1
|
Brusatol: A potential sensitizing agent for cancer therapy from Brucea javanica. Biomed Pharmacother 2023; 158:114134. [PMID: 36525821 DOI: 10.1016/j.biopha.2022.114134] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Cancer is currently the most important problem endangering human health. As antitumor drugs have always been the most common methods for treating cancers, searching for new antitumor agents is of great significance. Brusatol, a quassinoid from the seeds of Brucea javanica, exhibits a potent tumor-suppressing effect with improved disease outcome. Studies have shown that brusatol not only shows potential tumor inhibition through multiple pharmacological effects, such as promoting apoptosis and inhibiting metastasis but also exhibits significant synergistic antitumor effects in combination with chemotherapeutic agents and overcoming chemical resistance in a wide range of cancer types. In this paper, the antitumor effects and mechanisms of brusatol were reviewed to provide evidence that brusatol has the exact antitumor efficacy of chemotherapeutic agents and show the potential of brusatol to be developed as a promising antitumor drug.
Collapse
|
2
|
Zuo Y, Li R, Zhang Y, Bao G, Le Y, Yan L. Design, synthesis and antitumor activity of 5-trifluoromethylpyrimidine derivatives as EGFR inhibitors. J Enzyme Inhib Med Chem 2022; 37:2742-2754. [PMID: 36176072 PMCID: PMC9542405 DOI: 10.1080/14756366.2022.2128797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
A new series of 5-trifluoromethylpyrimidine derivatives were designed and synthesised as EGFR inhibitors. Three tumour cells A549, MCF-7, PC-3 and EGFR kinase were employed to evaluate their biological activities. The results were shown that most of the target compounds existed excellent antitumor activities. In particular, the IC50 values of compound 9u (E)-3-((2-((4-(3-(3-fluorophenyl)acrylamido)phenyl)amino)-5-(trifluoromethyl)pyrimidin-4-yl)amino)-N-methylthiophene-2-carboxamide against A549, MCF-7, PC-3 cells and EGFR kinase reached to 0.35 μM, 3.24 μM, 5.12 μM, and 0.091 μM, respectively. Additionally, further researches revealed that compound 9u could induce early apoptosis of A549 cells and arrest the cells in G2/M phase. Taken together, these findings indicated that compound 9u was potential for developing as antitumor reagent.
Collapse
Affiliation(s)
- Yaqing Zuo
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China.,Guizhou Engineering Laboratory for Synthetic Drugs, Guiyang, China
| | - Rongrong Li
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China.,Guizhou Engineering Laboratory for Synthetic Drugs, Guiyang, China
| | - Yan Zhang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China.,Guizhou Engineering Laboratory for Synthetic Drugs, Guiyang, China
| | - Guochen Bao
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, Australia
| | - Yi Le
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China.,Guizhou Engineering Laboratory for Synthetic Drugs, Guiyang, China
| | - Longjia Yan
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China.,Guizhou Engineering Laboratory for Synthetic Drugs, Guiyang, China
| |
Collapse
|
3
|
Concept of Hybrid Drugs and Recent Advancements in Anticancer Hybrids. Pharmaceuticals (Basel) 2022; 15:ph15091071. [PMID: 36145292 PMCID: PMC9500727 DOI: 10.3390/ph15091071] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is a complex disease, and its treatment is a big challenge, with variable efficacy of conventional anticancer drugs. A two-drug cocktail hybrid approach is a potential strategy in recent drug discovery that involves the combination of two drug pharmacophores into a single molecule. The hybrid molecule acts through distinct modes of action on several targets at a given time with more efficacy and less susceptibility to resistance. Thus, there is a huge scope for using hybrid compounds to tackle the present difficulties in cancer medicine. Recent work has applied this technique to uncover some interesting molecules with substantial anticancer properties. In this study, we report data on numerous promising hybrid anti-proliferative/anti-tumor agents developed over the previous 10 years (2011–2021). It includes quinazoline, indole, carbazole, pyrimidine, quinoline, quinone, imidazole, selenium, platinum, hydroxamic acid, ferrocene, curcumin, triazole, benzimidazole, isatin, pyrrolo benzodiazepine (PBD), chalcone, coumarin, nitrogen mustard, pyrazole, and pyridine-based anticancer hybrids produced via molecular hybridization techniques. Overall, this review offers a clear indication of the potential benefits of merging pharmacophoric subunits from multiple different known chemical prototypes to produce more potent and precise hybrid compounds. This provides valuable knowledge for researchers working on complex diseases such as cancer.
Collapse
|
4
|
Lyu X, Zeng L, Shi J, Ming Z, Li W, Liu B, Chen Y, Yuan B, Sun R, Yuan J, Zhao N, Yang X, Chen G, Yang S. Essential role for STAT3/FOXM1/ATG7 signaling-dependent autophagy in resistance to Icotinib. J Exp Clin Cancer Res 2022; 41:200. [PMID: 35690866 PMCID: PMC9188165 DOI: 10.1186/s13046-022-02390-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/15/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The contribution of autophagy to cancer therapy resistance remains complex, mainly owing to the discrepancy of autophagy mechanisms in different therapy. However, the potential mechanisms of autophagy-mediated resistance to icotinib have yet to be elucidated. METHODS The effect of autophagy in icotinib resistance was examined using a series of in vitro and in vivo assays. The results above were further verified in biopsy specimens of lung cancer patients before and after icotinib or gefitinib treatment. RESULTS Icotinib increased ATG3, ATG5, and ATG7 expression, but without affecting Beclin-1, VPS34 and ATBG14 levels in icotinib-resistant lung cancer cells. Autophagy blockade by 3-MA or silencing Beclin-1 had no effects on resistance to icotinib. CQ effectively restored lung cancer cell sensitivity to icotinib in vitro and in vivo. Notably, aberrantly activated STAT3 and highly expressed FOXM1 were required for autophagy induced by icotinib, without the involvement of AMPK/mTOR pathway in this process. Alterations of STAT3 activity using genetic and/or pharmacological methods effectively affected FOXM1 and ATG7 levels increased by icotinib, with altering autophagy and icotinib-mediated apoptosis in resistant cells. Furthermore, silencing FOXM1 impaired up-regulated ATG7 induced by STAT3-CA and icotinib. STAT3/FOXM1 signalling blockade also reversed resistance to icotinib in vivo. Finally, we found a negative correlation between STAT3/FOXM1/ATG7 signalling activity and epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) treatment efficacy in patients undergoing EGFR-TKIs treatment. CONCLUSIONS Our findings support that STAT3/FOXM1/ATG7 signalling-induced autophagy is a novel mechanism of resistance to icotinib, and provide insights into potential clinical values of ATG7-dependent autophagy in icotinib treatment.
Collapse
Affiliation(s)
- Xin Lyu
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital, Xi’an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Lizhong Zeng
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital, Xi’an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Jie Shi
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital, Xi’an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Zongjuan Ming
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital, Xi’an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Wei Li
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital, Xi’an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Boxuan Liu
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital, Xi’an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Yang Chen
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital, Xi’an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Bo Yuan
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital, Xi’an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Ruiying Sun
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital, Xi’an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Jingyan Yuan
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital, Xi’an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Nannan Zhao
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital, Xi’an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Xia Yang
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital, Xi’an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi’an, 710004 Shaanxi People’s Republic of China
| | - Guoan Chen
- School of Medicine, Southern University of Science and Technology, No. 1088, Xueyuan Road, Nanshan District, Shenzhen, 518055 Guangdong China
| | - Shuanying Yang
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital, Xi’an Jiaotong University, No. 157, Xiwu Road, Xincheng District, Xi’an, 710004 Shaanxi People’s Republic of China
| |
Collapse
|
5
|
Guo SB, Huang WJ, Tian XP. Brusatol modulates diverse cancer hallmarks and signaling pathways as a potential cancer therapeutic. ACTA MATERIA MEDICA 2022; 1. [DOI: 10.15212/amm-2022-0014] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2024]
Abstract
Cancer is a consequence of uncontrolled cell proliferation that is associated with cell-cycle disruption. It is a multifactorial disease that depends on the modulation of numerous oncogenic signaling pathways and targets. Although a battle against cancer has been waged for centuries, this disease remains a major cause of death worldwide. Because of the development of resistance to current anticancer drugs, substantial effort has been focused on discovering more effective agents for tumor therapy. Natural products have powerful prospects as anticancer drugs. Brusatol, a component isolated from the plant Brucea javanica, has been demonstrated to efficiently combat a wide variety of tumors. Extensive studies have indicated that brusatol exhibits anticancer effects by arresting the cell cycle; promoting apoptosis; inducing autophagy; attenuating epithelial-mesenchymal transition; inhibiting migration, invasion and angiogenesis; and increasing chemosensitivity and radiosensitivity. These effects involve various oncogenic signaling pathways, including the MAPK, NF-κB, PI3K/AKT/mTOR, JAK/STAT and Keap1/Nrf2/ARE signaling pathways. This review describes the evidence suggesting that brusatol is a promising drug candidate for cancer therapeutics.
Collapse
Affiliation(s)
- Song-Bin Guo
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Wei-Juan Huang
- Department of Pharmacology, College of Pharmacy, Jinan University, Guangzhou, P.R. China
| | - Xiao-Peng Tian
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| |
Collapse
|
6
|
Shalini, Kumar V. Have molecular hybrids delivered effective anti-cancer treatments and what should future drug discovery focus on? Expert Opin Drug Discov 2020; 16:335-363. [PMID: 33305635 DOI: 10.1080/17460441.2021.1850686] [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] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Cancer continues to be a big threat and its treatment is a huge challenge among the medical fraternity. Conventional anti-cancer agents are losing their efficiency which highlights the need to introduce new anti-cancer entities for treating this complex disease. A hybrid molecule has a tendency to act through varied modes of action on multiple targets at a given time. Thus, there is the significant scope with hybrid compounds to tackle the existing limitations of cancer chemotherapy. AREA COVERED This perspective describes the most significant hybrids that spring hope in the field of cancer chemotherapy. Several hybrids with anti-proliferative/anti-tumor properties currently approved or in clinical development are outlined, along with a description of their mechanism of action and identified drug targets. EXPERT OPINION The success of molecular hybridization in cancer chemotherapy is quite evident by the number of molecules entering into clinical trials and/or have entered the drug market over the past decade. Indeed, the recent advancements and co-ordinations in the interface between chemistry, biology, and pharmacology will help further the advancement of hybrid chemotherapeutics in the future.List of abbreviations: Deoxyribonucleic acid, DNA; national cancer institute, NCI; peripheral blood mononuclear cells, PBMC; food and drug administration, FDA; histone deacetylase, HDAC; epidermal growth factor receptor, EGFR; vascular endothelial growth factor receptor, VEGFR; suberoylanilide hydroxamic acid, SAHA; farnesyltransferase inhibitor, FTI; adenosine triphosphate, ATP; Tamoxifen, TAM; selective estrogen receptor modulator, SERM; structure activity relationship, SAR; estrogen receptor, ER; lethal dose, LD; half maximal growth inhibitory concentration, GI50; half maximal inhibitory concentration, IC50.
Collapse
Affiliation(s)
- Shalini
- Department of Chemistry, Guru Nanak Dev University, Amritsar-India
| | - Vipan Kumar
- Department of Chemistry, Guru Nanak Dev University, Amritsar-India
| |
Collapse
|
7
|
Bhardwaj M, Leli NM, Koumenis C, Amaravadi RK. Regulation of autophagy by canonical and non-canonical ER stress responses. Semin Cancer Biol 2020; 66:116-128. [PMID: 31838023 PMCID: PMC7325862 DOI: 10.1016/j.semcancer.2019.11.007] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/05/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022]
Abstract
Cancer cells encounter numerous stresses that pose a threat to their survival. Tumor microenviroment stresses that perturb protein homeostasis can produce endoplasmic reticulum (ER) stress, which can be counterbalanced by triggering the unfolded protein response (UPR) which is considered the canonical ER stress response. The UPR is characterized by three major proteins that lead to specific changes in transcriptional and translational programs in stressed cells. Activation of the UPR can induce apoptosis, but also can induce cytoprotective programs such as autophagy. There is increasing appreciation for the role that UPR-induced autophagy plays in supporting tumorigenesis and cancer therapy resistance. More recently several new pathways that connect cell stresses, components of the UPR and autophagy have been reported, which together can be viewed as non-canonical ER stress responses. Here we review recent findings on the molecular mechanisms by which canonical and non-canonical ER stress responses can activate cytoprotective autophagy and contribute to tumor growth and therapy resistance. Autophagy has been identified as a druggable pathway, however the components of autophagy (ATG genes) have proven difficult to drug. It may be the case that targeting the UPR or non-canonical ER stress programs can more effectively block cytoprotective autophagy to enhance cancer therapy. A deeper understanding of these pathways could provide new therapeutic targets in cancer.
Collapse
Affiliation(s)
- Monika Bhardwaj
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nektaria Maria Leli
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ravi K Amaravadi
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
8
|
Yu XQ, Shang XY, Huang XX, Yao GD, Song SJ. Brusatol: A potential anti-tumor quassinoid from Brucea javanica. CHINESE HERBAL MEDICINES 2020; 12:359-366. [PMID: 36120179 PMCID: PMC9476775 DOI: 10.1016/j.chmed.2020.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/08/2020] [Accepted: 05/25/2020] [Indexed: 01/20/2023] Open
Abstract
Brusatol, a triterpene lactone compound mainly from Brucea javanica, sensitizes a broad spectrum of cancer cells. It is known as a specific inhibitor of nuclear factor-erythroid 2-related factor 2 (Nrf2) pathway. In this review, we provide a comprehensive overview on the antitumor effect and molecular mechanisms of brusatol in vitro and in vivo. This review also covers pharmacokinetics studies, modification of dosages forms of brusatol. Increasing evidences have validated the value of brusatol as a chemotherapeutic agent in cancers, which may contribute to drug development and clinical application.
Collapse
|
9
|
Wu J, Ni Y, Yang Q, Mao J, Zhu X, Tao S, Kato K, Zhang J, Wang D, Yamanaka K, An Y. Long-term arsenite exposure decreases autophagy by increased release of Nrf2 in transformed human keratinocytes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 734:139425. [PMID: 32450402 DOI: 10.1016/j.scitotenv.2020.139425] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/07/2020] [Accepted: 05/12/2020] [Indexed: 06/11/2023]
Abstract
Autophagy dysfunction in arsenite toxicity plays critical roles in cancer development and progression. However, the precise mechanisms of arsenite-induced skin cancer by blocking autophagy remain uncertain. Herein, this study investigated molecular mechanisms of arsenite-induced autophagy dysfunction mediated by nuclear factor erythroid-2 related factor 2 (Nrf2) in human keratinocyte (HaCaT) cells. The effects of long-term arsenite exposure on Nrf2 activation and autophagy were established using a siRNA interference assay and western blots. A specific siRNA of Nrf2 was used to verify that autophagy induced by arsenite can be influenced by Nrf2. Specific inhibitors of PI3K (LY294002) and mTOR (Rapamycin) and siRNA of Nrf2 were employed to verify that upregulation of Nrf2 correlated with activating the PI3K/Akt pathway. Downstream mTOR and Bcl2 were upregulated by Nrf2 signaling, inhibiting autophagy initiation in arsenite-exposed HaCaT cells. In conclusion, our data suggest that long-term exposure to arsenite promotes Nrf2 upregulation via the PI3K/Akt pathway and, along with upregulation of downstream mTOR and Bcl2, contributes to autophagy dysfunction in transformed HaCaT cells. This work provides new insights into the mechanisms underlying arsenite-induced autophagy dysfunction in cancer promotion and malignancy progression.
Collapse
Affiliation(s)
- Jing Wu
- Department of Toxicology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Yiping Ni
- Department of Toxicology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Qianlei Yang
- Department of Toxicology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Jiayuan Mao
- Department of Toxicology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Xuerui Zhu
- Department of Toxicology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Shasha Tao
- Department of Toxicology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Koichi Kato
- Laboratory of Environmental Toxicology and Carcinogenesis, School of Pharmacy, Nihon University, Chiba, Japan
| | - Jie Zhang
- Department of Toxicology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Dapeng Wang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, Guizhou Medical University, Guiyang 550025, China.
| | - Kenzo Yamanaka
- Laboratory of Environmental Toxicology and Carcinogenesis, School of Pharmacy, Nihon University, Chiba, Japan.
| | - Yan An
- Department of Toxicology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou 215123, Jiangsu, China.
| |
Collapse
|
10
|
Potential Applications of NRF2 Inhibitors in Cancer Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8592348. [PMID: 31097977 PMCID: PMC6487091 DOI: 10.1155/2019/8592348] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/10/2019] [Accepted: 02/28/2019] [Indexed: 02/07/2023]
Abstract
The NRF2/KEAP1 pathway represents one of the most important cell defense mechanisms against exogenous or endogenous stressors. Indeed, by increasing the expression of several cytoprotective genes, the transcription factor NRF2 can shelter cells and tissues from multiple sources of damage including xenobiotic, electrophilic, metabolic, and oxidative stress. Importantly, the aberrant activation or accumulation of NRF2, a common event in many tumors, confers a selective advantage to cancer cells and is associated to malignant progression, therapy resistance, and poor prognosis. Hence, in the last years, NRF2 has emerged as a promising target in cancer treatment and many efforts have been made to identify therapeutic strategies aimed at disrupting its prooncogenic role. By summarizing the results from past and recent studies, in this review, we provide an overview concerning the NRF2/KEAP1 pathway, its biological impact in solid and hematologic malignancies, and the molecular mechanisms causing NRF2 hyperactivation in cancer cells. Finally, we also describe some of the most promising therapeutic approaches that have been successfully employed to counteract NRF2 activity in tumors, with a particular emphasis on the development of natural compounds and the adoption of drug repurposing strategies.
Collapse
|
11
|
Xue D, Zhou C, Shi Y, Lu H, Xu R, He X. Nuclear transcription factor Nrf2 suppresses prostate cancer cells growth and migration through upregulating ferroportin. Oncotarget 2018; 7:78804-78812. [PMID: 27788496 PMCID: PMC5346678 DOI: 10.18632/oncotarget.12860] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/04/2016] [Indexed: 12/31/2022] Open
Abstract
VTo investigate the effect of nuclear transcription factor Nrf2 on the transcription of Ferroportin (FPN) in prostate cancer cells, and the regulation mechanisms of FPN on cell viability, migration and apoptosis of prostate cancer cells.Empty vectors, pEGFPC1-Nrf2, pEGFPC1-FPN, Si-FPN and Si-Nrf2 were transfected into prostate cancer cell line PC3. The expression of mRNA and protein were measured by real time-PCR (RT-PCR) and western blot. Cell viability, migration, cycle and apoptosis were tested by CCK-8 assay, wound healing and flow cytometry, respectively. The interaction between FPN and Nrf2 was confirmed by chromatin immunoprecipitation (CHIP) assay.The viability, migration and mitosis of PC3 cells could be repressed by over-expressed FPN, with decreased intracellular ferritin. The CHIP assay demonstrated that Nrf2 is one transcription factor of FPN and promotes its transcription. With the increase of Nrf2 in PC3 cells, the viability, migration ability and concentration of ferritin were suppressed, while the apoptosis rate was increased. The above effects were counteracted by down-regulating FPN.FPN could inhibit the prostate cancer cell viability, migration and mitosis, which is also related to a decrease of intracellular ferritin content. In conclusion, Nrf2 suppresses prostate cancer cells viability, migration, and mitosis through upregulating FPN.
Collapse
Affiliation(s)
- Dong Xue
- Department of Urology, Third Affiliated Hospital, Suzhou University, Changzhou 213003, Jiangsu, China
| | - Cuixing Zhou
- Department of Urology, Third Affiliated Hospital, Suzhou University, Changzhou 213003, Jiangsu, China
| | - Yunbo Shi
- Foreign Languages School, Changzhou Institute of Technology, Changzhou 213002, Jiangsu, China
| | - Hao Lu
- Department of Urology, Third Affiliated Hospital, Suzhou University, Changzhou 213003, Jiangsu, China
| | - Renfang Xu
- Department of Urology, Third Affiliated Hospital, Suzhou University, Changzhou 213003, Jiangsu, China
| | - Xiaozhou He
- Department of Urology, Third Affiliated Hospital, Suzhou University, Changzhou 213003, Jiangsu, China
| |
Collapse
|
12
|
Catanzaro E, Calcabrini C, Turrini E, Sestili P, Fimognari C. Nrf2: a potential therapeutic target for naturally occurring anticancer drugs? Expert Opin Ther Targets 2017; 21:781-793. [PMID: 28675319 DOI: 10.1080/14728222.2017.1351549] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Nuclear factor (erythroid-derived-2)-like 2 is one of the most efficient cytoprotective rheostats against exogenous or endogenous oxidative insults. At present, the modulation of the Nrf2 pathway represents an interesting and highly explored strategy in the oncological area. Area covered: In this review, we present and discuss the different modulation of the Nrf2 pathway by some natural compounds with a well demonstrated anticancer activity, and critically analyze the challenges associated with the development of an Nrf2-based anticancer strategy. Expert opinion: Many natural compounds with a well-defined anticancer activity are able to modulate this pathway. Both Nrf2 inducers and inhibitors can be useful as anticancer strategy. However, since Nrf2 modulates many networks potentially involved in the detoxification process of anticancer drugs, its activation in cancer cells could lead to chemoresistance. The switch between a beneficial or detrimental role of Nrf2 in cancer cells essentially depends on the tight control of its activity, the specific conditions of tumor microenvironment, and cell type. In line with the paucity of clear data related to the mechanisms underpinning the role of Nrf2 in cancer development and chemoresistance, discovery and development of Nrf2-based strategies is one of the most critical and challenging assignments for fighting cancers.
Collapse
Affiliation(s)
- Elena Catanzaro
- a Department for Life Quality Studies , Alma Mater Studiorum-University of Bologna , Rimini , Italy
| | - Cinzia Calcabrini
- a Department for Life Quality Studies , Alma Mater Studiorum-University of Bologna , Rimini , Italy
| | - Eleonora Turrini
- a Department for Life Quality Studies , Alma Mater Studiorum-University of Bologna , Rimini , Italy
| | - Piero Sestili
- b Department of Biomolecular Sciences , University of Urbino Carlo Bo , Urbino , Italy
| | - Carmela Fimognari
- a Department for Life Quality Studies , Alma Mater Studiorum-University of Bologna , Rimini , Italy
| |
Collapse
|
13
|
Design, synthesis and cytotoxic evaluation of a novel series of benzo[d]thiazole-2-carboxamide derivatives as potential EGFR inhibitors. Med Chem Res 2017. [DOI: 10.1007/s00044-017-1925-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|