PDGFRA with a dissociatin constant (KD) value of 90.2 nM. These promising findings revealed that structurally novel guaianolide-germacranolide heterodimers might provide a new inspiration for the discovery of antihepatoma agents.
Collapse
Affiliation(s)
- Jia-Xin Yan
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education and School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, People's Republic of China
| | - Qi-Hao Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China
| | - Tian-Ze Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China
| | - Zhi-Yan Huang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education and School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an, People's Republic of China
| | - Yun-Bao Ma
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China
| | - Ji-Jun Chen
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, People's Republic of China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, People's Republic of China
| |
Collapse
|
6
|
Gong Y, Peng Q, Gao Y, Yang J, Lu J, Zhang Y, Yang Y, Liang H, Yue Y, Shi X. Dihydroartemisinin inhibited interleukin-18 expression by decreasing YAP1 in hepatocellular carcinoma cells. Acta Histochem 2023; 125:152040. [PMID: 37119608 DOI: 10.1016/j.acthis.2023.152040] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/22/2023] [Accepted: 04/22/2023] [Indexed: 05/01/2023]
Abstract
BACKGROUND Yes-associated protein 1 (YAP1) is highly expressed in liver cancer and has been used as an independent prognostic marker for hepatocellular carcinoma (HCC), while inhibition of YAP1 slows down the progression of HCC. Interleukin-18 (IL-18) also tends to be highly expressed in liver cancer. Previous research has proved that dihydroartemisinin (DHA) plays an important role in HCC treatment by reducing YAP1 expression. However, the relationship between YAP1 and IL-18 has not been reported in HCC, especially during DHA therapy. OBJECTIVE The purpose of this study was to clarify the relationship between YAP1 and IL-18 in HCC cells, and to explicit the role of IL-18 in the treatment of HCC by DHA. METHODS AND RESULTS We found that YAP1 and IL-18 were highly expressed in patients with hepatocellular carcinoma by bioinformatics analysis. Moreover, YAP1 was positively correlated with IL18 in liver cancer. YAP1 and IL18 correlated with immune cell infiltration, notably T cell exhaustion. YAP1 knockdown decreased IL-18 expression, while YAP1 overexpression increased the IL-18 expression in HCC cells. DHA reduced IL-18 expression through YAP1 in HCC cells. Further, DHA reduced the growth of Hepa1-6 cells subcutaneous xenograft tumors by inhibiting the expression of YAP1 and IL-18. However, DHA improved IL-18 in serum and adjacent tissues from DEN/TCPOBOP-induced liver tumor model in C57BL/6 mice. CONCLUSION YAP1 was positively correlated with IL-18 in HCC. DHA reduced the expression of IL-18 by inhibiting YAP1 and plays a role in the treatment of HCC. Our study suggested that IL-18 is a potential target for the treatment of HCC, and DHA is a promising drug for HCC therapy. DATA AVAILABILITY The dataset that supports the findings of this study is available from the corresponding author upon reasonable request.
Collapse
Affiliation(s)
- Yi Gong
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Qing Peng
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yuting Gao
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; School of Basic Medical Sciences, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Jiali Yang
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Junlan Lu
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yuman Zhang
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yanguang Yang
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Hua Liang
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yuan Yue
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Xinli Shi
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China.
| |
Collapse
|
7
|
Liu X, Du S, Wang S, Ye K. Ferroptosis in osteosarcoma: A promising future. Front Oncol 2022; 12:1031779. [PMID: 36457488 PMCID: PMC9705963 DOI: 10.3389/fonc.2022.1031779] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/01/2022] [Indexed: 10/25/2023] Open
Abstract
The incidence of osteosarcoma (OS) is increasing year by year, and the prognosis of patients with advanced OS is extremely poor due to the tendency of recurrence and chemotherapy resistance after surgery. Ferroptosis is a novel form of programmed cell death (PCD) that kills cells through iron-dependent lipid peroxidation. Current studies have shown that ferroptosis is closely related to OS and could reduce chemotherapy resistance to a certain extent, which has great therapeutic potential. In this paper, we review the regulatory mechanism of ferroptosis and its research progress in OS, hoping to provide new help for the clinical treatment of OS.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Shaowen Du
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Shengdong Wang
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Kaishan Ye
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
8
|
Xu DL, Fan K, Zhang H, Tang LX, Wang Y, Xiang Z, Shi AM, Qu YC, Su CJ, Pan J. Anti-proliferation and apoptosis-inducing effects of dihydroartemisinin on SH-SY5Y cells and metabolomic analysis. Transl Pediatr 2022; 11:1346-1361. [PMID: 36072536 PMCID: PMC9442203 DOI: 10.21037/tp-22-331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/11/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND In childhood, metastatic neuroblastoma (NB) is the most common extracranial solid tumor, but there are no appropriate drugs for its treatment. Dihydroartemisinin (DHA), a drug for malaria treatment, has therapeutic potential in several cancers; however, its mechanisms remain unclear. This study aimed to investigate the anti-proliferation effect of DHA on SH-SY5Y cells and to explore its mechanism in vitro. METHODS We used 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay to measure the half-maximal inhibitory concentration (IC50) of DHA; western blot was used to determine protein levels; propidium iodide (PI) staining was used to determine apoptotic cells; JC-1 staining to measure mitochondrial membrane potential; and dichloro-dihydro-fluorescein diacetate (DCFH-DA) staining was used to determine reactive oxygen species (ROS). Metabonomic analysis was performed by using ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS)-based untargeted metabolomics. Multivariate statistical analysis was performed to screen potential metabolites associated with DHA treatment in SH-SY5Y cells. RESULTS It was shown that DHA inhibited SH-SY5Y cell proliferation and increased poly (ADP-ribose) polymerase (PARP-1) and caspase 3 in a dose-dependent manner. In Further, DHA promoted ROS generation and γH2AX expression. In addition, a total of 125 proposed metabolites in SH-SY5Y cells and 45 vital metabolic pathways were identified through UHPLC-MS/MS-based untargeted metabolomic analysis. CONCLUSIONS These data suggest that DHA could regulate taurine, linoleic acid, phenylalanine metabolism, and tryptophan metabolism, which are involved in the anti-proliferation effect of DHA in SH-SY5Y cells.
Collapse
Affiliation(s)
- De-Lai Xu
- Department of Pharmacy, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Kai Fan
- Department of Pharmacy, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hua Zhang
- Department of Pharmacy, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liu-Xing Tang
- Department of Pharmacy, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Wang
- Department of Pharmacy, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhen Xiang
- Department of Pharmacy, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ai-Ming Shi
- Department of Pharmacy, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu-Chen Qu
- Department of Pharmacy, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Cun-Jin Su
- Department of Pharmacy, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie Pan
- Department of Pharmacy, the Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
9
|
Gunder LC, Blaine-Sauer S, Johnson HR, Shin MK, Auyeung AS, Zhang W, Leverson GE, Ward-Shaw ET, King RE, McGregor SM, Matkowskyj KA, Lambert PF, Carchman EH. Efficacy of Topically Administered Dihydroartemisinin in Treating Papillomavirus-Induced Anogenital Dysplasia in Preclinical Mouse Models. Viruses 2022; 14:1632. [PMID: 35893697 PMCID: PMC9332511 DOI: 10.3390/v14081632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/18/2022] [Accepted: 07/22/2022] [Indexed: 02/06/2023] Open
Abstract
The artemisinin family of compounds is cytopathic in certain cancer cell lines that are positive for human papillomaviruses (HPV) and can potentially drive the regression of dysplastic lesions. We evaluated the efficacy of topical dihydroartemisinin (DHA) on cervical dysplasia and anal dysplasia in two papillomavirus mouse models: K14E6/E7 transgenic mice, which express HPV16 oncogenes; and immunodeficient NOD/SCID gamma (NSG) mice infected with Mus musculus papillomavirus (MmuPV1). Mice started treatment with DHA at 25 weeks of age (K14E6/E7) or 20 weeks post infection (MmuPV1-infected), when the majority of mice are known to have papillomavirus-induced low- to high-grade dysplasia. Mice were treated with or without topical DHA at the cervix or anus and with or without topical treatment with the chemical carcinogen 7,12 dimethylbenz(a)anthracene (DMBA) at the anus of in transgenic mice to induce neoplastic progression. Mice were monitored for overt tumor growth, and tissue was harvested after 20 weeks of treatment and scored for severity of histological disease. For MmuPV1-infected mice, anogenital lavages were taken to monitor for viral clearance. Tissues were also evaluated for viral gene expression at the RNA and/or protein levels. Treatment with topical DHA did not reduce dysplasia in the anogenital tract in either papillomavirus-induced mouse model and did not prevent progression to anal cancer in the DMBA-treated K14E6/E7 mice.
Collapse
Affiliation(s)
- Laura C. Gunder
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, 600 Highland Ave, Madison, WI 53792, USA; (L.C.G.); (H.R.J.); (A.S.A.); (G.E.L.)
| | - Simon Blaine-Sauer
- McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin, 1111 Highland Ave, Madison, WI 53705, USA; (S.B.-S.); (M.-K.S.); (E.T.W.-S.); (R.E.K.); (P.F.L.)
| | - Hillary R. Johnson
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, 600 Highland Ave, Madison, WI 53792, USA; (L.C.G.); (H.R.J.); (A.S.A.); (G.E.L.)
| | - Myeong-Kyun Shin
- McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin, 1111 Highland Ave, Madison, WI 53705, USA; (S.B.-S.); (M.-K.S.); (E.T.W.-S.); (R.E.K.); (P.F.L.)
| | - Andrew S. Auyeung
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, 600 Highland Ave, Madison, WI 53792, USA; (L.C.G.); (H.R.J.); (A.S.A.); (G.E.L.)
| | - Wei Zhang
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, 3170 UW Medical Foundation Centennial Building (MFCB), 1685 Highland Avenue, Madison, WI 53705, USA; (W.Z.); (S.M.M.); (K.A.M.)
| | - Glen E. Leverson
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, 600 Highland Ave, Madison, WI 53792, USA; (L.C.G.); (H.R.J.); (A.S.A.); (G.E.L.)
| | - Ella T. Ward-Shaw
- McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin, 1111 Highland Ave, Madison, WI 53705, USA; (S.B.-S.); (M.-K.S.); (E.T.W.-S.); (R.E.K.); (P.F.L.)
| | - Renee E. King
- McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin, 1111 Highland Ave, Madison, WI 53705, USA; (S.B.-S.); (M.-K.S.); (E.T.W.-S.); (R.E.K.); (P.F.L.)
| | - Stephanie M. McGregor
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, 3170 UW Medical Foundation Centennial Building (MFCB), 1685 Highland Avenue, Madison, WI 53705, USA; (W.Z.); (S.M.M.); (K.A.M.)
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, 600 Highland Ave, Madison, WI 53705, USA
| | - Kristina A. Matkowskyj
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin, 3170 UW Medical Foundation Centennial Building (MFCB), 1685 Highland Avenue, Madison, WI 53705, USA; (W.Z.); (S.M.M.); (K.A.M.)
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, 600 Highland Ave, Madison, WI 53705, USA
- William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, WI 53705, USA
| | - Paul F. Lambert
- McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin, 1111 Highland Ave, Madison, WI 53705, USA; (S.B.-S.); (M.-K.S.); (E.T.W.-S.); (R.E.K.); (P.F.L.)
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, 600 Highland Ave, Madison, WI 53705, USA
| | - Evie H. Carchman
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, 600 Highland Ave, Madison, WI 53792, USA; (L.C.G.); (H.R.J.); (A.S.A.); (G.E.L.)
- University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, 600 Highland Ave, Madison, WI 53705, USA
- William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, WI 53705, USA
| |
Collapse
|
10
|
Meng Y, Liang Y, Liao B, He W, Liu Q, Shen X, Xu J, Chen S. Genome-Wide Identification, Characterization and Expression Analysis of Lipoxygenase Gene Family in Artemisia annua L. PLANTS (BASEL, SWITZERLAND) 2022; 11:plants11050655. [PMID: 35270126 PMCID: PMC8912875 DOI: 10.3390/plants11050655] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 05/17/2023]
Abstract
Lipoxygenase (LOX) is a ubiquitous oxygenase found in animals and plants and plays a pivotal role in diverse biological processes, including defense and development. Artemisinin, which can only be obtained from Artemisia annua L., is the most effective therapeutic drug for malaria without serious side effects. This study identified and analyzed LOX gene family members in the A. annua genome at the chromosomal level. Twenty LOX genes with various molecular weights, isoelectric points, and amino acid numbers were identified and named AaLOX, which were located in the cytoplasm or chloroplast. The average protein length of all AaLOX was 850 aa. Phylogenetic tree analysis revealed that the AaLOX was divided into two major groups, 9-LOX and 13-LOX. The exon numbers ranged from 1 to 12, indicating that different AaLOX genes have different functions. The secondary structure was mainly composed of alpha helix and random coil, and the tertiary structure was similar for most AaLOX. Upstream promoter region analysis revealed that a large number of cis-acting elements were closely related to plant growth and development, light response, hormone, and other stress responses. Transcriptome data analysis of different tissues suggested that the gene family was differently expressed in the roots, stems, leaves, and flowers of two A. annua strains HAN1 and LQ9. qRT-PCR confirmed that AaLOX5 and AaLOX17 had the highest expression in flowers and leaves. This study provides a theoretical basis for the further functional analysis of the AaLOX gene family.
Collapse
Affiliation(s)
- Ying Meng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250000, China;
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100000, China; (Y.L.); (B.L.); (Q.L.); (X.S.)
| | - Yu Liang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100000, China; (Y.L.); (B.L.); (Q.L.); (X.S.)
- College of Pharmaceutical Science, Dali University, Dali 671000, China;
| | - Baosheng Liao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100000, China; (Y.L.); (B.L.); (Q.L.); (X.S.)
| | - Wenrui He
- College of Pharmaceutical Science, Dali University, Dali 671000, China;
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Qianwen Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100000, China; (Y.L.); (B.L.); (Q.L.); (X.S.)
| | - Xiaofeng Shen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100000, China; (Y.L.); (B.L.); (Q.L.); (X.S.)
| | - Jiang Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100000, China; (Y.L.); (B.L.); (Q.L.); (X.S.)
- Correspondence: (J.X.); (S.C.)
| | - Shilin Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100000, China; (Y.L.); (B.L.); (Q.L.); (X.S.)
- Correspondence: (J.X.); (S.C.)
| |
Collapse
|
11
|
Zhou X, Suo F, Haslinger K, Quax WJ. Artemisinin-Type Drugs in Tumor Cell Death: Mechanisms, Combination Treatment with Biologics and Nanoparticle Delivery. Pharmaceutics 2022; 14:395. [PMID: 35214127 PMCID: PMC8875250 DOI: 10.3390/pharmaceutics14020395] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
Artemisinin, the most famous anti-malaria drug initially extracted from Artemisia annua L., also exhibits anti-tumor properties in vivo and in vitro. To improve its solubility and bioavailability, multiple derivatives have been synthesized. However, to reveal the anti-tumor mechanism and improve the efficacy of these artemisinin-type drugs, studies have been conducted in recent years. In this review, we first provide an overview of the effect of artemisinin-type drugs on the regulated cell death pathways, which may uncover novel therapeutic approaches. Then, to overcome the shortcomings of artemisinin-type drugs, we summarize the recent advances in two different therapeutic approaches, namely the combination therapy with biologics influencing regulated cell death, and the use of nanocarriers as drug delivery systems. For the former approach, we discuss the superiority of combination treatments compared to monotherapy in tumor cells based on their effects on regulated cell death. For the latter approach, we give a systematic overview of nanocarrier design principles used to deliver artemisinin-type drugs, including inorganic-based nanoparticles, liposomes, micelles, polymer-based nanoparticles, carbon-based nanoparticles, nanostructured lipid carriers and niosomes. Both approaches have yielded promising findings in vitro and in vivo, providing a strong scientific basis for further study and upcoming clinical trials.
Collapse
Affiliation(s)
| | | | - Kristina Haslinger
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (X.Z.); (F.S.)
| | - Wim J. Quax
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (X.Z.); (F.S.)
| |
Collapse
|
12
|
Zhang Y, Wang Y, Li Y, Huang C, Xiao X, Zhong Z, Tang J, Lu H, Tang Y, Yang J. Dihydroartemisinin and Artesunate Inhibit Aerobic Glycolysis via Suppressing c-Myc Signaling in non-small Cell Lung Cancer. Biochem Pharmacol 2022; 198:114941. [PMID: 35149053 DOI: 10.1016/j.bcp.2022.114941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 11/02/2022]
Abstract
Non-small cell lung cancer (NSCLC) cells exhibit aberrant metabolism characterized by high glycolysis even in the presence of abundant oxygen. Inhibition of aerobic glycolysis remains challenging when identifying potential cancer-specific inhibitors while maintaining or even boosting the anti-cancer immunity. Artemisinin derivatives DHA and AS have shown excellent anti-tumor and immunoenhancing roles in numerous malignancies, but the molecular mechanism of DHA and AS in regulating cancer glucose metabolism is largely unknown. In this study, we proved that DHA and AS inhibit NSCLC growth via prohibiting cancer cell aerobic glycolysis through ERK/c-Myc pathway. First, we proved that DHA and AS have comparable anti-cancer growth roles in both NSCLC cell lines and mouse Lewis Lung Cancer model. Then, our data clearly showed that DHA and AS dose- and time-dependently reduce the uptake of glucose, the production of ATP, and the secretion of lactate, the expression of glucose transporter GLUT1 and two key glycolysis-related enzymes hexokinase and lactate dehydrogenase, as well as the level of c-Myc. Finally, we generated c-Mychigh stable-expressing NSCLC cell line and treated it with DHA or AS, respectively. Our data clearly showed that c-Myc overexpression can partially reverse the glycolysis-repressing role of DHA and AS which strongly supported our proposal that AS and DHA suppress aerobic glycolysis in a c-Myc-dependent manner in NSCLC cells. This study extends our knowledge of artemisinin derivatives in regulating tumor glucose metabolism and provides potential strategy in the therapy of lung cancer.
Collapse
Affiliation(s)
- Yuxi Zhang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yi Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yanping Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cong Huang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaoqian Xiao
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhanqiong Zhong
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jingyi Tang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Haolan Lu
- School of Medical and Life sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yibei Tang
- School of Medical and Life sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jiahui Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
13
|
Hao L, Guo Y, Peng Q, Zhang Z, Ji J, Liu Y, Xue Y, Li C, Zheng K, Shi X. Dihydroartemisinin reduced lipid droplet deposition by YAP1 to promote the anti-PD-1 effect in hepatocellular carcinoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 96:153913. [PMID: 35026515 DOI: 10.1016/j.phymed.2021.153913] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 12/04/2021] [Accepted: 12/24/2021] [Indexed: 05/16/2023]
Abstract
BACKGROUND Anti-PD-1 was used to treat for many cancers, but the overall response rate of monoclonal antibodies blocking the inhibitory PD-1/PD-L1 was less than 20%. Lipid droplet (LD) deposition reduced chemotherapy efficacy, but whether LD deposition affects anti-PD-1 treatment and its mechanism remains unclear. Dihydroartemisinin (DHA) was FDA proved antimalarial medicine, but its working mechanism on LD deposition has not been clarified. PURPOSE This study aimed to elucidate the mechanism of DHA reducing LDs deposition and improving the efficacy of anti-PD-1. METHODS LD numbers and area were separately detected by electron microscopy and oil Red O staining. The expression of YAP1 and PLIN2 was detected by immunohistochemical staining in liver cancer tissues. Transcription and protein expression levels of YAP1 and PLIN2 in cells were detected by qRT-PCR and Western blot after DHA treated HepG2215 cells and Yap1LKO mice. RESULTS LD accumulation was found in the liver tumor cells of DEN/TOPBCOP-induced liver tumor mice with anti-PD-1 treatment. But DHA treatment or YAP1 knockdown reduced LD deposition and PLIN2 expression in HepG2215 cells. Furthermore, DHA reduced the LD deposition, PLIN2 expression and triglycerides (TG) content in the liver tumor cells of Yap1LKO mice with liver tumor. CONCLUSION Anti-PD-1 promoted LD deposition, while YAP1 knockdown/out reduced LD deposition in HCC. DHA reduced LD deposition by inhibiting YAP1, enhancing the effect of anti-PD-1 therapy.
Collapse
Affiliation(s)
- Liyuan Hao
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yinglin Guo
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Qing Peng
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Zhiqin Zhang
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Jingmin Ji
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yiwei Liu
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yu Xue
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Caige Li
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Kangning Zheng
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Xinli Shi
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China.
| |
Collapse
|
14
|
Zhou X, Soto-Gamez A, Nijdam F, Setroikromo R, Quax WJ. Dihydroartemisinin-Transferrin Adducts Enhance TRAIL-Induced Apoptosis in Triple-Negative Breast Cancer in a P53-Independent and ROS-Dependent Manner. Front Oncol 2022; 11:789336. [PMID: 35047402 PMCID: PMC8762273 DOI: 10.3389/fonc.2021.789336] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/03/2021] [Indexed: 01/25/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype independent of estrogen receptor, progesterone receptor, or human epidermal growth factor receptor 2. It has a poor prognosis and high recurrence. Due to its limited treatment options in the clinic, novel therapies are urgently needed. Single treatment with the death receptor ligand TRAIL was shown to be poorly effective. Recently, we have shown that artemisinin derivatives enhance TRAIL-induced apoptosis in colon cancer cells. Here, we utilized transferrin (TF) to enhance the effectiveness of dihydroartemisinin (DHA) in inducing cell death in TNBC cell lines (MDA-MB-231, MDA-MB-436, MDA-MB-468 and BT549). We found that the combination of DHA-TF and the death receptor 5-specific TRAIL variant DHER leads to an increase in DR5 expression in all four TNBC cell lines, while higher cytotoxicity was observed in MDA-MB-231, and MDA-MB-436. All the data point to the finding that DHA-TF stimulates cell death in TNBC cells, while the combination of DHA-TF with TRAIL variants will trigger more cell death in TRAIL-sensitive cells. Overall, DHA-TF in combination with TRAIL variants represents a potential novel combination therapy for triple-negative breast cancer.
Collapse
Affiliation(s)
- Xinyu Zhou
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
| | - Abel Soto-Gamez
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands.,European Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), Groningen, Netherlands
| | - Fleur Nijdam
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
| | - Rita Setroikromo
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
| | - Wim J Quax
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
| |
Collapse
|
15
|
Mollah F, Varamini P. Overcoming Therapy Resistance and Relapse in TNBC: Emerging Technologies to Target Breast Cancer-Associated Fibroblasts. Biomedicines 2021; 9:1921. [PMID: 34944738 PMCID: PMC8698629 DOI: 10.3390/biomedicines9121921] [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: 11/03/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most diagnosed cancer and is the leading cause of cancer mortality in women. Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer. Often, TNBC is not effectively treated due to the lack of specificity of conventional therapies and results in relapse and metastasis. Breast cancer-associated fibroblasts (BCAFs) are the predominant cells that reside in the tumor microenvironment (TME) and regulate tumorigenesis, progression and metastasis, and therapy resistance. BCAFs secrete a wide range of factors, including growth factors, chemokines, and cytokines, some of which have been proved to lead to a poor prognosis and clinical outcomes. This TME component has been emerging as a promising target due to its crucial role in cancer progression and chemotherapy resistance. A number of therapeutic candidates are designed to effectively target BCAFs with a focus on their tumor-promoting properties and tumor immune response. This review explores various agents targeting BCAFs in TNBC, including small molecules, nucleic acid-based agents, antibodies, proteins, and finally, nanoparticles.
Collapse
Affiliation(s)
- Farhana Mollah
- Faculty of Medicine and Health, School of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia;
| | - Pegah Varamini
- Faculty of Medicine and Health, School of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia;
- Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
16
|
Nandi D, Cheema PS, Singal A, Bharti H, Nag A. Artemisinin Mediates Its Tumor-Suppressive Activity in Hepatocellular Carcinoma Through Targeted Inhibition of FoxM1. Front Oncol 2021; 11:751271. [PMID: 34900697 PMCID: PMC8652299 DOI: 10.3389/fonc.2021.751271] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 11/04/2021] [Indexed: 12/28/2022] Open
Abstract
The aberrant up-regulation of the oncogenic transcription factor Forkhead box M1 (FoxM1) is associated with tumor development, progression and metastasis in a myriad of carcinomas, thus establishing it as an attractive target for anticancer drug development. FoxM1 overexpression in hepatocellular carcinoma is reflective of tumor aggressiveness and recurrence, poor prognosis and low survival in patients. In our study, we have identified the antimalarial natural product, Artemisinin, to efficiently curb FoxM1 expression and activity in hepatic cancer cells, thereby exhibiting potential anticancer efficacy. Here, we demonstrated that Artemisinin considerably mitigates FoxM1 transcriptional activity by disrupting its interaction with the promoter region of its downstream targets, thereby suppressing the expression of numerous oncogenic drivers. Augmented level of FoxM1 is implicated in drug resistance of cancer cells, including hepatic tumor cells. Notably, FoxM1 overexpression rendered HCC cells poorly responsive to Artemisinin-mediated cytotoxicity while FoxM1 depletion in resistant liver cancer cells sensitized them to Artemisinin treatment, manifested in lower proliferative and growth index, drop in invasive potential and repressed expression of EMT markers with a concomitantly increased apoptosis. Moreover, Artemisinin, when used in combination with Thiostrepton, an established FoxM1 inhibitor, markedly reduced anchorage-independent growth and displayed more pronounced death in liver cancer cells. We found this effect to be evident even in the resistant HCC cells, thereby putting forth a novel combination therapy for resistant cancer patients. Altogether, our findings provide insight into the pivotal involvement of FoxM1 in the tumor suppressive activities of Artemisinin and shed light on the potential application of Artemisinin for improved therapeutic response, especially in resistant hepatic malignancies. Considering that Artemisinin compounds are in current clinical use with favorable safety profiles, the results from our study will potentiate its utility in juxtaposition with established FoxM1 inhibitors, promoting maximal therapeutic efficacy with minimal adverse effects in liver cancer patients.
Collapse
Affiliation(s)
| | | | - Aakriti Singal
- Department of Biochemistry, University of Delhi, New Delhi, India
| | - Hina Bharti
- Department of Biochemistry, University of Delhi, New Delhi, India
| | - Alo Nag
- Department of Biochemistry, University of Delhi, New Delhi, India
| |
Collapse
|
17
|
Yang Y, Liu Q, Shi X, Zheng Q, Chen L, Sun Y. Advances in plant-derived natural products for antitumor immunotherapy. Arch Pharm Res 2021; 44:987-1011. [PMID: 34751930 DOI: 10.1007/s12272-021-01355-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 10/29/2021] [Indexed: 12/28/2022]
Abstract
In recent years, immunotherapy has emerged as a novel antitumor strategy in addition to traditional surgery, radiotherapy and chemotherapy. It uniquely focuses on immune cells and immunomodulators in the tumor microenvironment and helps eliminate tumors at the root by rebuilding the immune system. Despite remarkable breakthroughs, cancer immunotherapy still faces many challenges: lack of predictable and prognostic biomarkers, adverse side effects, acquired treatment resistance, high costs, etc. Therefore, more efficacious and efficient, safer and cheaper antitumor immunomodulatory drugs have become an urgent requirement. For decades, plant-derived natural products obtained from land and sea have provided the most important source for the development of antitumor drugs. Currently, more attention is being paid to the discovery of potential cancer immunotherapy modulators from plant-derived natural products, such as polysaccharides, phenols, terpenoids, quinones and alkaloids. Some of these agents have outstanding advantages of multitargeting and low side effects and low cost compared to conventional immunotherapeutic agents. We intend to summarize the progress of comprehensive research on these plant-derived natural products and their derivatives and discuss their possible mechanisms in regulating the immune system and their efficacy as monotherapies or in combination with regular chemotherapeutic agents.
Collapse
Affiliation(s)
- Yi Yang
- Fujian Provincial Key Laboratory of Medical Instrument and Pharmaceutical Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, 350108, China
| | - Qinying Liu
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Xianai Shi
- Fujian Provincial Key Laboratory of Medical Instrument and Pharmaceutical Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, 350108, China
| | - Qiuhong Zheng
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Li Chen
- Fujian Provincial Key Laboratory of Medical Instrument and Pharmaceutical Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, 350108, China.
| | - Yang Sun
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China.
- Department of Gyn-Surgical Oncology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China.
| |
Collapse
|
18
|
Peng J, Wang Q, Zhou J, Zhao S, Di P, Chen Y, Tao L, Du Q, Shen X, Chen Y. Targeted Lipid Nanoparticles Encapsulating Dihydroartemisinin and Chloroquine Phosphate for Suppressing the Proliferation and Liver Metastasis of Colorectal Cancer. Front Pharmacol 2021; 12:720777. [PMID: 34690764 PMCID: PMC8531263 DOI: 10.3389/fphar.2021.720777] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/24/2021] [Indexed: 12/23/2022] Open
Abstract
Antimalarial drugs Dihydroartemisinin (DHA) and chloroquine phosphate (CQ) exhibit evident anti-cancer activity, particularly as combination therapy. DHA and CQ combination therapy has been proved to exhibit higher cytotoxic effect in tumor cells and lower toxicity to normal cells than combination of artemisinin derivatives (ARTs) and anticancer chemotherapy drugs. However, different physiochemical properties of DHA and CQ, leading to distinctive in vivo outcomes, considerably limited their synergistic effect in cancer treatment. Herein, we developed a lipid nanoparticle (LNP) for co-delivery of DHA and CQ to inhibit proliferation and metastasis of colorectal cancer. Considering the beneficial effects of acid/reactive oxide species (ROS)-sensitive phospholipids and targeting ligands for colorectal cancer cells, an RGD peptide-modified pH/ROS dual-sensitive LNP loaded with DHA and CQ (RLNP/DC) was prepared. It exhibited optimal cytotoxicity and suppression of invasion and metastasis in HCT116 cells in vitro, attributable to irreversible upregulation of intracellular ROS levels, downregulation of VEGF expression, and upregulation of paxillin expression. A mouse model of orthotopic metastasis of colorectal cancer was established to evaluate anti-proliferation and anti-metastasis effects of RLNP/DC in vivo. Thus, an optimized nanoplatform for DHA and CQ combination therapy was developed in this study that offered potential antitumor efficacy against colorectal cancer.
Collapse
Affiliation(s)
- Jianqing Peng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Qin Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Jia Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Shuli Zhao
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Pan Di
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Yan Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Ling Tao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Qianming Du
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiangchun Shen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| | - Yi Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China
| |
Collapse
|
19
|
Xu P, Wang X, Li T, Li L, Wu H, Tu J, Zhang R, Zhang L, Guo Z, Chen Q. Bioinspired Microenvironment Responsive Nanoprodrug as an Efficient Hydrophobic Drug Self-Delivery System for Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:33926-33936. [PMID: 34254767 DOI: 10.1021/acsami.1c09612] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Artemisinin compounds have shown satisfactory safety records in anti-malarial clinical practice over decades and have revealed value as inexpensive anti-tumor adjuvant chemotherapeutic drugs. However, the rational design and precise preparation of nanomedicines based on the artemisinin drugs are still limited due to their non-aromatic and fragile chemical structure. Herein, a bioinspired coordination-driven self-assembly strategy was developed to manufacture the artemisinin-based nanoprodrug with a significantly increased drug loading efficacy (∼70 wt %) and decreased preparation complexity compared to conventional nanodrugs. The nanoprodrug has suitable size distribution and robust colloidal stability for cancer targeting in vivo. The nanoprodrug was able to quickly disassemble in the tumor microenvironment with weak acidity and a high glutathione concentration, which guarantees a better tumor inhibitory effect than direct administration and fewer side effects on normal tissues in vivo. This work highlights a new strategy to harness a robust, simplified, organic solvent-free, and highly repeatable route for nanoprodrug manufacturing, which may offer opportunities to develop cost-effective, safe, and clinically available nanomedicines.
Collapse
Affiliation(s)
- Pengping Xu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xueying Wang
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Tuanwei Li
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Lingli Li
- Department of Pharmacy, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, China
| | - Huihui Wu
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Jinwei Tu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Ruoyang Zhang
- Changzhou Senior High School of Jiangsu Province, Changzhou, Jiangsu 213003, China
| | - Lei Zhang
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Department of Pharmacy, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, China
| | - Zhen Guo
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Qianwang Chen
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
- Hefei National Laboratory for Physical Science at Microscale and Department of Materials Science & Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
20
|
Zhao J, Zhao Y, Ma X, Zhang B, Feng H. Targeting ferroptosis in osteosarcoma. J Bone Oncol 2021; 30:100380. [PMID: 34345580 PMCID: PMC8319509 DOI: 10.1016/j.jbo.2021.100380] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/03/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary bone tumour in children and adolescents, with high degree of malignancy and an extremely poor prognosis. Ferroptosis, a non-traditional mode of regulated cell death (RCD) characterised by iron-dependent accumulation of lipid reactive oxygen species (ROS), is closely associated with a variety of cancers. It has been demonstrated that ferroptosis can regulate OS progression and exert an essential role in the treatment of OS, which is potentially of great value. By targeting ferroptosis in OS, the present review article summarises the relevant mechanisms and therapeutic applications along with discussing current limitations and future directions, which may provide a new strategy for the treatment of OS.
Collapse
Affiliation(s)
- Jiazheng Zhao
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, 12 Health Road, Shijiazhuang, Hebei 050011, PR China
| | - Yi Zhao
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, 12 Health Road, Shijiazhuang, Hebei 050011, PR China
| | - Xiaowei Ma
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, 12 Health Road, Shijiazhuang, Hebei 050011, PR China
| | - Benzheng Zhang
- Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, Hebei 050011, PR. China
| | - Helin Feng
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, 12 Health Road, Shijiazhuang, Hebei 050011, PR China
| |
Collapse
|
21
|
Wei MX, Yu JY, Liu XX, Li XQ, Yang JH, Zhang MW, Yang PW, Zhang SS, He Y. Synthesis and biological evaluation of novel artemisone-piperazine-tetronamide hybrids. RSC Adv 2021; 11:18333-18341. [PMID: 35480921 PMCID: PMC9033422 DOI: 10.1039/d1ra00750e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/13/2021] [Indexed: 11/21/2022] Open
Abstract
For the first time, six novel artemisone-piperazine-tetronamide hybrids (12a-f) were efficiently synthesised from dihydroartemisinin (DHA) and investigated for their in vitro cytotoxicity against some human cancer cells and benign cells. All the targets showed good cytotoxic activity in vitro. Hybrid 12a exhibited much better inhibitory activity against human liver cancer cell line SMMC-7721 (IC50 = 0.03 ± 0.04 μM for 24 h) than the parent DHA (IC50 > 0.7 μM), and two references, vincristine (VCR; IC50 = 0.27 ± 0.03 μM) & cytosine arabinoside (ARA; IC50 = 0.63 ± 0.04 μM). Furthermore, hybrid 12a had low toxicity against human benign liver cell line LO2 (IC50 = 0.70 ± 0.02 μM for 24 h) compared with VCR, ARA, and DHA in vitro. Moreover, the inhibitory activity of hybrid 12a was obviously enhanced when human liver cancer cell line MHCC97H absorbed Fe2+ in vitro.
Collapse
Affiliation(s)
- Meng-Xue Wei
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, Ningxia Engineering Research Center for Natural Medicine, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Chemical Engineering, Ningxia University 489 Helanshan West Road Yinchuan 750021 China
| | - Jia-Ying Yu
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, Ningxia Engineering Research Center for Natural Medicine, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Chemical Engineering, Ningxia University 489 Helanshan West Road Yinchuan 750021 China
| | - Xin-Xin Liu
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, Ningxia Engineering Research Center for Natural Medicine, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Chemical Engineering, Ningxia University 489 Helanshan West Road Yinchuan 750021 China
| | - Xue-Qiang Li
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, Ningxia Engineering Research Center for Natural Medicine, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Chemical Engineering, Ningxia University 489 Helanshan West Road Yinchuan 750021 China
| | - Jin-Hui Yang
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, Ningxia Engineering Research Center for Natural Medicine, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Chemical Engineering, Ningxia University 489 Helanshan West Road Yinchuan 750021 China
| | - Meng-Wei Zhang
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, Ningxia Engineering Research Center for Natural Medicine, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Chemical Engineering, Ningxia University 489 Helanshan West Road Yinchuan 750021 China
| | - Pei-Wen Yang
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, Ningxia Engineering Research Center for Natural Medicine, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Chemical Engineering, Ningxia University 489 Helanshan West Road Yinchuan 750021 China
| | - Si-Si Zhang
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, Ningxia Engineering Research Center for Natural Medicine, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Chemical Engineering, Ningxia University 489 Helanshan West Road Yinchuan 750021 China
| | - Yu He
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, Ningxia Engineering Research Center for Natural Medicine, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Chemical Engineering, Ningxia University 489 Helanshan West Road Yinchuan 750021 China
| |
Collapse
|
22
|
Li Y, Wang W, Li A, Huang W, Chen S, Han F, Wang L. Dihydroartemisinin induces pyroptosis by promoting the AIM2/caspase-3/DFNA5 axis in breast cancer cells. Chem Biol Interact 2021; 340:109434. [PMID: 33689708 DOI: 10.1016/j.cbi.2021.109434] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/01/2020] [Accepted: 03/04/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Breast cancer is a complex disease. Recent research has examined the anticancer effects of dihydroartemisinin (DHA) on breast cancer. However, the molecular mechanism of the antitumour effect of DHA is unclear. METHODS MCF-7 and MDA-MB-231 cell lines were used for in vitro research. BALB/c nude mice were used to establish breast cancer xenografts. The mRNA and protein levels were analysed by qRT-PCR and western blotting, respectively. Flow cytometry was performed to examine cell apoptosis. ELISA kits were used to evaluate the production of interleukin-1β (IL-1β) and IL-18. LDH and ATP release were individually measured with the corresponding kits. A colony formation assay was used to examine the proliferation of breast cancer cells. RESULTS DHA inhibited proliferation and induced pyroptosis in breast cancer cells. Mechanistically, DHA activated the expression of absent in melanoma 2 (AIM2), caspase-3 and gasdermin E (DFNA5). In addition, AIM2 promoted DFNA5 expression by activating caspase-3. Knockdown of AIM2 and DFNA5 significantly enhanced breast cancer cell resistance to DHA. In vivo experiments showed that the tumorigenicity of breast cancer cells was significantly suppressed by DHA. Moreover, the AIM2/caspase-3/DFNA5 axis was activated by DHA and then induced pyroptosis. CONCLUSIONS Our findings indicate that DHA inhibits tumorigenesis by inducing pyroptosis in breast cancer cells, highlighting a promising therapeutic strategy for breast cancer.
Collapse
Affiliation(s)
- Yaqiong Li
- Department of Thyroid and Breast and Vascular Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, PR China
| | - Wei Wang
- Department of Thyroid and Breast and Vascular Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, PR China
| | - Aixia Li
- Department of Otolaryngology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, PR China
| | - Wei Huang
- Department of Thyroid and Breast and Vascular Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, PR China
| | - Shiman Chen
- Department of Thyroid and Breast and Vascular Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, PR China
| | - Fei Han
- Department of Thyroid and Breast and Vascular Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, PR China
| | - Lingcheng Wang
- Department of Thyroid and Breast and Vascular Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei Province, PR China.
| |
Collapse
|
23
|
Wei MX, Yu JY, Liu XX, Li XQ, Zhang MW, Yang PW, Yang JH. Synthesis of artemisinin-piperazine-furan ether hybrids and evaluation of in vitro cytotoxic activity. Eur J Med Chem 2021; 215:113295. [PMID: 33636536 DOI: 10.1016/j.ejmech.2021.113295] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/25/2021] [Accepted: 02/07/2021] [Indexed: 11/29/2022]
Abstract
For the first time, eight novel artemisinin-piperazine-furane ether hybrids (5a-h) were efficiently synthesized and investigated for their in vitro cytotoxic activity against some human cancer and benign cells. The absolute configuration of hybrid 5c was determined by X-ray crystallographic analysis. Hybrids 5a-h exhibited more pronounced growth-inhibiting action on hepatocarcinoma cell lines than their parent dihydroartemisinin (DHA) and the reference cytosine arabinoside (ARA). The hybrid 5a showed the best cytotoxic activity against human hepatocarcinoma cells SMMC-7721 (IC50 = 0.26 ± 0.03 μM) after 24 h. Furthermore, hybrid 5a also showed good cytotoxic activity against human breast cancer cells MCF-7 and low cytotoxicity against human breast benign cells MCF-10A in vitro. We found the cytotoxicity of hybrid 5a did not change when tumour cells absorb iron sulfate (FeSO4); thus, we conclude the anti-tumour mechanism induced by iron ions (Fe2+) is unclear.
Collapse
Affiliation(s)
- Meng-Xue Wei
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, Ningxia Engineering Research Center for Natural Medicine, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Chemical Engineering, Ningxia University, 489 Helanshan West Road, Yinchuan, 750021, China.
| | - Jia-Ying Yu
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, Ningxia Engineering Research Center for Natural Medicine, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Chemical Engineering, Ningxia University, 489 Helanshan West Road, Yinchuan, 750021, China
| | - Xin-Xin Liu
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, Ningxia Engineering Research Center for Natural Medicine, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Chemical Engineering, Ningxia University, 489 Helanshan West Road, Yinchuan, 750021, China
| | - Xue-Qiang Li
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, Ningxia Engineering Research Center for Natural Medicine, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Chemical Engineering, Ningxia University, 489 Helanshan West Road, Yinchuan, 750021, China
| | - Meng-Wei Zhang
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, Ningxia Engineering Research Center for Natural Medicine, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Chemical Engineering, Ningxia University, 489 Helanshan West Road, Yinchuan, 750021, China
| | - Pei-Wen Yang
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, Ningxia Engineering Research Center for Natural Medicine, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Chemical Engineering, Ningxia University, 489 Helanshan West Road, Yinchuan, 750021, China
| | - Jin-Hui Yang
- State Key Laboratory of High-efficiency Utilization of Coal and Green Chemical Engineering, Ningxia Engineering Research Center for Natural Medicine, National Demonstration Center for Experimental Chemistry Education, College of Chemistry and Chemical Engineering, Ningxia University, 489 Helanshan West Road, Yinchuan, 750021, China
| |
Collapse
|
24
|
Zhu S, Yu Q, Huo C, Li Y, He L, Ran B, Chen J, Li Y, Liu W. Ferroptosis: A Novel Mechanism of Artemisinin and its Derivatives in Cancer Therapy. Curr Med Chem 2021; 28:329-345. [PMID: 31965935 DOI: 10.2174/0929867327666200121124404] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/06/2019] [Accepted: 12/12/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Artemisinin is a sesquiterpene lactone compound with a special peroxide bridge that is tightly linked to the cytotoxicity involved in fighting malaria and cancer. Artemisinin and its derivatives (ARTs) are considered to be potential anticancer drugs that promote cancer cell apoptosis, induce cell cycle arrest and autophagy, inhibit cancer cell invasion and migration. Additionally, ARTs significantly increase intracellular Reactive Oxygen Species (ROS) in cancer cells, which result in ferroptosis, a new form of cell death, depending on the ferritin concentration. Ferroptosis is regarded as a cancer suppressor and as well as considered a new mechanism for cancer therapy. METHODS The anticancer activities of ARTs and reference molecules were compared by literature search and analysis. The latest research progress on ferroptosis was described, with a special focus on the molecular mechanism of artemisinin-induced ferroptosis. RESULTS Artemisinin derivatives, artemisinin-derived dimers, hybrids and artemisinin-transferrin conjugates, could significantly improve anticancer activity, and their IC50 values are lower than those of reference molecules such as doxorubicin and paclitaxel. The biological activities of linkers in dimers and hybrids are important in the drug design processes. ARTs induce ferroptosis mainly by triggering intracellular ROS production, promoting the lysosomal degradation of ferritin and regulating the System Xc-/Gpx4 axis. Interestingly, ARTs also stimulate the feedback inhibition pathway. CONCLUSION Artemisinin and its derivatives could be used in the future as cancer therapies with broader applications due to their induction of ferroptosis. Meanwhile, more attention should be paid to the development of novel artemisinin-related drugs based on the mechanism of artemisinininduced ferroptosis.
Collapse
Affiliation(s)
- Shunqin Zhu
- School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Qin Yu
- School of Chemistry and Chemical Engineering, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Chunsong Huo
- School of Chemistry and Chemical Engineering, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Yuanpeng Li
- School of Chemistry and Chemical Engineering, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Linshen He
- School of Chemistry and Chemical Engineering, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Botian Ran
- School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Ji Chen
- School of Chemistry and Chemical Engineering, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Yonghao Li
- School of Chemistry and Chemical Engineering, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Wanhong Liu
- School of Life Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
25
|
Pagliaro L, Marchesini M, Roti G. Targeting oncogenic Notch signaling with SERCA inhibitors. J Hematol Oncol 2021; 14:8. [PMID: 33407740 PMCID: PMC7789735 DOI: 10.1186/s13045-020-01015-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/02/2020] [Indexed: 12/26/2022] Open
Abstract
P-type ATPase inhibitors are among the most successful and widely prescribed therapeutics in modern pharmacology. Clinical transition has been safely achieved for H+/K+ ATPase inhibitors such as omeprazole and Na+/K+-ATPase inhibitors like digoxin. However, this is more challenging for Ca2+-ATPase modulators due to the physiological role of Ca2+ in cardiac dynamics. Over the past two decades, sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) modulators have been studied as potential chemotherapy agents because of their Ca2+-mediated pan-cancer lethal effects. Instead, recent evidence suggests that SERCA inhibition suppresses oncogenic Notch1 signaling emerging as an alternative to γ-secretase modulators that showed limited clinical activity due to severe side effects. In this review, we focus on how SERCA inhibitors alter Notch1 signaling and show that Notch on-target-mediated antileukemia properties of these molecules can be achieved without causing overt Ca2+ cellular overload.
Collapse
Affiliation(s)
- Luca Pagliaro
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy
| | - Matteo Marchesini
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy
| | - Giovanni Roti
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy.
| |
Collapse
|
26
|
Zhao F, Yu J, Gao W, Yang X, Liang L, Sun X, Su D, Ying Y, Li W, Li J, Zheng J, Qiao L, Cai W, Che S, Mou X. H 2O 2-independent chemodynamic therapy initiated from magnetic iron carbide nanoparticle-assisted artemisinin synergy. RSC Adv 2021; 11:37504-37513. [PMID: 35496387 PMCID: PMC9043768 DOI: 10.1039/d1ra04975e] [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: 06/28/2021] [Accepted: 10/19/2021] [Indexed: 11/21/2022] Open
Abstract
Chemodynamic therapy (CDT) is a booming technology that utilizes Fenton reagents to kill tumor cells by transforming intracellular H2O2 into reactive oxygen species (ROS), but insufficient endogenous H2O2 makes it difficult to attain satisfactory antitumor results.
Collapse
Affiliation(s)
- Fan Zhao
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
- Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jing Yu
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
- Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou 310014, China
| | - Weiliang Gao
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
- Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xue Yang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Liying Liang
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
- Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xiaolian Sun
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing 210009, China
| | - Dan Su
- Department of Oncology, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Yao Ying
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
- Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou 310014, China
| | - Wangchang Li
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
- Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou 310014, China
| | - Juan Li
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
- Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jingwu Zheng
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
- Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou 310014, China
| | - Liang Qiao
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
- Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou 310014, China
| | - Wei Cai
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
- Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou 310014, China
| | - Shenglei Che
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
- Research Center of Magnetic and Electronic Materials, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| |
Collapse
|
27
|
Trendafilova A, Moujir LM, Sousa PMC, Seca AML. Research Advances on Health Effects of Edible Artemisia Species and Some Sesquiterpene Lactones Constituents. Foods 2020; 10:E65. [PMID: 33396790 PMCID: PMC7823681 DOI: 10.3390/foods10010065] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/22/2020] [Accepted: 12/25/2020] [Indexed: 12/20/2022] Open
Abstract
The genus Artemisia, often known collectively as "wormwood", has aroused great interest in the scientific community, pharmaceutical and food industries, generating many studies on the most varied aspects of these plants. In this review, the most recent evidence on health effects of edible Artemisia species and some of its constituents are presented and discussed, based on studies published until 2020, available in the Scopus, Web of Sciences and PubMed databases, related to food applications, nutritional and sesquiterpene lactones composition, and their therapeutic effects supported by in vivo and clinical studies. The analysis of more than 300 selected articles highlights the beneficial effect on health and the high clinical relevance of several Artemisia species besides some sesquiterpene lactones constituents and their derivatives. From an integrated perspective, as it includes therapeutic and nutritional properties, without ignoring some adverse effects described in the literature, this review shows the great potential of Artemisia plants and some of their constituents as dietary supplements, functional foods and as the source of new, more efficient, and safe medicines. Despite all the benefits demonstrated, some gaps need to be filled, mainly related to the use of raw Artemisia extracts, such as its standardization and clinical trials on adverse effects and its health care efficacy.
Collapse
Affiliation(s)
- Antoaneta Trendafilova
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 9, 1113 Sofia, Bulgaria
| | - Laila M. Moujir
- Department of Biochemistry, Microbiology, Genetics and Cell Biology, Facultad de Farmacia, Universidad de La Laguna, 38206 La Laguna, Tenerife, Spain;
| | - Pedro M. C. Sousa
- Faculty of Sciences and Technology, University of Azores, 9500-321 Ponta Delgada, Portugal;
| | - Ana M. L. Seca
- cE3c—Centre for Ecology, Evolution and Environmental Changes/Azorean Biodiversity Group & Faculty of Sciences and Technology, University of Azores, Rua Mãe de Deus, 9500-321 Ponta Delgada, Portugal
- LAQV-REQUIMTE, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
28
|
Jiang X, Lin M, Huang J, Mo M, Liu H, Jiang Y, Cai X, Leung W, Xu C. Smart Responsive Nanoformulation for Targeted Delivery of Active Compounds From Traditional Chinese Medicine. Front Chem 2020; 8:559159. [PMID: 33363102 PMCID: PMC7758496 DOI: 10.3389/fchem.2020.559159] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022] Open
Abstract
Traditional Chinese medicine (TCM) has been used to treat disorders in China for ~1,000 years. Growing evidence has shown that the active ingredients from TCM have antibacterial, antiproliferative, antioxidant, and apoptosis-inducing features. However, poor solubility and low bioavailability limit clinical application of active compounds from TCM. “Nanoformulations” (NFs) are novel and advanced drug-delivery systems. They show promise for improving the solubility and bioavailability of drugs. In particular, “smart responsive NFs” can respond to the special external and internal stimuli in targeted sites to release loaded drugs, which enables them to control the release of drug within target tissues. Recent studies have demonstrated that smart responsive NFs can achieve targeted release of active compounds from TCM at disease sites to increase their concentrations in diseased tissues and reduce the number of adverse effects. Here, we review “internal stimulus–responsive NFs” (based on pH and redox status) and “external stimulus–responsive NFs” (based on light and magnetic fields) and focus on their application for active compounds from TCM against tumors and infectious diseases, to further boost the development of TCM in modern medicine.
Collapse
Affiliation(s)
- Xuejun Jiang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Mei Lin
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jianwen Huang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Mulan Mo
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Houhe Liu
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuan Jiang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaowen Cai
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wingnang Leung
- Asia-Pacific Institute of Aging Studies, Lingnan University, Hong Kong, China
| | - Chuanshan Xu
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
29
|
Traditional application and modern pharmacological research of Artemisia annua L. Pharmacol Ther 2020; 216:107650. [DOI: 10.1016/j.pharmthera.2020.107650] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/30/2022]
|
30
|
Gao Y, Chen DL, Zhou M, Zheng ZS, He MF, Huang S, Liao XZ, Zhang JX. Cordycepin enhances the chemosensitivity of esophageal cancer cells to cisplatin by inducing the activation of AMPK and suppressing the AKT signaling pathway. Cell Death Dis 2020; 11:866. [PMID: 33067427 PMCID: PMC7567864 DOI: 10.1038/s41419-020-03079-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 01/22/2023]
Abstract
Although cisplatin (cDDP), is a first-line chemotherapy drug for esophageal cancer, it still has the potential to develop drug resistance and side effects. There is increasing evidence that cordycepin can work synergistically with other chemotherapy drugs. Therefore, we investigated whether combination therapy of cordycepin and cDDP may enhance the therapeutic effect of cDDP. We performed a series of functional tests to study the effect of medical treatment on esophageal cancer cells. We then used GO analysis to examine the pathways affected by treatment with cordycepin and cDDP. Next, we observed changes in the abundance of the selected pathway proteins. The in vivo animal model supported the results of the in vitro experiments. Co-treatment with cordycepin and cDDP inhibited cell growth, migration, and metastasis, as well as induced apoptosis. Cordycepin was found to effectively enhance activation of AMPK and inhibited activity of AKT. In all treatment groups, the expression levels of p-PI3K, p-Akt, p-p70S6K, Caspase-3, and Bcl-2 were significantly reduced, while the expression levels of p-AMPK, cleaved Caspase-3, and Bax increased, and the total levels of Akt, PI3K, and p70S6K levels remained unchanged. Overall, cordycepin was found to enhance the chemical sensitivity of esophageal cancer cells to cisplatin by inducing AMPK activation and inhibiting the AKT signaling pathway. Combination therapy of cordycepin and cisplatin represent a novel potential treatment of esophageal cancer.
Collapse
Affiliation(s)
- Ying Gao
- Department of Oncology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Dan-Lei Chen
- Department of Oncology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Mi Zhou
- Department of Oncology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhou-San Zheng
- Department of Oncology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Mei-Fang He
- Department of Oncology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Sheng Huang
- Department of Orthopaedics, the First Affiliated Hospital of Nanchang University, 330006, Nanchang, China.
| | - Xiao-Zhong Liao
- Department of Oncology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China. .,Department of Oncology, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Jia-Xing Zhang
- Department of Oncology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
31
|
Jiang T, Sun X, Wei L, Li M. Determination of hydrogen peroxide released from cancer cells by a Fe-Organic framework/horseradish peroxidase-modified electrode. Anal Chim Acta 2020; 1135:132-141. [PMID: 33070850 DOI: 10.1016/j.aca.2020.09.040] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/16/2020] [Accepted: 09/23/2020] [Indexed: 01/05/2023]
Abstract
Multi-walled carbon nanotubes (MWCNTs) were used as conductive carrier on the glassy carbon electrode (GCE), and the hybrid of metal organic framework [NH2-MIL-53(Fe)] and horseradish peroxidase (HRP) was prepared by simple physical mechanical mixture. The GCE modified by the above material with immobilization, namely NH2-MIL-53(Fe)/HRP/MWCNTs/GCE, was used to construct an electrochemical biosensor toward H2O2. The results indicated that the addition of NH2-MIL-53(Fe) had a good synergistic effect on the electron transfer of HRP and the detection of H2O2. Under the optimized condition, the biosensor exhibited excellent electrochemical performances such as low detection limit, high sensitivity, good stability and so on. The H2O2 biosensor showed two linear ranges of 0.1-1 μM and 1-600 μM with a calculated detection limit of 0.028 μM (signal-to-noise ratio, S/N = 3). In addition, the stability of the hybrid of NH2-MIL-53(Fe) and HRP were discussed by SEM, XRD and UV-vis methods. Furthermore, the reported biosensors were practically used in direct detection of H2O2 released from HeLa and HepG2 cells successfully. Thus, this work provides a new strategy to fabricate electrochemical biosensors using MOFs and biomolecules.
Collapse
Affiliation(s)
- Tian Jiang
- Anhui Key Laboratory of Chemo-Biosensing, College of Chemistry and Materials Science, Anhui Normal University, Wuhu, 241000, PR China
| | - Xiuxiu Sun
- Anhui Key Laboratory of Chemo-Biosensing, College of Chemistry and Materials Science, Anhui Normal University, Wuhu, 241000, PR China
| | - Lingli Wei
- Anhui Key Laboratory of Chemo-Biosensing, College of Chemistry and Materials Science, Anhui Normal University, Wuhu, 241000, PR China
| | - Maoguo Li
- Anhui Key Laboratory of Chemo-Biosensing, College of Chemistry and Materials Science, Anhui Normal University, Wuhu, 241000, PR China.
| |
Collapse
|
32
|
Zhou X, Zijlstra SN, Soto-Gamez A, Setroikromo R, Quax WJ. Artemisinin Derivatives Stimulate DR5-Specific TRAIL-Induced Apoptosis by Regulating Wildtype P53. Cancers (Basel) 2020; 12:E2514. [PMID: 32899699 PMCID: PMC7563660 DOI: 10.3390/cancers12092514] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/28/2020] [Accepted: 09/01/2020] [Indexed: 12/26/2022] Open
Abstract
Artemisinin derivatives, widely known as commercial anti-malaria drugs, may also have huge potential in treating cancer cells. It has been reported that artemisinin derivatives can overcome resistance to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in liver and cervical cancer cells. In our study, we demonstrated that artesunate (ATS) and dihydroartemisinin (DHA) are more efficient in killing colon cancer cells compared to artemisinin (ART). ATS/DHA induces the expression of DR5 in a P53 dependent manner in HCT116 and DLD-1 cells. Both ATS and DHA overcome the resistance to DHER-induced apoptosis in HCT116, mainly through upregulating death receptor 5 (DR5). We also demonstrate that DHA sensitizes HCT116 cells to DHER-induced apoptosis via P53 regulated DR5 expression in P53 knockdown assays. Nevertheless, a lower effect was observed in DLD-1 cells, which has a single Ser241Phe mutation in the P53 DNA binding domain. Thus, the status of P53 could be one of the determinants of TRAIL resistance in some cancer cells. Finally, the combination treatment of DHA and the TRAIL variant DHER increases cell death in 3D colon cancer spheroid models, which shows its potential as a novel therapy.
Collapse
Affiliation(s)
| | | | | | | | - Wim J. Quax
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (X.Z.); (S.N.Z.); (A.S.-G.); (R.S.)
| |
Collapse
|
33
|
Ginosyan S, Grabski H, Tiratsuyan S. In vitro and in silico Determination of the Interaction of Artemisinin with Human Serum Albumin. Mol Biol 2020. [DOI: 10.1134/s0026893320040056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
34
|
Wang T, Wang J, Ren W, Liu ZL, Cheng YF, Zhang XM. Combination treatment with artemisinin and oxaliplatin inhibits tumorigenesis in esophageal cancer EC109 cell through Wnt/β-catenin signaling pathway. Thorac Cancer 2020; 11:2316-2324. [PMID: 32657048 PMCID: PMC7396387 DOI: 10.1111/1759-7714.13570] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 01/20/2023] Open
Abstract
Background Esophageal cancer (EC) is a prevalent malignant cancer worldwide. Interestingly, the antimalaria compound artemisinin (ART) is also reported to have anticancer potential, although its underlying mechanism in EC is unclear. In this study, we explored the anticancer role of ART in EC109 and further explored the combination of ART and oxaliplatin (OXA) for their synergetic anticancer functions. Methods Human EC cell line EC109 was used. After ART or oxaliplatin (OXA) treatment, cell proliferation, migration, and invasion were measured by MTT, transwell, and scratch wound assays, respectively. Flow cytometry was performed to examine the cell cycle and apoptosis. The mRNA and protein levels were determined using qRT‐PCR and western blotting. Results The migration and invasion abilities of EC109 were suppressed by ART. This was due to the inhibitory effect of ART on the Wnt/β‐catenin signaling pathway. The levels of β‐catenin, c‐myc, and survivin were also downregulated by ART. ART inhibits the proliferation of EC109 cells by arresting the cells in the G1‐phase of cell cycle. By using LiCl, an activator of the Wnt/β‐catenin pathway, we further verified that the inhibition of the Wnt/β‐catenin pathway was indeed due to ART. Remarkably, ART enhanced the anticancer effects of OXA in EC109 cells. OXA combined with ART was found to be more efficient in decreasing tumor growth compared to the individual drugs. Conclusions ART could suppress tumor progression by inhibiting Wnt/β‐catenin signaling pathway, and it may also enhance the antitumor effect of OXA in EC. Thus, ART could be a novel anticancer drug for EC treatment. Key points Significant findings of the study ART could be a novel anticancer drug for esophageal cancer (EC) treatment. What this study adds Combination treatment with artemisinin and oxaliplatin inhibits tumorigenesis in esophageal cancer EC109 cells through the Wnt/β‐catenin signaling pathway.
Collapse
Affiliation(s)
- Tao Wang
- Department of Radiotherapy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jian Wang
- Department of Ultrasound, Shandong Province Coal Taishan Sanatorium, Taian, China
| | - Wei Ren
- Department of Radiotherapy, The People's Hospital of Lanling County, Linyi, China
| | - Zhu-Long Liu
- Department of Radiotherapy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yu-Feng Cheng
- Department of Radiotherapy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiao-Mei Zhang
- Department of Radiotherapy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
35
|
García Manzano MF, Joray MB, Laiolo J, Palacios SM, Carpinella MC. Cytotoxic Activity of Germacrane-Type Sesquiterpene Lactones from Dimerostemma aspilioides. JOURNAL OF NATURAL PRODUCTS 2020; 83:1909-1918. [PMID: 32496057 DOI: 10.1021/acs.jnatprod.0c00115] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The need for effective candidates as cytotoxic drugs that at the same time challenge cancer multidrug resistance encouraged a search for these in plants of central Argentina. Bioassay-guided fractionation of the cytotoxic extract from Dimerostemma aspilioides led to the isolation of the germacranolide tomenphantin A (1), along with three new analogues (2-4). These efficiently inhibited the proliferation of the leukemia cell lines K562 and CCRF-CEM and their resistant variants, Lucena 1 and CEM/ADR5000, respectively, with IC50 values ranging from 0.40 to 7.7 μM. The structures and relative configurations of compounds 1-4 were elucidated by analysis of the spectroscopic data, in particular NMR spectroscopy. The most active among these was compound 1 (IC50 = 0.40-5.1 μM), and, therefore, this was selected as a model for a mechanistic study, which revealed that its antiproliferative effect was mediated by cell cycle arrest in the G2/M phase followed by apoptosis. The activity of compound 1 was selective, given the absence of cytotoxicity toward peripheral blood mononuclear cells. The results show the potential of these compounds, and in particular of compound 1, as leads for the development of drug candidates to fight sensitive and resistant leukemia cells.
Collapse
Affiliation(s)
- María F García Manzano
- Fine Chemical and Natural Products Laboratory, Research Institute of Natural Resources and Sustainability José Sánchez Labrador S.J. (IRNASUS-CONICET), School of Chemistry, Catholic University of Córdoba, Córdoba X5016DHK, Argentina
| | - Mariana B Joray
- Fine Chemical and Natural Products Laboratory, Research Institute of Natural Resources and Sustainability José Sánchez Labrador S.J. (IRNASUS-CONICET), School of Chemistry, Catholic University of Córdoba, Córdoba X5016DHK, Argentina
| | - Jerónimo Laiolo
- Fine Chemical and Natural Products Laboratory, Research Institute of Natural Resources and Sustainability José Sánchez Labrador S.J. (IRNASUS-CONICET), School of Chemistry, Catholic University of Córdoba, Córdoba X5016DHK, Argentina
| | - Sara M Palacios
- Fine Chemical and Natural Products Laboratory, Research Institute of Natural Resources and Sustainability José Sánchez Labrador S.J. (IRNASUS-CONICET), School of Chemistry, Catholic University of Córdoba, Córdoba X5016DHK, Argentina
| | - María C Carpinella
- Fine Chemical and Natural Products Laboratory, Research Institute of Natural Resources and Sustainability José Sánchez Labrador S.J. (IRNASUS-CONICET), School of Chemistry, Catholic University of Córdoba, Córdoba X5016DHK, Argentina
| |
Collapse
|
36
|
Zhang XW, Zhao X, Liu KH, Sub HM. Kinetics study on reaction between dihydroartemisinic acid and singlet oxygen: An essential step to photochemical synthesis of artemisinin. CHINESE J CHEM PHYS 2020. [DOI: 10.1063/1674-0068/cjcp2002021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Xian-wang Zhang
- Beijing National Laboratory for Molecular Science, Institute of Chemistry, Chinese Academy of Science, Beijing 100190, China
- College of Chemistry, Beijing Normal University, Beijing 100875, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuan Zhao
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Kun-hui Liu
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Hong-mei Sub
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
37
|
Medicinal Plants Used in Traditional Management of Cancer in Uganda: A Review of Ethnobotanical Surveys, Phytochemistry, and Anticancer Studies. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3529081. [PMID: 32256639 PMCID: PMC7102457 DOI: 10.1155/2020/3529081] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 02/18/2020] [Indexed: 01/29/2023]
Abstract
The burden of neoplastic diseases is a significant global health challenge accounting for thousands of deaths. In Uganda, about 32,617 cancer cases were reported in 2018, accompanied by 21,829 deaths. In a view to identify some potential anticancer plant candidates for possible drug development, the current study was designed to compile the inventory of plants with reported anticancer activity used in rural Uganda and the evidences supporting their use in cancer therapy. An electronic survey in multidisciplinary databases revealed that 29 plant species belonging to 28 genera distributed among 24 families have been reported to be used in the management of cancer in Uganda. Anticancer plants were majorly from the families Bignoniaceae (7%), Caricaceae (7%), Fabaceae (7%), Moraceae (7%), and Rutaceae (7%). Most species occur in the wild (52%), though some are cultivated (48%). The growth habit of the plants is as trees (55%) or herbs (45%). Anticancer extracts are usually prepared from leaves (29%), bark (24%), roots (21%), and fruits (13%) through decoctions (53%), as food spices (23%) or pounded to produce ointments that are applied topically (10%). Prunus africana (Hook.f.) Kalkman, Opuntia species, Albizia coriaria (Welw. ex Oliver), Daucus carota L., Cyperus alatus (Nees) F. Muell., Markhamia lutea (Benth.) K. Schum., and Oxalis corniculata L. were the most frequently encountered species. As per global reports, Allium sativum L., Annona muricata L., Carica papaya L., Moringa oleifera Lam., Opuntia species, Prunus africana (Hook.f.) Kalkman, and Catharanthus roseus (L.) G. Don. are the most studied species, with the latter having vincristine and vinblastine anticancer drugs developed from it. Prostate, cervical, breast, and skin cancers are the top traditionally treated malignancies. There is a need to isolate and evaluate the anticancer potential of the bioactive compounds in the unstudied claimed plants, such as Cyperus alatus (Nees) F. Muell., Ficus dawei Hutch., Ficus natalensis Hochst., and Lovoa trichilioides Harms, and elucidate their mechanism of anticancer activity.
Collapse
|
38
|
Hou C, Guo D, Yu X, Wang S, Liu T. TMT-based proteomics analysis of the anti-hepatocellular carcinoma effect of combined dihydroartemisinin and sorafenib. Biomed Pharmacother 2020; 126:109862. [PMID: 32120157 DOI: 10.1016/j.biopha.2020.109862] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC), as the major primary liver cancer, is one of the most prevalent malignant diseases with a high mortality rate worldwide. Prior studies have demonstrated that dihydroartemisinin (DHA), the semisynthetic derivative of artemisinin, possesses anti-HCC activity. The multikinase inhibitor sorafenib has been approved for the treatment of HCC. However, the anti-HCC efficacy of DHA combined with sorafenib has not been reported. In this study, we confirmed the significantly enhanced anti-HCC efficacy of DHA in combination with sorafenib compared with that of each agent alone. Tandem Mass Tag (TMT) peptide labeling coupled with LC-MS/MS was used to quantify the proteins from the control, DHA, sorafenib, and DHA + sorafenib groups. In total, 532, 426, 628 differentially expressed proteins (fold change >1.20 or <0.83 and P-value <0.05) were determined by comparing DHA versus control, sorafenib versus control and DHA + sorafenib versus control groups, respectively. Moreover, optimized screening was performed, and 101 optimized differentially expressed proteins were identified. The results of functional analysis of the optimized differentially expressed proteins suggested that they were enriched in cell components such as membrane-bound vesicles, extracellular vesicles, and organelle lumens, and they were mainly involved in biological processes such as cellular component organization, response to stress, and response to chemicals; in addition, they were related to various molecular functions such as protein binding, chromatin binding and enzyme binding. KEGG pathway analysis showed that the optimized differentially expressed proteins were enriched in pyrimidine metabolism, RNA polymerase, base excision repair, and osteoclast differentiation. Protein-protein interaction (PPI) networks of some of the optimized upregulated proteins suggested that they might not only affect vitamin and fat digestion and absorption but may also be involved in tight junctions. In the PPI network, some of the optimized downregulated proteins were enriched in base excision repair, RNA polymerase, purine metabolism, pyrimidine metabolism and mucin type O-glycan biosynthesis. Overall, this research explored the anti-HCC efficacy of DHA combined with sorafenib by using the TMT-based quantitative proteomics technique and might facilitate the understanding of the related anti-HCC molecular mechanism.
Collapse
Affiliation(s)
- Chunying Hou
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Dongqing Guo
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xue Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Shuyan Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tianhua Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
39
|
Li Y, Zhou X, Liu J, Gao N, Yang R, Wang Q, Ji J, Ma L, He Q. Dihydroartemisinin inhibits the tumorigenesis and metastasis of breast cancer via downregulating CIZ1 expression associated with TGF-β1 signaling. Life Sci 2020; 248:117454. [PMID: 32088211 DOI: 10.1016/j.lfs.2020.117454] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/05/2020] [Accepted: 02/19/2020] [Indexed: 12/25/2022]
Abstract
AIMS Dihydroartemisinin (DHA) is currently considered as the promising cancer therapeutic drug. In this study, we aimed to investigate the anti-proliferative and anti-metastasis effects of DHA. MAIN METHODS Utilizing breast cancer cells MCF-7, MDA-MB-231 and BT549, cell proliferation, migration and invasion were detected. RT-qPCR was performed to detect CIZ1, TGF-β1 and Snail expression, and the interactions of these related molecules were analyzed by GeneMANIA database. Western blot detected CIZ1, TGF-β1/Smads signaling and Snail expression in DHA-treated cells, in TGFβ1-induced cells with enhanced metastatic capacity, and in cells treated with DHA plus TGFβ1/TGFβ1 inhibitor SD-208. KEY FINDINGS Results indicated DHA inhibited breast cancer cell proliferation and migration, with more potent effects compared with that of artemisinin. RT-qPCR and Western blot showed DHA inhibited CIZ1, TGF-β1 and Snail expression, and these molecules were shown to have protein-protein interactions by bioinformatics. Furthermore, TGFβ1-treatment enhanced MCF-7 migration and invasion, and CIZ1, TGF-β1/Smads signaling and snail activities; DHA, SD-208, combination of DHA and SD-208 reversed these conditions, preliminarily proving the cascade regulation between TGF-β1 signaling and CIZ1. MCF-7 xenografts model demonstrated the inhibition of DHA on tumor burden, and its mechanisms and well-tolerance in vivo; combination of DHA and SD-208 tried by us for the first time showed better treatment effects, but possible liver impairment made its use still keep cautious. SIGNIFICANCE DHA treatment inhibits the proliferation and metastasis of breast cancer, through suppressing TGF-β1/Smad signaling and CIZ1, suggesting the promising potential of DHA as a well-tolerated antitumor TGF-β1 pathway inhibitor.
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
| | - Ning Gao
- Department of Clinical Laboratories, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Ruihua Yang
- Department of Clinical Laboratories, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Qi Wang
- Department of Clinical Laboratories, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Jing Ji
- Department of Clinical Laboratories, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Ling Ma
- 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
|
40
|
Akbarian A, Ebtekar M, Pakravan N, Hassan ZM. Folate receptor alpha targeted delivery of artemether to breast cancer cells with folate-decorated human serum albumin nanoparticles. Int J Biol Macromol 2020; 152:90-101. [PMID: 32057865 DOI: 10.1016/j.ijbiomac.2020.02.106] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 02/05/2020] [Accepted: 02/10/2020] [Indexed: 12/24/2022]
Abstract
The pharmaceutical application of artemether (ARM) as an anticancer natural agent is hampered due to its poor solubility and bioavailability. In the present study, ARM was encapsulated in human serum albumin nanoparticles (HSA NPs) via desolvation method led to improvement of the water solubility by 50 folds. In further, folate-decorated ARM-HSA NPs (F-ARM-HSA NPs) were developed to enhance targeted delivery to folate receptor alpha (FRα)-overexpressing breast cancer cells. The hydrodynamic diameter and the zeta potential value of F-ARM-HSA NPs were 198 ± 11.22 nm and -23 ± 0.88 mV, respectively. Fluorescent microscopy demonstrated an enhanced cellular uptake of F-ARM-HSA NPs by high FRα-expressing MDA-MB-231 breast cancer cells compared to low FRα-expressing SK-BR-3 breast cancer cells. Cytotoxicity assay revealed a small significant difference between cytotoxicity effect of targeted and non-targeted NPs in SK-BR-3 cells. However, in MDA-MB-231 cells due to FRα-mediated endocytosis, the F-conjugated NPs had less inhibitory concentration (IC50) value (19.82 μg/mL) and higher cytotoxicity after 72 h compared to non-targeted ARM-HSA NPs. Flow cytometry analysis indicated a more potent drug-induced apoptosis rather than necrosis. The results suggest that our novel F-ARM-HSA NPs are likely to be recommended as a promising candidate for combination therapy of FRα-overexpressing breast cancer cells.
Collapse
Affiliation(s)
- Asiye Akbarian
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, PO Box: 14155-114, Iran
| | - Masoumeh Ebtekar
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, PO Box: 14155-114, Iran.
| | - Nafiseh Pakravan
- Department of Immunology, Medical School, Alborz University of Medical Sciences, Karaj, PO Box 31497-79453, Iran
| | - Zuhair Mohammad Hassan
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, PO Box: 14155-114, Iran
| |
Collapse
|
41
|
Palve V, Liao Y, Remsing Rix LL, Rix U. Turning liabilities into opportunities: Off-target based drug repurposing in cancer. Semin Cancer Biol 2020; 68:209-229. [PMID: 32044472 DOI: 10.1016/j.semcancer.2020.02.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/29/2020] [Accepted: 02/03/2020] [Indexed: 12/12/2022]
Abstract
Targeted drugs and precision medicine have transformed the landscape of cancer therapy and significantly improved patient outcomes in many cases. However, as therapies are becoming more and more tailored to smaller patient populations and acquired resistance is limiting the duration of clinical responses, there is an ever increasing demand for new drugs, which is not easily met considering steadily rising drug attrition rates and development costs. Considering these challenges drug repurposing is an attractive complementary approach to traditional drug discovery that can satisfy some of these needs. This is facilitated by the fact that most targeted drugs, despite their implicit connotation, are not singularly specific, but rather display a wide spectrum of target selectivity. Importantly, some of the unintended drug "off-targets" are known anticancer targets in their own right. Others are becoming recognized as such in the process of elucidating off-target mechanisms that in fact are responsible for a drug's anticancer activity, thereby revealing potentially new cancer vulnerabilities. Harnessing such beneficial off-target effects can therefore lead to novel and promising precision medicine approaches. Here, we will discuss experimental and computational methods that are employed to specifically develop single target and network-based off-target repurposing strategies, for instance with drug combinations or polypharmacology drugs. By illustrating concrete examples that have led to clinical translation we will furthermore examine the various scientific and non-scientific factors that cumulatively determine the success of these efforts and thus can inform the future development of new and potentially lifesaving off-target based drug repurposing strategies for cancers that constitute important unmet medical needs.
Collapse
Affiliation(s)
- Vinayak Palve
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Yi Liao
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Lily L Remsing Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Uwe Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
42
|
Laleve A, Panozzo C, Kühl I, Bourand-Plantefol A, Ostojic J, Sissoko A, Tribouillard-Tanvier D, Cornu D, Burg A, Meunier B, Blondel M, Clain J, Bonnefoy N, Duval R, Dujardin G. Artemisinin and its derivatives target mitochondrial c-type cytochromes in yeast and human cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118661. [PMID: 31987792 DOI: 10.1016/j.bbamcr.2020.118661] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/17/2020] [Accepted: 01/23/2020] [Indexed: 12/27/2022]
Abstract
Artemisinin and its derivatives kill malaria parasites and inhibit the proliferation of cancer cells. In both processes, heme was shown to play a key role in artemisinin bioactivation. We found that artemisinin and clinical artemisinin derivatives are able to compensate for a mutation in the yeast Bcs1 protein, a key chaperon involved in biogenesis of the mitochondrial respiratory complex III. The equivalent Bcs1 variant causes an encephalopathy in human by affecting complex III assembly. We show that artemisinin derivatives decrease the content of mitochondrial cytochromes and disturb the maturation of the complex III cytochrome c1. This last effect is likely responsible for the compensation by decreasing the detrimental over-accumulation of the inactive pre-complex III observed in the bcs1 mutant. We further show that a fluorescent dihydroartemisinin probe rapidly accumulates in the mitochondrial network and targets cytochromes c and c1 in yeast, human cells and isolated mitochondria. In vitro this probe interacts with purified cytochrome c only under reducing conditions and we detect cytochrome c-dihydroartemisinin covalent adducts by mass spectrometry analyses. We propose that reduced mitochondrial c-type cytochromes act as both targets and mediators of artemisinin bioactivation in yeast and human cells.
Collapse
Affiliation(s)
- Anais Laleve
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France.
| | - Cristina Panozzo
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Inge Kühl
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Alexa Bourand-Plantefol
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Jelena Ostojic
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Abdoulaye Sissoko
- Université de Paris, MERIT, IRD, 4 Avenue de l'Observatoire, 75006 Paris, France
| | - Déborah Tribouillard-Tanvier
- Inserm UMR1078, Université de Bretagne Occidentale, Faculté de Médecine et des Sciences de la Santé; Etablissement Français du Sang (EFS) Bretagne; CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, 22 avenue Camille Desmoulins, 29200 Brest, France
| | - David Cornu
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Angélique Burg
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Brigitte Meunier
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Marc Blondel
- Inserm UMR1078, Université de Bretagne Occidentale, Faculté de Médecine et des Sciences de la Santé; Etablissement Français du Sang (EFS) Bretagne; CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, 22 avenue Camille Desmoulins, 29200 Brest, France
| | - Jerome Clain
- Université de Paris, MERIT, IRD, 4 Avenue de l'Observatoire, 75006 Paris, France
| | - Nathalie Bonnefoy
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| | - Romain Duval
- Université de Paris, MERIT, IRD, 4 Avenue de l'Observatoire, 75006 Paris, France
| | - Geneviève Dujardin
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France.
| |
Collapse
|
43
|
Zhang B. Artemisinin‐derived dimers as potential anticancer agents: Current developments, action mechanisms, and structure–activity relationships. Arch Pharm (Weinheim) 2019; 353:e1900240. [DOI: 10.1002/ardp.201900240] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/26/2019] [Accepted: 11/15/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Bo Zhang
- School of Chemistry and Life ScienceAnshan Normal University Anshan Liaoning China
| |
Collapse
|
44
|
Chen CP, Chen K, Feng Z, Wen X, Sun H. Synergistic antitumor activity of artesunate and HDAC inhibitors through elevating heme synthesis via synergistic upregulation of ALAS1 expression. Acta Pharm Sin B 2019; 9:937-951. [PMID: 31649844 PMCID: PMC6804493 DOI: 10.1016/j.apsb.2019.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 12/31/2022] Open
Abstract
Artemisinin and its derivatives (ARTs) were reported to display heme-dependent antitumor activity. On the other hand, histone deacetylase inhibitors (HDACi) were known to be able to promote heme synthesis in erythroid cells. Nevertheless, the effect of HDACi on heme homeostasis in non-erythrocytes remains unknown. We envisioned that the combination of HDACi and artesunate (ARS) might have synergistic antitumor activity through modulating heme synthesis. In vitro studies revealed that combination of ARS and HDACi exerted synergistic tumor inhibition by inducing cell death. Moreover, this combination exhibited more effective antitumor activity than either ARS or HDACi monotherapy in xenograft models without apparent toxicity. Importantly, mechanistic studies revealed that HDACi coordinated with ARS to increase 5-aminolevulinate synthase (ALAS1) expression, and subsequent heme production, leading to enhanced cytotoxicity of ARS. Notably, knocking down ALAS1 significantly blunted the synergistic effect of ARS and HDACi on tumor inhibition, indicating a critical role of ALAS1 upregulation in mediating ARS cytotoxicity. Collectively, our study revealed the mechanism of synergistic antitumor action of ARS and HDACi. This finding indicates that modulation of heme synthesis pathway by the combination based on ARTs and other heme synthesis modulators represents a promising therapeutic approach to solid tumors.
Collapse
Key Words
- ALA, 5-aminolevulinic acid
- ALAD, 5-aminolevulinate dehydratase
- ALAS, 5-aminolevulinate synthase
- ALAS1
- ARS, artesunate
- ART, artemisinin
- Antitumor
- Artesunate
- CCK-8, cell counting kit 8
- CI, combination index
- CMCNa, carboxymethyl cellulose
- DHA, dihydroartemisinin
- DMAB, (dimethylamino)benzaldehyde
- FECH, ferrochelatase
- GSDME, gasdermin E
- HDAC inhibitor
- HDAC, histone deacetylase
- HDACi, HDAC inhibitor
- HMBS, hydroxymethylbilane synthase
- Heme
- KD, knockdown
- KO, knockout
- LBH589, panobinostat
- PDT, photodynamic therapy
- PI, propidium iodide
- PpIX, protoporphyrin IX
- ROS, reactive oxygen species
- SA, succinyl acetone
- SAHA, vorinostat
- WT, wild-type
- sgRNA, single guide RNA
Collapse
Affiliation(s)
| | | | | | | | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
45
|
Synthesis of novel S-linked dihydroartemisinin derivatives and evaluation of their anticancer activity. Eur J Med Chem 2019; 178:552-570. [DOI: 10.1016/j.ejmech.2019.06.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/24/2019] [Accepted: 06/05/2019] [Indexed: 11/19/2022]
|
46
|
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
|
47
|
Liu X, Cao J, Huang G, Zhao Q, Shen J. Biological Activities of Artemisinin Derivatives Beyond Malaria. Curr Top Med Chem 2019; 19:205-222. [DOI: 10.2174/1568026619666190122144217] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 12/26/2022]
Abstract
Artemisinin is isolated from Artemisia annua L. with peroxide-containing sesquiterpene lactone structure. Because of its unique structural characteristics and promising anticancer, antivirus activities, it has recently received increasing attention. The aim of this review is to summarize recent discoveries of artemisinin's novel derivatives with new pharmaceutical effects beyond malaria with a focus on its antitumor and antivirus activity, as well as potential results of combination therapy with other clinical drugs.
Collapse
Affiliation(s)
- Xiaoyan Liu
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jianguo Cao
- College of Life and Environmental Sciences, Shanghai Normal University, Shanghai, 201418, China
| | - Guozheng Huang
- College of Life and Environmental Sciences, Shanghai Normal University, Shanghai, 201418, China
| | - Qingjie Zhao
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jingshan Shen
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
48
|
Shi X, Wang L, Ren L, Li J, Li S, Cui Q, Li S. Dihydroartemisinin, an antimalarial drug, induces absent in melanoma 2 inflammasome activation and autophagy in human hepatocellular carcinoma HepG2215 cells. Phytother Res 2019; 33:1413-1425. [PMID: 30873702 DOI: 10.1002/ptr.6332] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 02/03/2019] [Accepted: 02/08/2019] [Indexed: 12/22/2022]
Abstract
As an effective antimalarial drug, Dihydroartemisinin (DHA) is readily isolated from the traditional Chinese medicine of Artemisia annua. DHA is not only an autophagy promoter but also a substance with strong antitumor efficiency. The relationship between autophagy and inflammasomes has been suggested in hepatocellular carcinoma (HCC). However, there are few reports describing relationships between inflammasomes and autophagy in HCC therapy. The present study demonstrated that DHA suppressed cell proliferation in HepG2215 cells in a dose- and time-dependent manner. The inhibitory activity is mediated by autophagy, in which reactive oxygen species (ROS) production induced nuclear and mitochondrial DNA damage. Then, DHA were first shown to promote AIM2/caspase-1 inflammasome. Compared with the DHA group, the autophagy inhibitor 3-MA significantly inhibited the expressions of activated Caspase-1, a pyroptotic marker proteins. Meanwhile, repression of mTOR by rapamycin promoted autophagy and AIM2/caspase-1 activation. The caspase-1 inhibitor Z-YVAD-FMK also notably blocked autophagy cell death characterized by the downexpression of Beclin-1 and LC3-II. Additionally, the study demonstrated that DHA suppressed pseudopodium formation and cell mobility. Therefore, we first reveal a novel mechanism that DHA promotes AIM2/caspase-1 inflammasome, which contributes to autophagy in HepG2215 cells. Moreover, nuclear and mitochondrial DNA damage was also involved in this process via ROS production.
Collapse
Affiliation(s)
- Xinli Shi
- Hebei Provincial Engineering Laboratory of Plant Bioreactor Preparation Technology, Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Li Wang
- Laboratory of Organ Fibrosis Prophylaxis and Treatment by Combine Traditional Chinese and Western Medicine, Research Center of Combine Traditional Chinese and Western Medicine, Clinical Laboratory, Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Laifeng Ren
- Department of Immunology, Affiliated Cancer Hospital of Shanxi Medical University and Shanxi Cancer Hospital, Taiyuan, China
| | - Jianchun Li
- Laboratory of Organ Fibrosis Prophylaxis and Treatment by Combine Traditional Chinese and Western Medicine, Research Center of Combine Traditional Chinese and Western Medicine, Clinical Laboratory, Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Shenghao Li
- Hebei Provincial Engineering Laboratory of Plant Bioreactor Preparation Technology, Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Qingzhuo Cui
- Hebei Provincial Engineering Laboratory of Plant Bioreactor Preparation Technology, Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Sheng Li
- Hebei Provincial Engineering Laboratory of Plant Bioreactor Preparation Technology, Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
49
|
Wang L, Li J, Shi X, Li S, Tang PMK, Li Z, Li H, Wei C. Antimalarial Dihydroartemisinin triggers autophagy within HeLa cells of human cervical cancer through Bcl-2 phosphorylation at Ser70. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 52:147-156. [PMID: 30599894 DOI: 10.1016/j.phymed.2018.09.221] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/25/2018] [Accepted: 09/25/2018] [Indexed: 05/21/2023]
Abstract
BACKGROUND As an effective antimalarial medicine, Dihydroartemisinin (DHA) has therapeutic potential on human cervical cancer. However, its working mechanism has not been elucidated. PURPOSE This study aimed to investigate the reversal effect of DHA on human cervical cancer HeLa cells, and explored its mechanism of action in vitro and in vivo. STUDY DESIGN/METHODS The effect and mechanism of DHA on HeLa cells was examined by using CCK-8 assay, flow cytometry, transmission electron microscopy, immunofluorescence, and Western blot analysis in human hepatocellular carcinoma cells. RESULTS In this study, it was confirmed that DHA had statistically equivalent anti-tumor efficiency in HeLa cells with a clinical chemotherapeutic agent of cisplatin. Meanwhile, DHA triggered autophagy, where LC3B-II expression was dose-dependently increased. Further, it was revealed that DHA promotes reactive oxygen species (ROS) generation, with DNA double-strand breaks (DSB) damage, as up-regulation of γH2AX protein and foci formation. Interestingly, we firstly demonstrated that DHA induced autophagy through promotion of the phosphorylation of Bcl-2 (Ser70), independent of the phosphorylated JNK1/2 (Thr183/Tyr185). Moreover, DHA-treated HeLa cells displayed an increase in the pro-autophagic protein Beclin-1 with downregulated the phospho-mTOR (Ser2448). Furthermore, upregulated pro-apoptotic protein Bak-1, but not Bax, suggesting Bak-1 is included in DHA-induced autophagy. CONCLUSION Therefore, DHA upregulates the phosphorylation of Bcl-2 (Ser70) and mTOR (Ser2448) and induces autophagic cell death in Hela cells. This study provided a mechanism to support DHA, an autophagy inducer, as a potential therapeutic agent for human cervical cancer.
Collapse
Affiliation(s)
- Li Wang
- Laboratory of Organ Fibrosis Prophylaxis and Treatment by Combine Traditional Chinese and Western Medicine, Research Center of Combine Traditional Chinese and Western Medicine, Clinical Laboratory, Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Jianchun Li
- Laboratory of Organ Fibrosis Prophylaxis and Treatment by Combine Traditional Chinese and Western Medicine, Research Center of Combine Traditional Chinese and Western Medicine, Clinical Laboratory, Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Xinli Shi
- Hebei Provincial Engineering Laboratory of Plant Bioreactor Preparation Technology, Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China; Department of Otolaryngology Head and Neck Surgery, Bethune International Peace Hospital, Shijiazhuang 050081, China.
| | - Shenghao Li
- Hebei Provincial Engineering Laboratory of Plant Bioreactor Preparation Technology, Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Zhen Li
- Department of Otolaryngology Head and Neck Surgery, Bethune International Peace Hospital, Shijiazhuang 050081, China
| | - Hui Li
- Department of Otolaryngology Head and Neck Surgery, Bethune International Peace Hospital, Shijiazhuang 050081, China
| | - Cong Wei
- Laboratory of Organ Fibrosis Prophylaxis and Treatment by Combine Traditional Chinese and Western Medicine, Research Center of Combine Traditional Chinese and Western Medicine, Clinical Laboratory, Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
50
|
Kumar MS, Yadav TT, Khair RR, Peters GJ, Yergeri MC. Combination Therapies of Artemisinin and its Derivatives as a Viable Approach for Future Cancer Treatment. Curr Pharm Des 2019; 25:3323-3338. [PMID: 31475891 DOI: 10.2174/1381612825666190902155957] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 08/30/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Many anticancer drugs have been developed for clinical usage till now, but the major problem is the development of drug-resistance over a period of time in the treatment of cancer. Anticancer drugs produce huge adverse effects, ultimately leading to death of the patient. Researchers have been focusing on the development of novel molecules with higher efficacy and lower toxicity; the anti-malarial drug artemisinin and its derivatives have exhibited cytotoxic effects. METHODS We have done extensive literature search for artemisinin for its new role as anti-cancer agent for future treatment. Last two decades papers were referred for deep understanding to strengthen its role. RESULT Literature shows changes at 9, 10 position in the artemisinin structure produces anticancer activity. Artemisinin shows anticancer activity in leukemia, hepatocellular carcinoma, colorectal and breast cancer cell lines. Artemisinin and its derivatives have been studied as combination therapy with several synthetic compounds, RNA interfaces, recombinant proteins and antibodies etc., for synergizing the effect of these drugs. They produce an anticancer effect by causing cell cycle arrest, regulating signaling in apoptosis, angiogenesis and cytotoxicity activity on the steroid receptors. Many novel formulations of artemisinin are being developed in the form of carbon nanotubes, polymer-coated drug particles, etc., for delivering artemisinin, since it has poor water/ oil solubility and is chemically unstable. CONCLUSION We have summarize the combination therapies of artemisinin and its derivatives with other anticancer drugs and also focussed on recent developments of different drug delivery systems in the last 10 years. Various reports and clinical trials of artemisinin type drugs indicated selective cytotoxicity along with minimal toxicity thus projecting them as promising anti-cancer agents in future cancer therapies.
Collapse
Affiliation(s)
- Maushmi S Kumar
- Department of Pharmaceutical Chemistry, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vile Parle west, Mumbai-400056, India
| | - Tanuja T Yadav
- Department of Pharmaceutical Chemistry, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vile Parle west, Mumbai-400056, India
| | - Rohan R Khair
- Department of Pharmaceutical Chemistry, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vile Parle west, Mumbai-400056, India
| | - Godefridus J Peters
- Department of Medical Oncology, VU University Medical Center, Amsterdam, Netherlands
| | - Mayur C Yergeri
- Department of Pharmaceutical Chemistry, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vile Parle west, Mumbai-400056, India
| |
Collapse
|