1
|
Praveen Kumar PK, Sundar H, Balakrishnan K, Subramaniam S, Ramachandran H, Kevin M, Michael Gromiha M. The Role of HSP90 and TRAP1 Targets on Treatment in Hepatocellular Carcinoma. Mol Biotechnol 2025; 67:1367-1381. [PMID: 38684604 DOI: 10.1007/s12033-024-01151-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/18/2024] [Indexed: 05/02/2024]
Abstract
Hepatocellular Carcinoma (HCC) is the predominant form of liver cancer and arises due to dysregulation of the cell cycle control machinery. Heat Shock Protein 90 (HSP90) and mitochondrial HSP90, also referred to as TRAP1 are important critical chaperone target receptors for early diagnosis and targeting HCC. Both HSP90 and TRAP1 expression was found to be higher in HCC patients. Hence, the importance of HSP90 and TRAP1 inhibitors mechanism and mitochondrial targeted delivery of those inhibitors function is widely studied. This review also focuses on importance of protein-protein interactions of HSP90 and TRAP1 targets and association of its interacting proteins in various pathways of HCC. To further elucidate the mechanism, systems biology approaches and computational biology approach studies are well explored in the association of inhibition of herbal plant molecules with HSP90 and its mitochondrial type in HCC.
Collapse
Affiliation(s)
- P K Praveen Kumar
- Department of Biotechnology, Sri Venkateswara College of Engineering, Pennalur, Sriperumbudur Tk, Tamil Nadu, 602117, India.
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India.
| | - Harini Sundar
- Department of Biotechnology, Sri Venkateswara College of Engineering, Pennalur, Sriperumbudur Tk, Tamil Nadu, 602117, India
| | - Kamalavarshini Balakrishnan
- Department of Biotechnology, Sri Venkateswara College of Engineering, Pennalur, Sriperumbudur Tk, Tamil Nadu, 602117, India
| | - Sakthivel Subramaniam
- Department of Biotechnology, Sri Venkateswara College of Engineering, Pennalur, Sriperumbudur Tk, Tamil Nadu, 602117, India
| | - Hemalatha Ramachandran
- Department of Biotechnology, Sri Venkateswara College of Engineering, Pennalur, Sriperumbudur Tk, Tamil Nadu, 602117, India
| | - M Kevin
- Department of Biotechnology, Sri Venkateswara College of Engineering, Pennalur, Sriperumbudur Tk, Tamil Nadu, 602117, India
| | - M Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| |
Collapse
|
2
|
Vento F, Privitera A, Caruso G, Nicosia A. A Silibinin-Poly(ε-Caprolactone) Conjugate as an Enhanced Anticancer Agent. Macromol Biosci 2025; 25:e2400510. [PMID: 39937435 PMCID: PMC11995842 DOI: 10.1002/mabi.202400510] [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: 10/19/2024] [Revised: 01/24/2025] [Indexed: 02/13/2025]
Abstract
Poly(ε-caprolactone) (PCL) is a hydrolytically degradable biopolyester used in drug delivery to enhance drug solubility and bioavailability, where drugs are typically incorporated physically within the biopolymeric matrix rather than covalently bonded, due to the limited availability of functional groups required for covalent attachment. In pursuit of developing a facile method for the production of a biopolyester-drug covalent conjugate with effective drug loading capacity, this study reports the synthesis of a covalent Silibinin-PCL conjugate (Sil-PCLHyd) through a two-step approach. This involves the controlled hydrolysis of a high molecular weight PCL to increase the concentration of carboxylic end groups, which are subsequently used for the catalyzed esterification with Silibinin. The Sil-PCLHyd is characterized with mass spectrometry, gel permeation chromatography, thermogravimetric analysis, differential scanning calorimetry, and NMR and UV-vis spectroscopies. The cytotoxic effects of Sil-PCLHyd against colorectal adenocarcinoma cells (Caco-2) are measured through the MTT assay. The results of the Sil-PCLHyd characterization revealed a Silibinin loading of ≈9.8 wt.%. The MTT assay demonstrated that Sil-PCLHyd induced cytotoxic effects at concentrations a hundred times lower than those required for free Silibinin. The proposed approach might represent a reliable pathway for the development of biopolyester-based covalent conjugates with a high drug loading capacity.
Collapse
Affiliation(s)
- Fabiana Vento
- Department of Chemical Sciences and INSTM UdR of CataniaUniversity of CataniaV.le A. Doria 6Catania95125Italy
| | - Anna Privitera
- Department of Drug and Health SciencesUniversity of CataniaV.le A. Doria 6Catania95125Italy
- Department of Biomedical and Biotechnological SciencesUniversity of CataniaVia Santa Sofia 97Catania95123Italy
| | - Giuseppe Caruso
- Department of Drug and Health SciencesUniversity of CataniaV.le A. Doria 6Catania95125Italy
- Unit of Neuropharmacology and Translational NeurosciencesOasi Research Institute‐IRCCSVia Conte Ruggero73Troina94018Italy
| | - Angelo Nicosia
- Department of Chemical Sciences and INSTM UdR of CataniaUniversity of CataniaV.le A. Doria 6Catania95125Italy
| |
Collapse
|
3
|
Ray PP, Islam MA, Islam MS, Han A, Geng P, Aziz MA, Mamun AA. A comprehensive evaluation of the therapeutic potential of silibinin: a ray of hope in cancer treatment. Front Pharmacol 2024; 15:1349745. [PMID: 38487172 PMCID: PMC10937417 DOI: 10.3389/fphar.2024.1349745] [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: 12/05/2023] [Accepted: 02/13/2024] [Indexed: 03/17/2024] Open
Abstract
Natural compounds hold promise in the search for cancer therapies due to their unique chemical structures and combinations that may effectively combat cancer while minimizing toxicity and side effects compared to conventional treatments. Silibinin, a natural lignan, has been found to possess strong anti-cancer activity against several types of human cancers based on emerging research. This study aims to provide an overview of the therapeutic potential of silibinin in the treatment and prevention of cancers. A comprehensive search was conducted using various internet databases such as PubMed, Google Scholar, and ScienceDirect to identify relevant research papers. Silibinin has been shown to exhibit anticancer activity against several types of cancers, including liver, lungs, breast, prostate, colorectal, skin, and bladder cancers. Its multifaceted mechanisms of action contribute to its therapeutic effects. Silibinin exerts antioxidant, anti-inflammatory, anti-proliferative, pro-apoptotic, anti-metastatic, and anti-angiogenic activities, making it a promising candidate for cancer therapy. One of the key mechanisms underlying the anticancer effects of silibinin is its ability to modulate multiple signaling pathways involved in cancer development and progression. It can inhibit the activation of various oncogenic pathways, including PI3K/Akt, NF-κB, Wnt/β-catenin, and MAPK pathways, thereby suppressing cancer cell proliferation, inducing cell cycle arrest, and promoting apoptosis. Silibinin possesses great potential as an effective treatment agent for cancer. The multifaceted mechanisms of action, favorable safety profile, and potential synergistic effects of silibinin with conventional therapies make it an attractive candidate for further investigation and development as a cancer treatment. However, more extensive clinical studies are necessary to fully establish the efficacy, optimal dosage, and long-term effects of silibinin in cancer treatment.
Collapse
Affiliation(s)
- Pantha Prodip Ray
- Department of Pharmacy, State University of Bangladesh, Dhaka, Bangladesh
| | | | - Mohammad Safiqul Islam
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Aixia Han
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui of The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, China
| | - Peiwu Geng
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui of The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, China
| | - Md. Abdul Aziz
- Department of Pharmacy, State University of Bangladesh, Dhaka, Bangladesh
| | - Abdullah Al Mamun
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui of The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, China
| |
Collapse
|
4
|
Chandrababu G, Varkey M, Devan AR, Anjaly MV, Unni AR, Nath LR. Kaempferide exhibits an anticancer effect against hepatocellular carcinoma in vitro and in vivo. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2461-2467. [PMID: 36988659 DOI: 10.1007/s00210-023-02468-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/17/2023] [Indexed: 03/30/2023]
Abstract
CONTEXT Phytochemicals have been promising candidates for cancer therapy, affecting various cancer initiation and progression stages. Kaempferide is a mono methoxy flavone that shows potent anticancer effects on multiple cancers both in vitro and in vivo. MATERIALS AND METHODS We evaluated the anticancer activity of kaempferide against HCC using an MTT ((3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide) assay. HepG2, Huh7, and N1S1 were used for preliminary in vitro studies. This is followed by an apoptosis analysis assessed by caspase-3 and 9. The in vivo effects of the compound were studied in the N1S1 orthotopically injected SD (Sprague Dawley) rat model, where the animal was given kaempferide (25 mg/kg thrice a week) and vehicle (Cremophor:ethanol) iv. The expression of caspase-9 and a critical tumor marker, transforming growth factor beta 1 (TGF-β 1), were assessed in both control and treatment tumor samples. RESULTS Kaempferide-induced dose-dependent cytotoxicity in three HCC cell lines (HepG2: IC50 = 27.94 ± 2.199 µM; Huh7: IC50 = 25.65 ± 0.956 µM; and N1S1: IC50 = 15.18 ± 3.68 µM). Furthermore, caspase-dependent apoptosis was confirmed in vitro. Kaempferide showed a significant reduction in tumor size and tumor volume in vivo. Histopathological evaluation by hematoxylin and eosin (H&E) staining confirmed that altered cells were significantly demolished in the kaempferide-treated animals, which correlates with tumor reduction compared to the vehicle-treated group. Caspase-9 levels were also found to be increased in the treatment group. TGF-β 1, a crucial marker in invasion and metastasis of liver cancer, was also downregulated in the treatment group (control = 207.8 ± 22.9 pg/mL and kaempferide-treated = 157.3 ± 13.8 pg/mL). CONCLUSION We report for the first time the potential of kaempferide as a promising alternative against HCC, which further warrants its clinical validation.
Collapse
Affiliation(s)
- Gopika Chandrababu
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P.O., Kochi, Kerala, 682041, India
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P.O., Kochi, Kerala, 682041, India
| | - Merlin Varkey
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P.O., Kochi, Kerala, 682041, India
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P.O., Kochi, Kerala, 682041, India
| | - Aswathy R Devan
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P.O., Kochi, Kerala, 682041, India
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P.O., Kochi, Kerala, 682041, India
| | - M V Anjaly
- Central Lab Animal Facility, Amrita Institute of Medical Sciences, Kochi, Kerala, 682041, India
| | - Ashok R Unni
- Central Lab Animal Facility, Amrita Institute of Medical Sciences, Kochi, Kerala, 682041, India
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P.O., Kochi, Kerala, 682041, India.
| |
Collapse
|
5
|
Koltai T, Fliegel L. Role of Silymarin in Cancer Treatment: Facts, Hypotheses, and Questions. J Evid Based Integr Med 2022; 27:2515690X211068826. [PMID: 35018864 PMCID: PMC8814827 DOI: 10.1177/2515690x211068826] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/20/2021] [Accepted: 12/06/2021] [Indexed: 12/14/2022] Open
Abstract
The flavonoid silymarin extracted from the seeds of Sylibum marianum is a mixture of 6 flavolignan isomers. The 3 more important isomers are silybin (or silibinin), silydianin, and silychristin. Silybin is functionally the most active of these compounds. This group of flavonoids has been extensively studied and they have been used as hepato-protective substances for the mushroom Amanita phalloides intoxication and mainly chronic liver diseases such as alcoholic cirrhosis and nonalcoholic fatty liver. Hepatitis C progression is not, or slightly, modified by silymarin. Recently, it has also been proposed for SARS COVID-19 infection therapy. The biochemical and molecular mechanisms of action of these substances in cancer are subjects of ongoing research. Paradoxically, many of its identified actions such as antioxidant, promoter of ribosomal synthesis, and mitochondrial membrane stabilization, may seem protumoral at first sight, however, silymarin compounds have clear anticancer effects. Some of them are: decreasing migration through multiple targeting, decreasing hypoxia inducible factor-1α expression, inducing apoptosis in some malignant cells, and inhibiting promitotic signaling among others. Interestingly, the antitumoral activity of silymarin compounds is limited to malignant cells while the nonmalignant cells seem not to be affected. Furthermore, there is a long history of silymarin use in human diseases without toxicity after prolonged administration. The ample distribution and easy accessibility to milk thistle-the source of silymarin compounds, its over the counter availability, the fact that it is a weed, some controversial issues regarding bioavailability, and being a nutraceutical rather than a drug, has somehow led medical professionals to view its anticancer effects with skepticism. This is a fundamental reason why it never achieved bedside status in cancer treatment. However, in spite of all the antitumoral effects, silymarin actually has dual effects and in some cases such as pancreatic cancer it can promote stemness. This review deals with recent investigations to elucidate the molecular actions of this flavonoid in cancer, and to consider the possibility of repurposing it. Particular attention is dedicated to silymarin's dual role in cancer and to some controversies of its real effectiveness.
Collapse
Affiliation(s)
- Tomas Koltai
- Hospital del Centro Gallego de Buenos Aires, Buenos Aires, Argentina
| | | |
Collapse
|
6
|
Abid F, Saleem M, Yasir S, Arshad S, Qureshi S, Bajwa MA, Ashiq S, Tanveer S, Qayyum M, Ashiq K. CANCER EPIGENETICS AND THE ROLE OF DIETARY ELEMENTS. GOMAL JOURNAL OF MEDICAL SCIENCES 2020. [DOI: 10.46903/gjms/17.03.2070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Cancer has been a fatal disease since many decades. Over the time, it is presented in multiple ways and is a matter of consideration as accounts for the high rate of mortality. The aim of the current review was to focus on the genetics, epigenetics factors and role of medicinal plants for the cure of this inimical disease. Related articles available in English language (2002-2018) were reviewed with help of different database, including PubMed, Springer Link, Medline, Google Scholar and ScienceDirect. In order to ensure credibility and accuracy of data only those articles were considered which are published in indexed journals i.e. Web of Science and Scopus. This project was conducted at the Department of Pharmacy, Government College University, Faisalabad, Pakistan from 02-01-2019 to 28-02-2019. The genetic machinery is vibrantly involved in the interpretation of the signals and is observed to be affected by various dietary factors. A sequence of modified activities is observed with use of these dietary elements. However, the modification is reviewed through the histone acetyltransferase (HAT), histone deacetylase (HDAC) and DNA methyl transferase (DNMTs), effecting the expression of gene. These modified genes, in turn then express the signals in multiple reformed ways. Different dietary elements that are used such as polyphenol, alkaloid and flavonoids are effective against cancer. The progression of disease involves genetics and epigenetics due to amplification, translocation and mutation during gene expression. Though, many studies have been conducted elaborating the role of plants and their ingredients which play a part in inhibition of cancerous cells by blockade of cell cycle and apoptosis; more in-depth investigations are still required to identify the new drug target and novel therapeutic modalities.
Collapse
|
7
|
Fan Y, Hou T, Dan W, Liu T, Luan J, Liu B, Li L, Zeng J. Silibinin inhibits epithelial‑mesenchymal transition of renal cell carcinoma through autophagy‑dependent Wnt/β‑catenin signaling. Int J Mol Med 2020; 45:1341-1350. [PMID: 32323735 PMCID: PMC7138295 DOI: 10.3892/ijmm.2020.4521] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/22/2020] [Indexed: 01/07/2023] Open
Abstract
Silibinin is a flavonoid extracted from milk thistle seeds which has been widely used as a hepatoprotective and antioxidant agent. Recently, accumulating evidence has demonstrated the anti-cancer effects of silibinin in various cancer models. It was previously reported that silibinin induced apoptosis and decreased metastasis by activating autophagy in renal cell carcinoma (RCC). However, the underlying molecular mechanisms by which silibinin regulates autophagy remain largely unknown. The aim of the present study was to investigate the effects of silibinin on RCC metastasis in vitro and in vivo, with a focus on autophagy-dependent Wnt/β-catenin signaling. Human RCC 786-O and ACHN cell lines were used as the model system in vitro and RCC xenografts of nude mice were used for in vivo studies. Silibinin inhibited metastasis and epithelial-mesenchymal transition (EMT) of RCC in vitro and in vivo, by regulating the Wnt/β-catenin signaling pathway. Furthermore, silibinin inhibited the Wnt/β-catenin signaling pathway in an autophagy-dependent manner. Autophagic degradation of β-catenin induced by silibinin was associated with the anti-metastatic effects of silibinin against RCC. These findings identify a novel mechanism by which silibinin inhibits EMT and metastasis of RCC, highlighting a potential novel strategy for treating metastatic RCC.
Collapse
Affiliation(s)
- Yizeng Fan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Tao Hou
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Weichao Dan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Tianjie Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jiaxin Luan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Bo Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Lei Li
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jin Zeng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
8
|
Chi C, Zhang C, Liu Y, Nie H, Zhou J, Ding Y. Phytosome-nanosuspensions for silybin-phospholipid complex with increased bioavailability and hepatoprotection efficacy. Eur J Pharm Sci 2020; 144:105212. [PMID: 31923602 DOI: 10.1016/j.ejps.2020.105212] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/30/2019] [Accepted: 01/06/2020] [Indexed: 12/15/2022]
Abstract
Silybin, a natural compound for treating liver disease, has been shown to provide diverse biological activities such as anticancer, antioxidant and hepatoprotective. However, it is still challenging to develop silybin product due to its poor aqueous solubility and limited gastrointestinal absorption. In order to improve the low bioavailability of silybin, a novel formulation of phytosome-nanosuspensions for silybin shielding termed as SPCs-NPs, has been developed herein for hepatoprotection efficacy. We found that SPCs-NPs formulation not only possessed an increased in vitro dissolution rate but also improved plasma concentration in the in vivo pharmacokinetic study. Moreover, SPCs-NPs was provided with more potent hepatoprotective effects in pharmacodynamic assessments. Moreover, physicochemical features including interactions between silybin and phospholipid, and crystalline variation of the optimized SPCs-NPs formulation were confirmed by using Fourier-transform infrared spectrometry (FTIR), 1H nuclear magnetic resonance spectroscopy (H-NMR), differential scanning calorimetry (DSC), and powder X-ray diffraction spectroscopy (PXRD) respectively. Overall, the interesting finding of this study suggested that SPCs-NPs could be applied as a promising formulation for a higher drug bioavailability and better hepatoprotection efficacy.
Collapse
Affiliation(s)
- Cheng Chi
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Chenshuang Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Haichen Nie
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Station Mall Drive, West Lafayette, IN 47907, United States
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.
| | - Yang Ding
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.
| |
Collapse
|
9
|
Xiang Y, Guo Z, Zhu P, Chen J, Huang Y. Traditional Chinese medicine as a cancer treatment: Modern perspectives of ancient but advanced science. Cancer Med 2019; 8:1958-1975. [PMID: 30945475 PMCID: PMC6536969 DOI: 10.1002/cam4.2108] [Citation(s) in RCA: 483] [Impact Index Per Article: 80.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 02/26/2019] [Accepted: 03/07/2019] [Indexed: 12/24/2022] Open
Abstract
Traditional Chinese medicine (TCM) has been practiced for thousands of years and at the present time is widely accepted as an alternative treatment for cancer. In this review, we sought to summarize the molecular and cellular mechanisms underlying the chemopreventive and therapeutic activity of TCM, especially that of the Chinese herbal medicine-derived phytochemicals curcumin, resveratrol, and berberine. Numerous genes have been reported to be involved when using TCM treatments and so we have selectively highlighted the role of a number of oncogene and tumor suppressor genes in TCM therapy. In addition, the impact of TCM treatment on DNA methylation, histone modification, and the regulation of noncoding RNAs is discussed. Furthermore, we have highlighted studies of TCM therapy that modulate the tumor microenvironment and eliminate cancer stem cells. The information compiled in this review will serve as a solid foundation to formulate hypotheses for future studies on TCM-based cancer therapy.
Collapse
Affiliation(s)
- Yuening Xiang
- College of Life and Health SciencesNortheastern UniversityShenyangChina
| | - Zimu Guo
- College of Life and Health SciencesNortheastern UniversityShenyangChina
| | - Pengfei Zhu
- College of Life and Health SciencesNortheastern UniversityShenyangChina
| | - Jia Chen
- College of Life and Health SciencesNortheastern UniversityShenyangChina
| | - Yongye Huang
- College of Life and Health SciencesNortheastern UniversityShenyangChina
| |
Collapse
|
10
|
Flavonoids Effects on Hepatocellular Carcinoma in Murine Models: A Systematic Review. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:6328970. [PMID: 29681978 PMCID: PMC5850900 DOI: 10.1155/2018/6328970] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/06/2017] [Indexed: 02/07/2023]
Abstract
The hepatocellular carcinoma (HCC) is the second most common cause of cancer deaths worldwide. It occurs primarily as manifestation of other pathological processes, such as viral hepatitis, cirrhosis, and toxin exposure that affect directly the cellular process. Studies were selected from PubMed and Scopus databases according to the PRISMA statement. The research filters were constructed using three parameters: flavonoids, hepatocellular carcinoma, and animal model. The bias analysis of the 34 selected works was done using the ARRIVE guidelines. The most widely used flavonoid in the studies was epigallocatechin gallate extracted from green tea. In general, the treatment with different flavonoids presented inhibition of tumor growth and antiangiogenic, antimetastatic, antioxidant, and anti-inflammatory activities. The bias analysis evidenced the absence of methodological processes in all studies, such as the age or weight of the animals, the method of flavonoids' extraction, or the experimental designs, analytical methods, and outcome measures. It has been known that flavonoids have a protective effect against HCC. However, the absence or incomplete characterization of the animal models, treatment protocols, and phytochemical and toxicity analyses impaired the internal validity of the individual studies, making it difficult to determine the effectiveness of plant-derived products in the treatment of HCC.
Collapse
|
11
|
Sun T, Cheung KSC, Liu ZL, Leung F, Lu WW. Matrix metallopeptidase 9 targeted by hsa-miR-494 promotes silybin-inhibited osteosarcoma. Mol Carcinog 2017; 57:262-271. [DOI: 10.1002/mc.22753] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/09/2017] [Accepted: 10/23/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Tianhao Sun
- Li Ka Shing Faculty of Medicine; Department of Orthopaedics and Traumatology; The University of Hong Kong; Hong Kong SAR China
| | - Kelvin S. C. Cheung
- Li Ka Shing Faculty of Medicine; Department of Orthopaedics and Traumatology; The University of Hong Kong; Hong Kong SAR China
| | - Zhi-Li Liu
- Department of Orthopedic Surgery; The First Affiliated Hospital of Nanchang University; Nanchang China
| | - Frankie Leung
- Li Ka Shing Faculty of Medicine; Department of Orthopaedics and Traumatology; The University of Hong Kong; Hong Kong SAR China
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma; Department of Orthopaedics and Traumatology; The University of Hong Kong-Shenzhen Hospital; Shenzhen China
| | - William W. Lu
- Li Ka Shing Faculty of Medicine; Department of Orthopaedics and Traumatology; The University of Hong Kong; Hong Kong SAR China
| |
Collapse
|
12
|
Cytotoxic and toxicogenomic effects of silibinin in bladder cancer cells with different TP53 status. J Biosci 2017; 42:91-101. [PMID: 28229968 DOI: 10.1007/s12038-016-9654-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Silibinin is a natural phenol found in the seeds of the milk thistle plant. Recent data have shown its effectiveness for preventing/treating bladder tumours. Therefore, in this study we investigated the cytotoxic and toxicogenetic activity of silibinin in bladder cancer cells with different TP53 statuses. Two bladder urothelial carcinoma cell lines were used: RT4 (wild-type TP53 gene) and T24 (mutated TP53 gene). Cell proliferation, clonogenic survival, apoptosis rates, genotoxicity and relative expression profile of FRAP/mTOR, FGFR3, AKT2 and DNMT1 genes and of miR100 and miR203 were evaluated. Silibinin promoted decreased proliferation and increased late apoptosis in TP53 mutated cells. Increased early apoptosis rates, primary DNA damage, and decrease of cell colonies in the clonogenic survival assay were detected in both RT4 and T24 cell lines. Down-regulation of FRAP/mTOR, AKT2, FGFR3, DNMT1 and miR100 expression occurred in RT4 cells. Modulation of miR203 was observed in both cell lines. In conclusion, despite the reduction of clone formation in both cell lines, the toxicogenomic effect of silibinin on FRAP/mTOR, AKT2, FGFR3, DNMT1 and miR100 was dependent on the TP53 status. Taken together, the data confirmed the role of silibinin as an antiproliferative compound, whose mechanism of action was related to the TP53 status.
Collapse
|
13
|
Anestopoulos I, Sfakianos AP, Franco R, Chlichlia K, Panayiotidis MI, Kroll DJ, Pappa A. A Novel Role of Silibinin as a Putative Epigenetic Modulator in Human Prostate Carcinoma. Molecules 2016; 22:molecules22010062. [PMID: 28042859 PMCID: PMC6155798 DOI: 10.3390/molecules22010062] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 12/22/2016] [Indexed: 12/11/2022] Open
Abstract
Silibinin, extracted from milk thistle (Silybum marianum L.), has exhibited considerable preclinical activity against prostate carcinoma. Its antitumor and chemopreventive activities have been associated with diverse effects on cell cycle, apoptosis, and receptor-dependent mitogenic signaling pathways. Here we hypothesized that silibinin's pleiotropic effects may reflect its interference with epigenetic mechanisms in human prostate cancer cells. More specifically, we have demonstrated that silibinin reduces gene expression levels of the Polycomb Repressive Complex 2 (PRC2) members Enhancer of Zeste Homolog 2 (EZH2), Suppressor of Zeste Homolog 12 (SUZ12), and Embryonic Ectoderm Development (EED) in DU145 and PC3 human prostate cancer cells, as evidenced by Real Time Polymerase Chain Reaction (RT-PCR). Furthermore immunoblot and immunofluorescence analysis revealed that silibinin-mediated reduction of EZH2 levels was accompanied by an increase in trimethylation of histone H3 on lysine (Κ)-27 residue (H3K27me3) levels and that such response was, in part, dependent on decreased expression levels of phosphorylated Akt (ser473) (pAkt) and phosphorylated EZH2 (ser21) (pEZH2). Additionally silibinin exerted other epigenetic effects involving an increase in total DNA methyltransferase (DNMT) activity while it decreased histone deacetylases 1-2 (HDACs1-2) expression levels. We conclude that silibinin induces epigenetic alterations in human prostate cancer cells, suggesting that subsequent disruptions of central processes in chromatin conformation may account for some of its diverse anticancer effects.
Collapse
Affiliation(s)
- Ioannis Anestopoulos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus, Dragana, 68100 Alexandroupolis, Greece.
| | - Aristeidis P Sfakianos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus, Dragana, 68100 Alexandroupolis, Greece.
| | - Rodrigo Franco
- Redox Biology Center, School of Veterinary Medicine & Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| | - Katerina Chlichlia
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus, Dragana, 68100 Alexandroupolis, Greece.
| | - Mihalis I Panayiotidis
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne NE1 8ST, UK.
| | - David J Kroll
- Department of Pharmaceutical Sciences, College of Science & Technology, Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA.
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus, Dragana, 68100 Alexandroupolis, Greece.
| |
Collapse
|
14
|
Shang RZ, Qu SB, Wang DS. Reprogramming of glucose metabolism in hepatocellular carcinoma: Progress and prospects. World J Gastroenterol 2016; 22:9933-9943. [PMID: 28018100 PMCID: PMC5143760 DOI: 10.3748/wjg.v22.i45.9933] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 09/30/2016] [Accepted: 11/13/2016] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal cancers, and its rate of incidence is rising annually. Despite the progress in diagnosis and treatment, the overall prognoses of HCC patients remain dismal due to the difficulties in early diagnosis and the high level of tumor invasion, metastasis and recurrence. It is urgent to explore the underlying mechanism of HCC carcinogenesis and progression to find out the specific biomarkers for HCC early diagnosis and the promising target for HCC chemotherapy. Recently, the reprogramming of cancer metabolism has been identified as a hallmark of cancer. The shift from the oxidative phosphorylation metabolic pathway to the glycolysis pathway in HCC meets the demands of rapid cell proliferation and offers a favorable microenvironment for tumor progression. Such metabolic reprogramming could be considered as a critical link between the different HCC genotypes and phenotypes. The regulation of metabolic reprogramming in cancer is complex and may occur via genetic mutations and epigenetic modulations including oncogenes, tumor suppressor genes, signaling pathways, noncoding RNAs, and glycolytic enzymes etc. Understanding the regulatory mechanisms of glycolysis in HCC may enrich our knowledge of hepatocellular carcinogenesis and provide important foundations in the search for novel diagnostic biomarkers and promising therapeutic targets for HCC.
Collapse
|
15
|
Dietz BM, Hajirahimkhan A, Dunlap TL, Bolton JL. Botanicals and Their Bioactive Phytochemicals for Women's Health. Pharmacol Rev 2016; 68:1026-1073. [PMID: 27677719 PMCID: PMC5050441 DOI: 10.1124/pr.115.010843] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Botanical dietary supplements are increasingly popular for women's health, particularly for older women. The specific botanicals women take vary as a function of age. Younger women will use botanicals for urinary tract infections, especially Vaccinium macrocarpon (cranberry), where there is evidence for efficacy. Botanical dietary supplements for premenstrual syndrome (PMS) are less commonly used, and rigorous clinical trials have not been done. Some examples include Vitex agnus-castus (chasteberry), Angelica sinensis (dong quai), Viburnum opulus/prunifolium (cramp bark and black haw), and Zingiber officinale (ginger). Pregnant women have also used ginger for relief from nausea. Natural galactagogues for lactating women include Trigonella foenum-graecum (fenugreek) and Silybum marianum (milk thistle); however, rigorous safety and efficacy studies are lacking. Older women suffering menopausal symptoms are increasingly likely to use botanicals, especially since the Women's Health Initiative showed an increased risk for breast cancer associated with traditional hormone therapy. Serotonergic mechanisms similar to antidepressants have been proposed for Actaea/Cimicifuga racemosa (black cohosh) and Valeriana officinalis (valerian). Plant extracts with estrogenic activities for menopausal symptom relief include Glycine max (soy), Trifolium pratense (red clover), Pueraria lobata (kudzu), Humulus lupulus (hops), Glycyrrhiza species (licorice), Rheum rhaponticum (rhubarb), Vitex agnus-castus (chasteberry), Linum usitatissimum (flaxseed), Epimedium species (herba Epimedii, horny goat weed), and Medicago sativa (alfalfa). Some of the estrogenic botanicals have also been shown to have protective effects against osteoporosis. Several of these botanicals could have additional breast cancer preventive effects linked to hormonal, chemical, inflammatory, and/or epigenetic pathways. Finally, although botanicals are perceived as natural safe remedies, it is important for women and their healthcare providers to realize that they have not been rigorously tested for potential toxic effects and/or drug/botanical interactions. Understanding the mechanism of action of these supplements used for women's health will ultimately lead to standardized botanical products with higher efficacy, safety, and chemopreventive properties.
Collapse
Affiliation(s)
- Birgit M Dietz
- University of Illinois at Chicago/National Institutes of Health Center for Botanical Dietary Supplements, Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Atieh Hajirahimkhan
- University of Illinois at Chicago/National Institutes of Health Center for Botanical Dietary Supplements, Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Tareisha L Dunlap
- University of Illinois at Chicago/National Institutes of Health Center for Botanical Dietary Supplements, Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Judy L Bolton
- University of Illinois at Chicago/National Institutes of Health Center for Botanical Dietary Supplements, Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
16
|
Zhu XX, Ding YH, Wu Y, Qian LY, Zou H, He Q. Silibinin: a potential old drug for cancer therapy. Expert Rev Clin Pharmacol 2016; 9:1323-1330. [PMID: 27362364 DOI: 10.1080/17512433.2016.1208563] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Silibinin is mixture of flavonolignans extracted from milk thistle and often has been used in the treatment of acute and chronic liver disorders caused by toxins, drug, alcohol and hepatitis and gall bladder disorders for its antioxidant and hepatoprotective properties. Areas covered: However, increasing evidence suggest that silibinin is not solely limited in the treatment of these diseases. Further research suggests that silymarin may function diversely and may serve as a novel therapy for cancer therapy, such as lung cancer, prostatic cancer, colon cancer, breast cancer, bladder cancer and hepatocellular carcinoma by regulating cancer cells growth, proliferation, apoptosis, angiogenesis and many other mechanism. Expert commentary: In this review, in order to provide potential new treatment for these cancer, we summarize the recent anti-cancer findings of silibinin in these cancer and clarify the mechanisms of this effect.
Collapse
Affiliation(s)
- Xing-Xing Zhu
- a Department of Nephrology , Zhejiang Provincial People's Hospital , Hangzhou , China
| | - Ya-Hui Ding
- b Department of Cardiology , Zhejiang Provincial People's Hospital , Hangzhou , China
| | - Yi Wu
- c Department of Hematology , Zhejiang Provincial People's Hospital , Hangzhou , China
| | - Lin-Yan Qian
- b Department of Cardiology , Zhejiang Provincial People's Hospital , Hangzhou , China
| | - Hai Zou
- b Department of Cardiology , Zhejiang Provincial People's Hospital , Hangzhou , China
| | - Qiang He
- a Department of Nephrology , Zhejiang Provincial People's Hospital , Hangzhou , China
| |
Collapse
|
17
|
Novel Investigations of Flavonoids as Chemopreventive Agents for Hepatocellular Carcinoma. BIOMED RESEARCH INTERNATIONAL 2015; 2015:840542. [PMID: 26858957 PMCID: PMC4695650 DOI: 10.1155/2015/840542] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/19/2015] [Indexed: 12/16/2022]
Abstract
We would like to highlight the application of natural products to hepatocellular carcinoma (HCC). We will focus on the natural products known as flavonoids, which target this disease at different stages of hepatocarcinogenesis. In spite of the use of chemotherapy and radiotherapy in treating HCC, patients with HCC still face poor prognosis because of the nature of multidrug resistance and toxicity derived from chemotherapy and radiotherapy. Flavonoids can be found in many vegetables, fruits, and herbal medicines that exert their different anticancer effects via different intracellular signaling pathways and serve as antioxidants. In this review, we will discuss seven common flavonoids that exert different biological effects against HCC via different pathways.
Collapse
|
18
|
Busch C, Burkard M, Leischner C, Lauer UM, Frank J, Venturelli S. Epigenetic activities of flavonoids in the prevention and treatment of cancer. Clin Epigenetics 2015; 7:64. [PMID: 26161152 PMCID: PMC4497414 DOI: 10.1186/s13148-015-0095-z] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 06/17/2015] [Indexed: 02/07/2023] Open
Abstract
Aberrant epigenetic modifications are described in an increasing number of pathological conditions, including neurodegenerative diseases, cardiovascular diseases, diabetes mellitus type 2, obesity and cancer. The general reversibility of epigenetic changes makes them an attractive and promising target e.g. in the treatment of cancer. Thus, a growing number of epigenetically active compounds are currently tested in clinical trials for their therapeutic potential. Interestingly, many phytochemicals present in plant foods, particularly flavonoids, are suggested to be able to alter epigenetic cellular mechanisms. Flavonoids are natural phenol compounds that form a large group of secondary plant metabolites with interesting biological activities. They can be categorized into six major subclasses, which display diverse properties affecting the two best characterized epigenetic mechanisms: modulation of the DNA methylation status and histone acetylation. High dietary flavonoid intake has strongly been suggested to reduce the risk of numerous cancer entities in a large body of epidemiological studies. Established health-promoting effects of diets rich in fruit and vegetables are faced by efforts to use purified flavonoids as supplements or pharmaceuticals, whereupon data on the latter applications remain controversial. The purpose of this review is to give an overview of current research on flavonoids to further elucidate their potential in cancer prevention and therapy, thereby focusing on their distinct epigenetic activities.
Collapse
Affiliation(s)
- Christian Busch
- Division of Dermatologic Oncology, Department of Dermatology and Allergology, Medical University Hospital, Tuebingen, Germany
| | - Markus Burkard
- Division of Dermatologic Oncology, Department of Dermatology and Allergology, Medical University Hospital, Tuebingen, Germany ; Department of Internal Medicine I, Medical University Hospital, Otfried-Mueller-Str. 27, 72076 Tuebingen, Germany
| | - Christian Leischner
- Department of Internal Medicine I, Medical University Hospital, Otfried-Mueller-Str. 27, 72076 Tuebingen, Germany
| | - Ulrich M Lauer
- Department of Internal Medicine I, Medical University Hospital, Otfried-Mueller-Str. 27, 72076 Tuebingen, Germany
| | - Jan Frank
- Institute of Biological Chemistry and Nutrition, University of Hohenheim, Stuttgart, Germany
| | - Sascha Venturelli
- Department of Internal Medicine I, Medical University Hospital, Otfried-Mueller-Str. 27, 72076 Tuebingen, Germany
| |
Collapse
|
19
|
Mastron JK, Siveen KS, Sethi G, Bishayee A. Silymarin and hepatocellular carcinoma: a systematic, comprehensive, and critical review. Anticancer Drugs 2015; 26:475-86. [PMID: 25603021 DOI: 10.1097/cad.0000000000000211] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The blessed milk thistle (Silybum marianum L.), a flowering plant native to Mediterranean Europe, has been consumed and extensively used as a cure for various chronic liver ailments over several centuries. Milk thistle extract, known as silymarin, is a complex mixture of seven major flavonolignans and one flavonoid. The phytoconstituents of silymarin owe their therapeutic and hepatoprotective effects to their strong antioxidant and anti-inflammatory properties. Primary liver cancer, also known as hepatocellular carcinoma (HCC), occurs in a milieu of oxidative stress and inflammation. The etiology of HCC includes chronic infection with hepatitis B and C viruses, cirrhosis, and exposure to dietary and environmental hepatocarcinogens. Current therapeutic options for HCC, including surgical resection and liver transplantation, have limited benefits and are essentially ineffective. Chemoprevention, using phytochemicals with potent antioxidant and anti-inflammatory properties, represents a fascinating strategy, which has been a subject of intense investigation in the recent years. In this review, we explore the potential role of silymarin as a chemopreventive and therapeutic agent for HCC. The review systematically evaluates the preclinical in-vitro and in-vivo studies investigating the effects of silymarin and its constituents on HCC. The biochemical mechanisms involved in the anti-liver-cancer effects of silymarin have been presented. The current status of clinical studies evaluating the potential of role of silymarin in liver cancer, especially that caused by hepatitis C virus, has also been examined. Potential challenges and future directions of research involved in the 'bench-to-bedside' transition of silymarin phytoconstituents for the chemoprevention and treatment of HCC have also been discussed.
Collapse
Affiliation(s)
- Jeanetta K Mastron
- aAmerican University of Health Sciences, Signal Hill bDepartment of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, California, USA cDepartment of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore dInterim Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | | | | | | |
Collapse
|
20
|
Herbal product silibinin-induced programmed cell death is enhanced by metformin in cervical cancer cells at the dose without influence on nonmalignant cells. J Appl Biomed 2015. [DOI: 10.1016/j.jab.2014.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
21
|
Tsai CC, Chuang TW, Chen LJ, Niu HS, Chung KM, Cheng JT, Lin KC. Increase in apoptosis by combination of metformin with silibinin in human colorectal cancer cells. World J Gastroenterol 2015; 21:4169-4177. [PMID: 25892866 PMCID: PMC4394077 DOI: 10.3748/wjg.v21.i14.4169] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 10/25/2014] [Accepted: 12/22/2014] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the effect of metformin on silibinin-induced apoptosis in human colorectal cancer (COLO 205) cells. METHODS MTT assays were performed to quantify cell viability. Western blot assays were applied to identify the expression of signaling proteins. RESULTS The combined treatment of COLO 205 cells with metformin and silibinin decreased cell survival at a dose insufficient to influence the non-malignant cells [Human colonic epithelial cells (HCoEpiC)]. Silibinin and metformin increased phosphatase and tensin homolog and 5'-adenosine monophosphate-activated protein kinase expression in COLO 205 cells and inhibited the phosphorylation of mammol/Lalian target of rapamycin. This combined treatment resulted in an increase in the expression of activated caspase 3 and apoptosis inducing factor, indicating apoptosis. CONCLUSION The combined treatment of human colorectal cancer cells with silibinin and metformin may induce apoptosis at a dose that does not affect HCoEpiC. This finding reveals a potential therapeutic strategy for the treatment of colorectal cancer.
Collapse
|
22
|
Schnekenburger M, Dicato M, Diederich M. Plant-derived epigenetic modulators for cancer treatment and prevention. Biotechnol Adv 2014; 32:1123-32. [DOI: 10.1016/j.biotechadv.2014.03.009] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 03/14/2014] [Accepted: 03/16/2014] [Indexed: 12/12/2022]
|
23
|
Bishayee A. The role of inflammation and liver cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 816:401-35. [PMID: 24818732 DOI: 10.1007/978-3-0348-0837-8_16] [Citation(s) in RCA: 193] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Persistent inflammation is known to promote and exacerbate malignancy. Primary liver cancer, mostly hepatocellular carcinoma (HCC), is a clear example of inflammation-related cancer as more than 90 % of HCCs arise in the context of hepatic injury and inflammation. HCC represents the fifth most common malignancy and the third leading cause of cancer-related death worldwide with about one million new cases diagnosed every year with almost an equal number of deaths. Chronic unresolved inflammation is associated with persistent hepatic injury and concurrent regeneration, leading to sequential development of fibrosis, cirrhosis, and eventually HCC. Irrespective of the intrinsic differences among various etiological factors, a common denominator at the origin of HCC is the perpetuation of a wound-healing response activated by parenchymal cell death and the resulting inflammatory cascade. Hence, the identification of fundamental inflammatory signaling pathways causing transition from chronic liver injury to dysplasia and HCC could depict new predictive biomarkers and targets to identify and treat patients with chronic liver inflammation. This chapter critically discusses the roles of several major cytokines, chemokines, growth factors, transcription factors, and enzymes as well as a distinct network of inflammatory signaling pathways in the development and progression of HCC. It also highlights and analyzes preclinical animal studies showing innovative approaches of targeting inflammatory mediators and signaling by a variety of natural compounds and synthetic agents to achieve effective therapy as well as prevention of hepatic malignancy. Additionally, current limitations and potential challenges associated with the inhibition of inflammatory signaling as well as future directions of research to accelerate clinical development of anti-inflammatory agents to prevent and treat liver cancer are presented.
Collapse
Affiliation(s)
- Anupam Bishayee
- Department of Pharmaceutical Sciences, School of Pharmacy, American University of Health Sciences, 1600 East Hill Street, Signal Hill, CA, 90755, USA,
| |
Collapse
|
24
|
Anestopoulos I, Voulgaridou GP, Georgakilas AG, Franco R, Pappa A, Panayiotidis MI. Epigenetic therapy as a novel approach in hepatocellular carcinoma. Pharmacol Ther 2014; 145:103-19. [PMID: 25205159 DOI: 10.1016/j.pharmthera.2014.09.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 09/02/2014] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of liver malignancy and one with high fatality. Its 5-year survival rate remains low and thus, there is a need for improvement of current treatment strategies as well as development of novel targeted methodologies in order to optimize existing therapeutic protocols. To this end, only recently, it was discovered that its pathophysiology also involves epigenetic alterations in DNA methylation, histone modifications and/or non-coding microRNA patterns. Unlike genetic events, epigenetic alterations are reversible and thus potentially considered to be an alternative option in cancer treatment protocols. In this review, we describe the general characteristics and resulted major alterations of the epigenetic machinery as well as current state of progress of epigenetic therapy (via different single or combinatorial experimental approaches) in HCC.
Collapse
Affiliation(s)
- Ioannis Anestopoulos
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | | | - Alexandros G Georgakilas
- School of Applied Mathematical & Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Rodrigo Franco
- Redox Biology Center, School of Veterinary Medicine & Biomedical Sciences, Redox Biology Center, University of Nebraska-Lincoln, USA
| | - Aglaia Pappa
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | | |
Collapse
|
25
|
Kim TH, Woo JS, Kim YK, Kim KH. Silibinin induces cell death through reactive oxygen species-dependent downregulation of notch-1/ERK/Akt signaling in human breast cancer cells. J Pharmacol Exp Ther 2014; 349:268-78. [PMID: 24472723 DOI: 10.1124/jpet.113.207563] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
The present study was undertaken to determine the underlying mechanism of silibinin-induced cell death in human breast cancer cell lines MCF7 and MDA-MB-231. Silibinin-induced cell death was attenuated by antioxidants, N-acetylcysteine (NAC) and 6-hydroxy-2,5,7,8-tetramethylchromane-2-carboxylic acid, suggesting that the effect of silibinin was dependent on generation of reactive oxygen species (ROS). Western blot analysis showed that silibinin induced downregulation of extracellular signal-regulated kinase (ERK) and Akt. When cells were transiently transfected with constitutively active (ca) mitogen-activated protein kinase (MEK), an upstream kinase of ERK and caAkt, they showed resistance to silibinin-induced cell death. Silibinin decreased the cleavage of Notch-1 mRNA and protein levels. Notch-1-overexpressed cells were resistant to the silibinin-induced cell death. Inhibition of Notch-1 signaling was dependent on ROSgeneration. Overexpression of Notch-1 prevented silibinin-induced inhibition of ERK and Akt phosphorylation. Silibinin-induced cell death was accompanied by increased cleavage of caspase-3 and was prevented by caspase-3 inhibitor in MDA-MB-231 cells but not in MCF7 cells. Silibinin induced translocation of apoptosis-inducing factor (AIF), which was blocked by NAC, and transfection of caMEK and caAkt. Silibinin-induced cell death was prevented by silencing of AIF expression using small interfering AIF RNA in MCF7 cells but not in MDA-MB-231 cells. In conclusion, silibinin induces cell death through an AIF-dependent mechanism in MCF7 cells and a caspase-3-dependent mechanism in MDA-MB-231 cells, and ROS generation and Notch-1 signaling act upstream of the ERK and Akt pathway. These data suggest that silibinin may serve as a potential agent for induction of apoptosis in human breast cancer cells.
Collapse
Affiliation(s)
- Thae Hyun Kim
- Department of Physiology (T.H.K., J.S.W., Y.K.K.) and Department of Obstetrics and Gynecology (K.H.K.), Pusan National University School of Medicine, Yangsan, Republic of Korea; and Pusan National University Hospital Biomedical Research Institute and Pusan Cancer Center (K.H.K.), Pusan, Republic of Korea
| | | | | | | |
Collapse
|
26
|
Ghasemi R, Ghaffari SH, Momeny M, Pirouzpanah S, Yousefi M, Malehmir M, Alimoghaddam K, Ghavamzadeh A. Multitargeting and antimetastatic potentials of silibinin in human HepG-2 and PLC/PRF/5 hepatoma cells. Nutr Cancer 2013; 65:590-9. [PMID: 23659451 DOI: 10.1080/01635581.2013.770043] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most common sort of primary liver malignancy with poor prognosis. This study aimed at examining the effects of silibinin (a putative antimetastatic agent) on some transcriptional markers mechanistically related to HCC recurrence and metastasis in HepG-2 [hepatitis B virus (HBV)-negative and P53 intact) and PLC/PRF/5 (HBV-positive and P53 mutated) cells. The expression of 27 genes in response to silibinin was evaluated by real-time RT-PCR. The MMP gelatinolytic assay and microculture tetrazolium test (MTT) were tested. Silibinin was capable of suppressing the transcriptional levels of ANGPT2, ATP6L, CAP2, CCR6, CCR7, CLDN-10, cortactin, CXCR4, GLI2, HK2, ID1, KIAA0101, mortalin, PAK1, RHOA, SPINK1, and STMN1 as well as the enzymatic activity of MMP-2 but promoted the transcripts of CREB3L3, DDX3X, and PROX1 in both cells. Some significant differences between the cells in response to silibinin were detected that might be related to the differences of the cells in terms of HBV infection and/or P53 mutation, suggesting the possible influence of silibinin on HCC through biological functions of these 2 prognostic factors. In conclusion, our findings suggest that silibinin could potentially function as a multitargeting antimetastatic agent and might provide new insights for HCC therapy particularly for HBV-related and/or P53-mutated HCCs.
Collapse
Affiliation(s)
- Reza Ghasemi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Hu Y, Wang S, Wu X, Zhang J, Chen R, Chen M, Wang Y. Chinese herbal medicine-derived compounds for cancer therapy: a focus on hepatocellular carcinoma. JOURNAL OF ETHNOPHARMACOLOGY 2013; 149:601-12. [PMID: 23916858 DOI: 10.1016/j.jep.2013.07.030] [Citation(s) in RCA: 148] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 07/23/2013] [Accepted: 07/24/2013] [Indexed: 05/20/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hepatocellular carcinoma (HCC) as the major histological subtype of primary liver cancer remains one of the most common malignancies worldwide. Due to the complicated molecular pathogenesis of HCC, the option for effective systemic treatment is quite limited. There exists a critical need to explore and evaluate possible alternative strategies for effective control of HCC. With a long history of clinical use, Chinese herbal medicine (CHM) is emerging as a noticeable choice for its multi-level, multi-target and coordinated intervention effects against HCC. With the aids of phytochemistry and molecular biological approaches, in the past decades many CHM-derived compounds have been carefully studied through both preclinical and clinical researches and have shown great potential in novel anti-HCC natural product development. The present review aimed at providing the most recent developments on anti-HCC compounds derived from CHM, especially their underlying pharmacological mechanisms. MATERIALS AND METHODS A systematic search of anti-HCC compounds from CHM was carried out focusing on literatures published both in English (PubMed, Scopus, Web of Science and Medline) and in Chinese academic databases (Wanfang and CNKI database). RESULTS In this review, we tried to give a timely and comprehensive update about the anti-HCC effects and targets of several representative CHM-derived compounds, namely curcumin, resveratrol, silibinin, berberine, quercetin, tanshinone II-A and celastrol. Their mechanisms of anti-HCC behaviors, potential side effects or toxicity and future research directions were discussed. CONCLUSION Herbal compounds derived from CHM are of much significance in devising new drugs and providing unique ideas for the war against HCC. We propose that these breakthrough findings may have important implications for targeted-HCC therapy and modernization of CHM.
Collapse
Affiliation(s)
- Yangyang Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | | | | | | | | | | | | |
Collapse
|
28
|
Su CH, Chen LJ, Liao JF, Cheng JT. Increase of phosphatase and tensin homolog by silymarin to inhibit human pharynx squamous cancer. J Med Food 2013; 16:778-84. [PMID: 23909904 PMCID: PMC3778994 DOI: 10.1089/jmf.2012.2534] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 05/24/2013] [Indexed: 01/11/2023] Open
Abstract
Silymarin is an active principle from the seeds of the milk thistle plant and is widely used as a hepatoprotective gent due to its antioxidant-like activity. In the present study, we evaluated the potential efficacy of silymarin against oral cancer and investigated its possible mechanism of action. Cell viability assay and western blotting analyses were used to identify silymarin-induced apoptotic cell death in human pharynx squamous cell carcinoma (FaDu) cells. The short interfering RNA (siRNA) is used to confirm the role of phosphatase and tensin homolog (PTEN) in silymarin-induced apoptosis. Treatment of FaDu cells with silymarin resulted in a significant decrease in cell viability (up to 70%). Silymarin inhibited the phosphorylation of Akt (over 10-fold) with an increase in expression of PTEN (five to sixfold). Consequently, the level of Bcl-2 expression was decreased five to sixfold and caspase 3 activated to induce apoptosis. Treatment with siRNA specific to PTEN gene diminished the action of silymarin. The results suggest that silymarin inhibits the Akt signaling pathway by increasing PTEN expression in FaDu cells and directly affects Bcl-2 family members. Also, we demonstrated the inhibitory activity of silymarin for oral cancer is related to cell survival. These mechanisms may in part explain the actions of silymarin and provide a rationale for the development of silymarin as an anticancer agent.
Collapse
Affiliation(s)
- Chin-Hui Su
- Institute of Pharmacology, National Yang-Ming University, Taipei City, Taiwan
- Department of Otorhinolaryngology, Mackay Memorial Hospital, Taipei City, Taiwan
- Mackay Medicine, Nursing and Management College, Taipei City, Taiwan
| | - Li-Jen Chen
- Department of Pharmacology, Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Jyh Fei Liao
- Institute of Pharmacology, National Yang-Ming University, Taipei City, Taiwan
| | - Juei-Tang Cheng
- Department of Pharmacology, Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
- Department of Medical Research, Chi-Mei Medical Center, Yung Kang, Tainan City, Taiwan
| |
Collapse
|
29
|
Ting H, Deep G, Agarwal R. Molecular mechanisms of silibinin-mediated cancer chemoprevention with major emphasis on prostate cancer. AAPS J 2013; 15:707-16. [PMID: 23588585 PMCID: PMC3691417 DOI: 10.1208/s12248-013-9486-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Accepted: 04/02/2013] [Indexed: 02/08/2023] Open
Abstract
Despite advances in early detection, prostate cancer remains the second highest cancer mortality in American men, and even successful interventions are associated with enormous health care costs as well as prolonged deleterious effects on quality of patient life. Prostate cancer chemoprevention is one potential avenue to alleviate these burdens. It is a regime whereby long-term treatments are intended to prevent or arrest cancer development, in contrast to more direct intervention upon disease diagnosis. Based on this intention, cancer chemoprevention generally focuses on the use of nontoxic chemical agents which are well-tolerated for prolonged usage that is necessary to address prostate cancer's multistage and lengthy period of progression. One such nontoxic natural agent is the flavonoid silibinin, derived from the milk thistle plant (Silybum marianum), which has ancient medicinal usage and potent antioxidant activity. Based on these properties, silibinin has been investigated in a host of cancer models where it exhibits broad-spectrum efficacy against cancer progression both in vitro and in vivo without noticeable toxicity. Specifically in prostate cancer models, silibinin has shown the ability to modulate cell signaling, proliferation, apoptosis, epithelial to mesenchymal transition, invasion, metastasis, and angiogenesis, which taken together provides strong support for silibinin as a candidate prostate cancer chemopreventive agent.
Collapse
Affiliation(s)
- Harold Ting
- />Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| | - Gagan Deep
- />Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
- />University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
| | - Rajesh Agarwal
- />Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
- />University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado USA
- />Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, 12850 E. Montview Blvd, Room V20-2118, Box C238, Aurora, Colorado 80045 USA
| |
Collapse
|
30
|
Anestopoulos I, Kavo A, Tentes I, Kortsaris A, Panayiotidis M, Lazou A, Pappa A. Silibinin protects H9c2 cardiac cells from oxidative stress and inhibits phenylephrine-induced hypertrophy: potential mechanisms. J Nutr Biochem 2013; 24:586-94. [PMID: 22818713 DOI: 10.1016/j.jnutbio.2012.02.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 01/26/2012] [Accepted: 02/17/2012] [Indexed: 02/07/2023]
Abstract
Cardiac hypertrophy is the main response of the heart to various extrinsic and intrinsic stimuli, and it is characterized by specific molecular and phenotypic changes. Recent in vitro and in vivo studies indicate the involvement of reactive oxygen species in the hypertrophic response. In this study, silibinin, a plant flavonolignan extracted from milk thistle with potent antioxidant activity, was evaluated for its effects in (a) preventing hydrogen peroxide (H2O2)-induced cellular damage and (b) blocking the phenylephrine-induced hypertrophic response. Using the in vitro model of embryonic rat heart-derived H9c2 cells, we showed that silibinin has a rather safe profile as concentrations up to 200μM did not affect cell viability. Pretreatment of H9c2 cells with silibinin resulted in better protection of H9c2 cells under conditions of H2O2-induced cellular stress compared to untreated cells as indicated by cell viability and DNA fragmentation assays. Furthermore, silibinin attenuated the phenylephrine-induced hypertrophic response as evidenced by the measurement of cell surface, up-regulation of atrial natriuretic peptide and increase of cellular protein levels. Moreover, silibinin repressed the phenylephrine-induced phosphorylation of ERK1/2 kinases, while it appeared to inhibit the weakly activated by phenylephrine phosphorylation of Akt. Based on our results, silibinin may attenuate the phenylephrine-induced hypertrophic response of H9c2 cells via antioxidant mechanisms involving mainly the inhibition of the intracellular signaling pathways mediated by ERK1/2 MAPKs and Akt.
Collapse
Affiliation(s)
- Ioannis Anestopoulos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus, Dragana, Alexandroupolis 68100, Greece
| | | | | | | | | | | | | |
Collapse
|
31
|
Kauntz H, Bousserouel S, Gossé F, Raul F. Epigenetic effects of the natural flavonolignan silibinin on colon adenocarcinoma cells and their derived metastatic cells. Oncol Lett 2013; 5:1273-1277. [PMID: 23599778 PMCID: PMC3629096 DOI: 10.3892/ol.2013.1190] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 01/18/2013] [Indexed: 12/22/2022] Open
Abstract
Epigenetic modifications are important in tumorigenesis. The most frequent epigenetic phenomena in cancer are histone deacetylation and DNA hypermethylation, which lead to gene silencing, particularly of tumor suppressor genes. However, monotherapies with histone deacetylase (HDAC) or DNA methyltransferase (DNMT) inhibitors lack efficacy, hence there is a need to enhance their anticancer action in a safe and effective combination therapy. The present study investigated the epigenetic effects of the natural flavonolignan silibinin in a model of colon cancer progression, the primary adenocarcinoma cells SW480 and their derived metastatic cells SW620. Silibinin did not change the activity of HDACs, but it was able to significantly inhibit DNMT activity in both SW480 and SW620 cells. The clinically used HDAC inhibitor, suberoylanilide hydroxamic acid (SAHA), and the broad spectrum HDAC inhibitor, trichostatin A (TSA), combined with silibinin demonstrated synergistic effects on cell death induction, may be related to its DNMT inhibition properties. The present data suggest that treatments combining silibinin and HDAC inhibitors may represent a promising approach, given the non-toxic nature of silibinin and the fact that HDAC inhibitors selectively target cancer cells.
Collapse
Affiliation(s)
- Henriette Kauntz
- Department of Nutritional Cancer Prevention, University of Strasbourg, Unit EA 4438, Faculty of Medicine; ; IRCAD-EITS, F-67000 Strasbourg, France
| | | | | | | |
Collapse
|
32
|
Stagos D, Amoutzias GD, Matakos A, Spyrou A, Tsatsakis AM, Kouretas D. Chemoprevention of liver cancer by plant polyphenols. Food Chem Toxicol 2012; 50:2155-70. [DOI: 10.1016/j.fct.2012.04.002] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 03/31/2012] [Accepted: 04/02/2012] [Indexed: 02/07/2023]
|
33
|
Sun H, Yu L, Wei H, Liu G. A novel antihepatitis drug, bicyclol, prevents liver carcinogenesis in diethylnitrosamine-initiated and phenobarbital-promoted mice tumor model. J Biomed Biotechnol 2012; 2012:584728. [PMID: 22500097 PMCID: PMC3303747 DOI: 10.1155/2012/584728] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 09/21/2011] [Accepted: 09/21/2011] [Indexed: 11/28/2022] Open
Abstract
Bicyclol, an antihepatitis drug developed by Chinese scientists, has been shown to prevent the malignant transformation induced by 3-methylcholanthrene and 12-O-tetradecanoylphorbol-13-acetate in WB-F344 rat liver epithelial cells. This study provides further evidence on its role as a chemopreventive agent in experimental mice with diethylnitrosamine- (DEN-) initiated and phenobarbital- (PB-) promoted liver carcinoma. Liver tissue and serum were collected. In the two-stage model of hepatocarcinogenesis in mice, oral administration of bicyclol (100, 200 mg/kg) before DEN injection showed significant reduction in the incidence of hepatocellular foci, nodules, or carcinoma. Histopathological examination revealed that there was no hepatocellular carcinoma (HCC) and hepatoma formation in the mice pretreated with bicyclol (200 mg/kg) at week 20, while the mice treated with DEN/PB developed 33.3% HCC and 55.6% hepatoma. Furthermore, the serum levels of alanine aminotransferase (ALT), alkaline phosphatase (ALP), and α-fetal protein (AFP) in serum significantly increased in the DEN/PB model group in comparison with the control group. Pretreatment with bicyclol showed a marked reduction in the above condition. Bicyclol also decreased the expression of AFP and proliferating cell nuclear antigen level in the liver tissue and attenuated the decrease in body weight. In this study, we also found that 10 weeks after stopping the administration of PB and drugs, the control and bicyclol-treated (200 mg/kg) animals showed no HCC and hepatoma formation at the time of termination whereas DEN/PB-induced mice developed 100% hepatoma and 50% HCC. These results further indicate that bicyclol has the chemopreventive potential for liver carcinogenesis induced by carcinogens.
Collapse
Affiliation(s)
- Hua Sun
- Department of Pharmacology, Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.
| | | | | | | |
Collapse
|
34
|
Yan X, Gardner TR, Grieco M, Herath SAC, Jang JH, Kirchoff D, Njoh L, Shantha Kumara HMC, Naffouje S, Whelan RL. Perioperative polyphenon E- and siliphos-inhibited colorectal tumor growth and metastases without impairment of gastric or abdominal wound healing in mouse models. Surg Endosc 2012; 26:1856-64. [PMID: 22258296 DOI: 10.1007/s00464-011-2114-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 11/14/2011] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Perioperative anticancer therapy that does not impair wound healing is needed to counter the persistent proangiogenic plasma compositional changes that occur after colorectal resection. Polyphenon E (PolyE), a green tea derivative (main component EGCG), and Siliphos (main component silibinin), from the milk thistle plant, both have antitumor effects. This study assessed the impact of PolyE/Siliphos (PES) on wound healing and the growth of CT-26 colon cancer in several murine models. METHODS One wound healing and three tumor studies were performed. Tumor Study (TS)1 assessed the impact of PES on subcutaneous tumor growth, whereas TS2 assessed PES's impact on subcutaneous growth when given pre- and post-CO(2) pneumoperitoneum (pneumo), sham laparotomy, or anesthesia alone. TS3 determined the ability of PES to limit hepatic metastases (mets) after portal venous injection of tumor cells. In the final study, laparotomy and gastrotomy wound healing were assessed several ways. BALB/c mice were used for all studies. The drugs were given via drinking water (PolyE) and gavage (Siliphos), daily, for 7-9 days preprocedure and for 7-21 days postoperatively. Tumor mass, number/size of hepatic mets, and proliferation and apoptosis rates were assessed. The abdominal breaking strength and energy to failure were measured postmortem as was gastric bursting pressures. RESULTS PES significantly inhibited subcutaneous growth in the nonoperative setting. PES also significantly decreased the number/size of liver mets when given perioperatively. Abdominal wound breaking strength, energy to wound failure, and collagen content were not altered by PES; gastrotomy bursting strength also was not affected by PES. Neither drug alone had a significant impact on tumor growth. CONCLUSIONS The PES combination inhibited subcutaneous and hepatic tumor growth yet did not impair wound healing. PES holds promise as a perioperative anticancer therapy.
Collapse
Affiliation(s)
- Xiaohong Yan
- Colon & Rectum Surgery, St. Luke's Roosevelt Hospital Center, 432 West, 58th Street, Room 517, New York, NY 10019, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Yu HC, Chen LJ, Cheng KC, Li YX, Yeh CH, Cheng JT. Silymarin inhibits cervical cancer cell through an increase of phosphatase and tensin homolog. Phytother Res 2011; 26:709-15. [PMID: 22016029 DOI: 10.1002/ptr.3618] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 06/12/2011] [Accepted: 06/25/2011] [Indexed: 12/18/2022]
Abstract
Silymarin is an active constituent contained in the seeds of the milk thistle plant and is widely used as a hepatic protection agent due to its antioxidant-like activity. In the present study we evaluated the potential action of silymarin against cervical cancer and investigated its mechanism of action. Treatment of cervical cancer cells (C-33A) with silymarin resulted in a significant decrease in cell viability. Silymarin induced apoptosis through the modulation of Bcl-2 family proteins and activation of caspase 3. Silymarin also inhibited the phosphorylation of Akt with an increase in expression of phosphatase and tensin homolog (PTEN). We also observed that silymarin suppressed C-33A cell invasion and wound-healing migration in a concentration-dependent manner. Western-blot analysis showed that silymarin significantly inhibited the expression of matrix metalloproteinase-9 (MMP-9) in C-33A cells. Furthermore, we applied siRNA to lower the PTEN gene, which diminished the anticancer actions of silymarin. Taken together, these results show that silymarin has the potential to suppress the survival, migration and invasion of C-33A cancer cells; thus, it could be developed as a promising agent for the treatment of cervical cancer in the future.
Collapse
Affiliation(s)
- Hann-Chin Yu
- Department of Obstetrics and Gynecology, Zhudong Veterans Hospital, Zhudong City, Taiwan
| | | | | | | | | | | |
Collapse
|
36
|
Tan W, Lu J, Huang M, Li Y, Chen M, Wu G, Gong J, Zhong Z, Xu Z, Dang Y, Guo J, Chen X, Wang Y. Anti-cancer natural products isolated from chinese medicinal herbs. Chin Med 2011. [PMID: 21777476 DOI: 10.1186/1749-8546-6- 27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
In recent years, a number of natural products isolated from Chinese herbs have been found to inhibit proliferation, induce apoptosis, suppress angiogenesis, retard metastasis and enhance chemotherapy, exhibiting anti-cancer potential both in vitro and in vivo. This article summarizes recent advances in in vitro and in vivo research on the anti-cancer effects and related mechanisms of some promising natural products. These natural products are also reviewed for their therapeutic potentials, including flavonoids (gambogic acid, curcumin, wogonin and silibinin), alkaloids (berberine), terpenes (artemisinin, β-elemene, oridonin, triptolide, and ursolic acid), quinones (shikonin and emodin) and saponins (ginsenoside Rg3), which are isolated from Chinese medicinal herbs. In particular, the discovery of the new use of artemisinin derivatives as excellent anti-cancer drugs is also reviewed.
Collapse
Affiliation(s)
- Wen Tan
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Jinjian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,College of Life Sciences, Zhejiang Chinese Medical University, 548 Binwen Rd., Binjiang Dist., Hangzhou 310053, Zhejiang, China
| | - Mingqing Huang
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,College of Pharmacy, Fujian University of Traditional Chinese Medicine, No.1 Huatuo Rd., Shangjie University Town, Fuzhou 350108, Fujian, China
| | - Yingbo Li
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Guosheng Wu
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Jian Gong
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Zhangfeng Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Zengtao Xu
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Yuanye Dang
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Jiajie Guo
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| |
Collapse
|
37
|
Tan W, Lu J, Huang M, Li Y, Chen M, Wu G, Gong J, Zhong Z, Xu Z, Dang Y, Guo J, Chen X, Wang Y. Anti-cancer natural products isolated from chinese medicinal herbs. Chin Med 2011; 6:27. [PMID: 21777476 PMCID: PMC3149025 DOI: 10.1186/1749-8546-6-27] [Citation(s) in RCA: 259] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 07/22/2011] [Indexed: 02/06/2023] Open
Abstract
In recent years, a number of natural products isolated from Chinese herbs have been found to inhibit proliferation, induce apoptosis, suppress angiogenesis, retard metastasis and enhance chemotherapy, exhibiting anti-cancer potential both in vitro and in vivo. This article summarizes recent advances in in vitro and in vivo research on the anti-cancer effects and related mechanisms of some promising natural products. These natural products are also reviewed for their therapeutic potentials, including flavonoids (gambogic acid, curcumin, wogonin and silibinin), alkaloids (berberine), terpenes (artemisinin, β-elemene, oridonin, triptolide, and ursolic acid), quinones (shikonin and emodin) and saponins (ginsenoside Rg3), which are isolated from Chinese medicinal herbs. In particular, the discovery of the new use of artemisinin derivatives as excellent anti-cancer drugs is also reviewed.
Collapse
Affiliation(s)
- Wen Tan
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Jinjian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,College of Life Sciences, Zhejiang Chinese Medical University, 548 Binwen Rd., Binjiang Dist., Hangzhou 310053, Zhejiang, China
| | - Mingqing Huang
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,College of Pharmacy, Fujian University of Traditional Chinese Medicine, No.1 Huatuo Rd., Shangjie University Town, Fuzhou 350108, Fujian, China
| | - Yingbo Li
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Guosheng Wu
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Jian Gong
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Zhangfeng Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Zengtao Xu
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Yuanye Dang
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Jiajie Guo
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China.,Institute of Chinese Medical Sciences, University of Macau, Av. Padre Toma's Pereira S.J., Taipa, Macao SAR, China
| |
Collapse
|
38
|
Dunnick JK, Singh B, Nyska A, Peckham J, Kissling GE, Sanders JM. Investigating the potential for toxicity from long-term use of the herbal products, goldenseal and milk thistle. Toxicol Pathol 2011; 39:398-409. [PMID: 21300790 DOI: 10.1177/0192623310394211] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Two-year toxicity studies were conducted on the widely used herbal products, goldenseal and milk thistle, in male and female F344/N rats and B6C3F1 mice. With goldenseal root powder, the primary finding was an increase in liver tumors in rats and mice, and with milk thistle extract, a decrease in spontaneous background tumors including mammary gland tumors in female rats and liver tumors in male mice. Increased tumorigenicity in rodents exposed to goldenseal root powder may be due in part to the topoisomerase inhibition properties of berberine, a major alkaloid constituent in goldenseal, or its metabolite, berberrubine. In the clinic, use of topoisomerase-inhibiting agents has been associated with secondary tumor formation and inhibition in DNA repair processes. In contrast, the radical-scavenging and antioxidant properties of silibinin and other flavonolignans in milk thistle extract may have contributed to the decrease in background tumors in rodents in the present studies. The fate of the active constituents of goldenseal and milk thistle is similar in humans and rodents; therefore, the modes of action may translate across species. Further studies are needed to extrapolate the findings to humans.
Collapse
Affiliation(s)
- June K Dunnick
- National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA.
| | | | | | | | | | | |
Collapse
|
39
|
DHF-18, a new synthetic flavonoid, induced a mitochondrial-mediated apoptosis of hepatocarcinoma cells in vivo and in vitro. Eur J Pharmacol 2011; 651:33-40. [DOI: 10.1016/j.ejphar.2010.10.067] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 10/07/2010] [Accepted: 10/29/2010] [Indexed: 11/20/2022]
|
40
|
Park CE, Yun H, Lee EB, Min BI, Bae H, Choe W, Kang I, Kim SS, Ha J. The antioxidant effects of genistein are associated with AMP-activated protein kinase activation and PTEN induction in prostate cancer cells. J Med Food 2010; 13:815-20. [PMID: 20673057 DOI: 10.1089/jmf.2009.1359] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Epidemiological evidence suggests a lower incidence of prostate cancer in Asian countries, where soy products are more frequently consumed than in Western countries, indicating that isoflavones from soy have chemopreventive activities in prostate cells. Here, we tested the effects of the soy isoflavone genistein on antioxidant enzymes in DU145 prostate cancer cells. Genistein significantly decreased reactive oxygen species levels and induced the expression of the antioxidant enzymes manganese (Mn) superoxide dismutase (SOD) and catalase, which were associated with AMP-activated protein kinase (AMPK) and phosphatase and tensin homolog deleted from chromosome 10 (PTEN) pathways. The induced expression of catalase, MnSOD, and PTEN were attenuated by pretreatment with a pharmacological inhibitor for AMPK, indicating the effects of genistein primarily depend on AMPK. Furthermore, PTEN is essential for genistein activity, as shown by PTEN transfection in PTEN-deficient PC3 cells. Thus, genistein induces antioxidant enzymes through AMPK activation and increased PTEN expression.
Collapse
Affiliation(s)
- Chang Eun Park
- Department of East-West Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Deep G, Agarwal R. Antimetastatic efficacy of silibinin: molecular mechanisms and therapeutic potential against cancer. Cancer Metastasis Rev 2010; 29:447-63. [PMID: 20714788 PMCID: PMC3928361 DOI: 10.1007/s10555-010-9237-0] [Citation(s) in RCA: 178] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer is a major health problem around the world. Research efforts in the last few decades have been successful in providing better and effective treatments against both early stage and localized cancer, but clinical options against advanced metastatic stage/s of cancer remain limited. The high morbidity and mortality in most of the cancers are attributed to their metastatic spread to distant organs. Due to its extreme clinical relevance, metastasis has been extensively studied and is now understood as a highly complex biological event that involves multiple steps including acquisition of invasiveness by cancer cells, intravasation into circulatory system, survival in the circulation, arrest in microvasculature, extravasation, and growth at distant organs. The increasing understanding of molecular underpinnings of these events has provided excellent opportunity to target metastasis especially through nontoxic and biologically effective nutraceuticals. Silibinin, a popular dietary supplement isolated from milk thistle seed extracts, is one such natural agent that has shown biological efficacy through pleiotropic mechanisms against a variety of cancers and is currently in clinical trials. Recent preclinical studies have also shown strong efficacy of silibinin to target cancer cell's migratory and invasive characteristics as well as their ability to metastasize to distant organs. Detailed mechanistic analyses revealed that silibinin targets signaling molecules involved in the regulation of epithelial-to-mesenchymal transition, proteases activation, adhesion, motility, invasiveness as well as the supportive tumor-microenvironment components, thereby inhibiting metastasis. Overall, the long history of human use, remarkable nontoxicity, and preclinical efficacy strongly favor the clinical use of silibinin against advanced metastatic cancers.
Collapse
Affiliation(s)
- Gagan Deep
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Denver, Aurora, Colorado 80045
- University of Colorado Cancer Center, University of Colorado Denver, Aurora, Colorado 80045
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Denver, Aurora, Colorado 80045
- University of Colorado Cancer Center, University of Colorado Denver, Aurora, Colorado 80045
| |
Collapse
|
42
|
Schmit TL, Ledesma MC, Ahmad N. Modulating polo-like kinase 1 as a means for cancer chemoprevention. Pharm Res 2010; 27:989-98. [PMID: 20107874 PMCID: PMC2873067 DOI: 10.1007/s11095-010-0051-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 01/05/2010] [Indexed: 12/18/2022]
Abstract
Naturally occurring agents have always been appreciated for their medicinal value for both their chemopreventive and therapeutic effects against cancer. In fact, the majority of the drugs we use today, including the anti-cancer agents, were originally derived from natural compounds, either in their native form or modified to enhance their bioavailability or specificity. It is believed that for maximum effectiveness, it will useful to design novel target-based agents for chemoprevention as well as the treatment of cancer. Recent studies have shown that the serine/threonine kinase polo-like kinase (Plk) 1 is widely overexpressed in a variety of cancers and is being increasingly appreciated as a target for cancer management. Additionally, several chemopreventive agents have been shown to inhibit Plk1 in cancer cells. In this review, we will discuss if Plk1 could also be a target for designing novel strategies for cancer chemoprevention.
Collapse
Affiliation(s)
- Travis L. Schmit
- Department of Dermatology, University of Wisconsin, 1300 University Avenue, Medical Science Center, Room 423, Madison, Wisconsin 53706, USA
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Mark C. Ledesma
- Department of Dermatology, University of Wisconsin, 1300 University Avenue, Medical Science Center, Room 423, Madison, Wisconsin 53706, USA
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, 1300 University Avenue, Medical Science Center, Room 423, Madison, Wisconsin 53706, USA
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, Wisconsin, USA
- University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
43
|
Abstract
IMPORTANCE OF THE FIELD Due to the failure and severe toxicity of cancer chemotherapy, silibinin, a natural flavonoid from the seeds of milk thistle, has recently received more attention for its potential anticancer and nontoxic roles in animals and humans. Silibinin has clearly demonstrated inhibition of multiple cancer cell signaling pathways, including growth inhibition, inhibition of angiogenesis, chemosensitization, and inhibition of invasion and metastasis. Cumulative evidence implicates that silibinin is a potential agent for cancer chemoprevention and chemotherapy. AREAS COVERED IN THIS REVIEW Our aim is to discuss the recent progress of silibinin in regulating multiple anticancer proliferative signaling pathways; the review covers literature mainly from the past 3 - 5 years. WHAT THE READER WILL GAIN One of the strategies for tumor therapy is eradication of cancer cells through targeting specific cell-proliferative pathways. This review highlights the current knowledge of silibinin in regulating multiple cellular proliferative pathways in cancer cells, including receptor tyrosine kinases, androgen receptor, STATs, NF-kappaB, cell cycle regulatory and apoptotic signaling pathways. TAKE HOME MESSAGE The molecular mechanisms of silibinin-mediated antiproliferative effects are mainly via receptor tyrosine kinases, androgen receptor, STATs, NF-kappaB, cell cycle regulatory and apoptotic signaling pathways in various cancer cells. Targeting inhibition of proliferative pathways through silibinin treatment may provide a new approach for improving chemopreventive and chemotherapeutic effects.
Collapse
Affiliation(s)
- Lei Li
- The First Hospital of Xi'an Jiaotong University, Department of Urology, 277 Yanta West Road, Xi'an 710061, People's Republic of China
| | | | | | | |
Collapse
|
44
|
McCarty MF, Barroso-Aranda J, Contreras F. Practical strategies for suppressing hypoxia-inducible factor activity in cancer therapy. Med Hypotheses 2010; 74:789-97. [PMID: 20089365 DOI: 10.1016/j.mehy.2009.12.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Accepted: 12/16/2009] [Indexed: 12/18/2022]
Abstract
The utility of anti-angiogenic strategies for cancer control is strongly compromised by hypoxia-driven phenotypic changes in cancer cells, which make cancer cells more invasive and more prone to give rise to metastases. A key mediator of this phenotypic shift is the transcription factor hypoxia-inducible factor-1 (HIF-1), which acts directly and indirectly to promote the epidermal-mesenchymal transition, boost cancer invasiveness, increase production of angiogenic factors, and induce chemoresistance. In some cancers, HIF-1 activity is constitutively elevated even in aerobic environments, making the cancer harder to treat and control. Practical strategies for suppressing HIF-1 activation may include the following: inhibiting NF-kappaB activation with salicylic acid and/or silibinin, which should decrease transcription of the HIF-1alpha gene; suppressing translation of HIF-1alpha mRNA with drugs that inhibit mTOR or topoisomerase I; supporting the effective activity of prolyl hydroxylases - which promote proteasomal degradation of HIF-1alpha under aerobic conditions - with antioxidant measures, alpha-ketoglutarate, and possibly dichloroacetate; promoting the O(2)-independent proteasomal degradation of HIF-1alpha with agents that inhibit the chaperone protein Hsp90; and blocking HIF-1 binding to its DNA response elements with anthracyclines. The utility of various combinations of these strategies should be tested in cancer cell cultures and rodent xenograft models; initial efforts in this regard have yielded encouraging results. Comprehensive strategies for suppressing HIF-1 activity can be expected to complement the efficacy of cancer chemotherapy and of effective anti-angiogenic regimens.
Collapse
Affiliation(s)
- Mark F McCarty
- Oasis of Hope Hospital, Paseo Playas 19, Playas de Tijuana, Tijuana, B.C., Mexico.
| | | | | |
Collapse
|