1
|
Malla S, Nyinawabera A, Neupane R, Pathak R, Lee D, Abou-Dahech M, Kumari S, Sinha S, Tang Y, Ray A, Ashby CR, Yang MQ, Babu RJ, Tiwari AK. Novel Thienopyrimidine-Hydrazinyl Compounds Induce DRP1-Mediated Non-Apoptotic Cell Death in Triple-Negative Breast Cancer Cells. Cancers (Basel) 2024; 16:2621. [PMID: 39123351 PMCID: PMC11311031 DOI: 10.3390/cancers16152621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 08/12/2024] Open
Abstract
Apoptosis induction with taxanes or anthracyclines is the primary therapy for TNBC. Cancer cells can develop resistance to anticancer drugs, causing them to recur and metastasize. Therefore, non-apoptotic cell death inducers could be a potential treatment to circumvent apoptotic drug resistance. In this study, we discovered two novel compounds, TPH104c and TPH104m, which induced non-apoptotic cell death in TNBC cells. These lead compounds were 15- to 30-fold more selective in TNBC cell lines and significantly decreased the proliferation of TNBC cells compared to that of normal mammary epithelial cell lines. TPH104c and TPH104m induced a unique type of non-apoptotic cell death, characterized by the absence of cellular shrinkage and the absence of nuclear fragmentation and apoptotic blebs. Although TPH104c and TPH104m induced the loss of the mitochondrial membrane potential, TPH104c- and TPH104m-induced cell death did not increase the levels of cytochrome c and intracellular reactive oxygen species (ROS) and caspase activation, and cell death was not rescued by incubating cells with the pan-caspase inhibitor, carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]-fluoromethylketone (Z-VAD-FMK). Furthermore, TPH104c and TPH104m significantly downregulated the expression of the mitochondrial fission protein, DRP1, and their levels determined their cytotoxic efficacy. Overall, TPH104c and TPH104m induced non-apoptotic cell death, and further determination of their cell death mechanisms will aid in the development of new potent and efficacious anticancer drugs to treat TNBC.
Collapse
Affiliation(s)
- Saloni Malla
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (S.M.); (A.N.); (R.N.); (D.L.); (M.A.-D.); (S.K.)
| | - Angelique Nyinawabera
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (S.M.); (A.N.); (R.N.); (D.L.); (M.A.-D.); (S.K.)
| | - Rabin Neupane
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (S.M.); (A.N.); (R.N.); (D.L.); (M.A.-D.); (S.K.)
| | - Rajiv Pathak
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Donghyun Lee
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (S.M.); (A.N.); (R.N.); (D.L.); (M.A.-D.); (S.K.)
| | - Mariam Abou-Dahech
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (S.M.); (A.N.); (R.N.); (D.L.); (M.A.-D.); (S.K.)
| | - Shikha Kumari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (S.M.); (A.N.); (R.N.); (D.L.); (M.A.-D.); (S.K.)
| | - Suman Sinha
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, UP, India;
| | - Yuan Tang
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43606, USA;
| | - Aniruddha Ray
- Department of Physics, College of Math’s and Natural Sciences, University of Toledo, Toledo, OH 43606, USA;
| | - Charles R. Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy, St. John’s University, Queens, NY 11439, USA;
| | - Mary Qu Yang
- MidSouth Bioinformatics Center and Joint Bioinformatics Graduate Program of University of Arkansas at Little Rock, University of Arkansas for Medical Sciences, Little Rock, AR 72204, USA;
| | - R. Jayachandra Babu
- Department of Drug Discovery & Development, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849, USA;
| | - Amit K. Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (S.M.); (A.N.); (R.N.); (D.L.); (M.A.-D.); (S.K.)
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| |
Collapse
|
2
|
Zhao T, Nong X, Zhang X, Zhou X, Yu Z, Li X, Chen G. Four new diterpenoids from the aerial parts of Leucas zeylanica (L.) R. Br. Fitoterapia 2024; 175:105948. [PMID: 38588904 DOI: 10.1016/j.fitote.2024.105948] [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: 12/25/2023] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/10/2024]
Abstract
Four new undescribed halimane- and labdane-type diterpenoids, named zeylleucapenoids E-H (1-4), along with four known analogues (5-8), were isolated from the aerial parts of Leucas zeylanica (L.) R. Br. Their structures were determined by comprehensive spectroscopic analysis and computational calculations. Compounds 1 and 2 are the highly modified halimane diterpenoids featuring a 6/6/6-fused tricyclic system with an unusual six-membered 6,11-ether ring. Compound 8 exhibits nontoxic effects for zebrafish embryo, while it displays efficient reduction against NO production in a dose-dependent manner and strongly suppresses the secretion of LPS-induced TNF-α and IL-6 cytokines in RAW264.7 macrophages. In addition, marked reductions of iNOS and COX-2 expression were observed. Molecular docking analysis indicated that 8 has high affinities with the target amino acid residues on protein-binding sites, which may be a possible mechanism contributing to the anti-inflammatory potential of this molecule.
Collapse
Affiliation(s)
- Ting Zhao
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou, Hainan 571158, China; School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, China; Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Xuhua Nong
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou, Hainan 571158, China; Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Xuan Zhang
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou, Hainan 571158, China; Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Xueming Zhou
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou, Hainan 571158, China; Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Zhangxin Yu
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou, Hainan 571158, China; Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Xiaobao Li
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou, Hainan 571158, China; Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China.
| | - Guangying Chen
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou, Hainan 571158, China; Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China.
| |
Collapse
|
3
|
Gupta J, Jalil AT, Riyad Muedii ZAH, Aminov Z, Alsaikhan F, Ramírez-Coronel AA, Ramaiah P, Farhood B. The Radiosensitizing Potentials of Silymarin/Silibinin in Cancer: A Systematic Review. Curr Med Chem 2024; 31:6992-7014. [PMID: 37921180 DOI: 10.2174/0109298673248404231006052436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/10/2023] [Accepted: 09/11/2023] [Indexed: 11/04/2023]
Abstract
INTRODUCTION Although radiotherapy is one of the main cancer treatment modalities, exposing healthy organs/tissues to ionizing radiation during treatment and tumor resistance to ionizing radiation are the chief challenges of radiotherapy that can lead to different adverse effects. It was shown that the combined treatment of radiotherapy and natural bioactive compounds (such as silymarin/silibinin) can alleviate the ionizing radiation-induced adverse side effects and induce synergies between these therapeutic modalities. In the present review, the potential radiosensitization effects of silymarin/silibinin during cancer radiation exposure/radiotherapy were studied. METHODS According to the PRISMA guideline, a systematic search was performed for the identification of relevant studies in different electronic databases of Google Scholar, PubMed, Web of Science, and Scopus up to October 2022. We screened 843 articles in accordance with a predefined set of inclusion and exclusion criteria. Seven studies were finally included in this systematic review. RESULTS Compared to the control group, the cell survival/proliferation of cancer cells treated with ionizing radiation was considerably less, and silymarin/silibinin administration synergistically increased ionizing radiation-induced cytotoxicity. Furthermore, there was a decrease in the tumor volume, weight, and growth of ionizing radiation-treated mice as compared to the untreated groups, and these diminutions were predominant in those treated with radiotherapy plus silymarin/ silibinin. Furthermore, the irradiation led to a set of biochemical and histopathological changes in tumoral cells/tissues, and the ionizing radiation-induced alterations were synergized following silymarin/silibinin administration (in most cases). CONCLUSION In most cases, silymarin/silibinin administration could sensitize the cancer cells to ionizing radiation through an increase of free radical formation, induction of DNA damage, increase of apoptosis, inhibition of angiogenesis and metastasis, etc. However, suggesting the use of silymarin/silibinin during radiotherapeutic treatment of cancer patients requires further clinical studies.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, U.P., India
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | | | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
- Department of Scientific Affairs, Tashkent State Dental Institute, 103 Makhtumkuli Str., Tashkent, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Andrés Alexis Ramírez-Coronel
- Psychometry and Ethology Laboratory, Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Catholic University of Cuenca, Cuenca, Ecuador
- Epidemiology and Biostatistics Research Group, CES University, Medellin, Colombia
- Educational Statistics Research Group (GIEE), National University of Education, Cuenca, Ecuador
| | | | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
4
|
Zhao T, Zhang X, Nong XH, Zhou XM, Chai RR, Li XB, Chen GY. Zeylleucapenoids A-D, Highly Oxygenated Diterpenoids with Anti-Inflammatory Activity from Leucas zeylanica (L.) R. Br. Molecules 2023; 28:molecules28114472. [PMID: 37298948 DOI: 10.3390/molecules28114472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Four previously undescribed highly oxygenated diterpenoids (1-4), zeylleucapenoids A-D, characterized by halimane and labdane skeletons, were isolated from the aerial parts of Leucas zeylanica. Their structures were elucidated primarily via NMR experiments. The absolute configuration of 1 was established using theoretical ECD calculations and X-ray crystallographic analysis, whereas those for 2-4 were assigned using theoretical ORD calculations. Zeylleucapenoids A-D were tested for anti-inflammatory activity against nitric oxide (NO) production in RAW264.7 macrophages, of which only 4 showed significant efficacy with an IC50 value of 38.45 μM. Further, active compound 4 was also evaluated for the inhibition of the release of pro-inflammatory cytokines TNF-α and IL-6 and was found to have a dose-dependent inhibitory effect, while it showed nontoxic activity for zebrafish embryos. A subsequent Western blotting experiment revealed that 4 inhibited the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Furthermore, molecular docking analysis indicated that the possible mechanism of action for 4 may be bind to targets via hydrogen and hydrophobic bond interactions.
Collapse
Affiliation(s)
- Ting Zhao
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou 571158, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Xuan Zhang
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou 571158, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Xu-Hua Nong
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou 571158, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Xue-Ming Zhou
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou 571158, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Ru-Ru Chai
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou 571158, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Xiao-Bao Li
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou 571158, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Guang-Ying Chen
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou 571158, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| |
Collapse
|
5
|
Singh M, Kadhim MM, Turki Jalil A, Oudah SK, Aminov Z, Alsaikhan F, Jawhar ZH, Ramírez-Coronel AA, Farhood B. A systematic review of the protective effects of silymarin/silibinin against doxorubicin-induced cardiotoxicity. Cancer Cell Int 2023; 23:88. [PMID: 37165384 PMCID: PMC10173635 DOI: 10.1186/s12935-023-02936-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/03/2023] [Indexed: 05/12/2023] Open
Abstract
PURPOSE Although doxorubicin chemotherapy is commonly applied for treating different malignant tumors, cardiotoxicity induced by this chemotherapeutic agent restricts its clinical use. The use of silymarin/silibinin may mitigate the doxorubicin-induced cardiac adverse effects. For this aim, the potential cardioprotective effects of silymarin/silibinin against the doxorubicin-induced cardiotoxicity were systematically reviewed. METHODS In this study, we performed a systematic search in accordance with PRISMA guideline for identifying all relevant studies on "the role of silymarin/silibinin against doxorubicin-induced cardiotoxicity" in different electronic databases up to June 2022. Sixty-one articles were obtained and screened based on the predefined inclusion and exclusion criteria. Thirteen eligible papers were finally included in this review. RESULTS According to the echocardiographic and electrocardiographic findings, the doxorubicin-treated groups presented a significant reduction in ejection fraction, tissue Doppler peak mitral annulus systolic velocity, and fractional shortening as well as bradycardia, prolongation of QT and QRS interval. However, these echocardiographic abnormalities were obviously improved in the silymarin plus doxorubicin groups. As well, the doxorubicin administration led to induce histopathological and biochemical changes in the cardiac cells/tissue; in contrast, the silymarin/silibinin co-administration could mitigate these induced alterations (for most of the cases). CONCLUSION According to the findings, it was found that the co-administration of silymarin/silibinin alleviates the doxorubicin-induced cardiac adverse effects. Silymarin/silibinin exerts its cardioprotective effects via antioxidant, anti-inflammatory, anti-apoptotic activities, and other mechanisms.
Collapse
Affiliation(s)
- Mandeep Singh
- Department of Physical Education, University of Jammu, Srinagar, Jammu, India
| | - Mustafa M Kadhim
- Department of Dentistry, Kut University College, Kut, Wasit, 52001, Iraq
- Medical Laboratory Techniques Department, Al-Farahidi University, Baghdad, 10022, Iraq
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq.
| | | | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
- Department of Scientific Affairs, Tashkent State Dental Institute, 103 Makhtumkuli Str., Tashkent, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| | - Zanko Hassan Jawhar
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Erbil, Kurdistan Region, Iraq
- Clinical Biochemistry Department, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region, Iraq
| | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Cuenca, Ecuador
- Epidemiology and Biostatistics Research Group, CES University, Medellín, Colombia
- Educational Statistics Research Group (GIEE), National University of Education, Cuenca, Ecuador
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
6
|
Balaji S, Neupane R, Malla S, Khupse R, Amawi H, Kumari S, Tukaramrao DB, Chattopadhyay S, Ashby CR, Boddu SHS, Karthikeyan C, Trivedi P, Raman D, Tiwari AK. IND-2, a Quinoline Derivative, Inhibits the Proliferation of Prostate Cancer Cells by Inducing Oxidative Stress, Apoptosis and Inhibiting Topoisomerase II. LIFE (BASEL, SWITZERLAND) 2022; 12:life12111879. [PMID: 36431014 PMCID: PMC9693996 DOI: 10.3390/life12111879] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022]
Abstract
In men, prostate cancer (PC) is the most frequently diagnosed cancer, causing an estimated 375,000 deaths globally. Currently, existing therapies for the treatment of PC, notably metastatic cases, have limited efficacy due to drug resistance and problematic adverse effects. Therefore, it is imperative to discover and develop novel drugs for treating PC that are efficacious and do not produce intolerable adverse or toxic effects. Condensed quinolines are naturally occurring anticancer compounds. In this study, we determined the in vitro efficacy of IND-2 (4-chloro-2-methylpyrimido[1″,2″:1,5]pyrazolo[3,4-b]quinolone) in the PC lines, PC-3 and DU-145. IND-2 significantly inhibited the proliferation of PC-3 and DU-145, with IC50 values of 3 µM and 3.5 µM, respectively. The incubation of PC-3 cells with 5 and 10 µM of IND-2 caused the loss of the mitochondrial membrane potential in PC-3 cells. Furthermore, IND-2, at 5 µM, increased the expression of cleaved caspase-3, cleaved caspase-7 and cleaved poly (ADP-ribose) polymerase (PARP). The incubation of PC-3 cells with 5 µM of IND-2 significantly decreased the expression of the apoptotic protein, B-cell lymphoma 2 (Bcl-2). Furthermore, 5 and 10 µM of IND-2 produced morphological changes in PC-3 cells characteristic of apoptosis. Interestingly, IND-2 (2.5, 5 and 10 µM) also induced mitotic catastrophe in PC-3 cells, characterized by the accumulation of multinuclei. The incubation of DU-145 cells with 1.25 and 5 μM of IND-2 significantly increased the levels of reactive oxygen species (ROS). Finally, IND-2, at 10 μM, inhibited the catalytic activity of topoisomerase IIα. Overall, our findings suggest that IND-2 could be a potential lead compound for the development of more efficacious compounds for the treatment of PC.
Collapse
Affiliation(s)
- Swapnaa Balaji
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Rabin Neupane
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Saloni Malla
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Rahul Khupse
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Findlay, Findlay, OH 43551, USA
| | - Haneen Amawi
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
- Department of Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, P.O. Box 566, Irbid 21163, Jordan
| | - Shikha Kumari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Diwakar Bastihalli Tukaramrao
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Srestha Chattopadhyay
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Charles R. Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy & Pharmaceutical Sciences, St. John’s University, New York, NY 11432, USA
| | - Sai H. S. Boddu
- College of Pharmacy and Health Sciences, Ajman University, Ajman P.O. Box 346, United Arab Emirates
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Chandrabose Karthikeyan
- Department of Pharmacy, Indira Gandhi National Tribal University, Lalpur, Amarkantak 484887, Madhya Pradesh, India
| | - Piyush Trivedi
- Center for Innovation and Translational Research, Poona College of Pharmacy, Bharati Vidyapeeth, Pune 411038, Maharashtra, India
| | - Dayanidhi Raman
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Amit K. Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
- Correspondence: ; Tel.: +1-419-383-1913
| |
Collapse
|
7
|
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: 30] [Impact Index Per Article: 15.0] [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
|
8
|
Amawi H, Aljabali AAA, Boddu SHS, Amawi S, Obeid MA, Ashby CR, Tiwari AK. The use of zebrafish model in prostate cancer therapeutic development and discovery. Cancer Chemother Pharmacol 2021; 87:311-325. [PMID: 33392639 DOI: 10.1007/s00280-020-04211-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/26/2020] [Indexed: 12/24/2022]
Abstract
Zebrafish is now among the leading in vivo model for cancer research, including prostate cancer. They are an alternative economic model being used to study cancer development, proliferation, and metastasis. They can also be effectively utilized for the development of cancer drugs at all levels, including target validation, and high-throughput screening for possible lead molecules. In this review, we provide a comprehensive overview of the role of zebrafish as an in vivo model in prostate cancer research. Globally, prostate cancer is a leading cause of death in men. Although many molecular mechanisms have been identified as playing a role in the pathogenesis of prostate cancer, there is still a significant need to understand the initial events of the disease. Furthermore, current treatments are limited by the emergence of severe toxicities and multidrug resistance. There is an essential need for economical and relevant research tools to improve our understanding and overcome these problems. This review provides a comprehensive summary of studies that utilized zebrafish for different aims in prostate cancer research. We discuss the use of zebrafish in prostate cancer cell proliferation and metastasis, defining signaling pathways, drug discovery and therapeutic development against prostate cancer, and toxicity studies. Finally, this review highlights limitations in this field and future directions to efficiently use zebrafish as a robust model for prostate cancer therapeutics development.
Collapse
Affiliation(s)
- Haneen Amawi
- Department of Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, P.O.BOX 566, Irbid, 21163, Jordan.
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Sai H S Boddu
- College of Pharmacy and Health Sciences, Ajman University, Ajman, UAE
| | - Sadam Amawi
- Department of Urology and General Surgery, Faculty of Medicine, King Abdullah University Hospital, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Mohammad A Obeid
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, St. John's University, Queens, USA
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, The University of Toledo, Toledo, OH, USA.
| |
Collapse
|
9
|
Namdeo AG, Boddu SHS, Amawi H, Ashby CR, Tukaramrao DB, Trivedi P, Babu RJ, Tiwari AK. Flavonoids as Multi-Target Compounds: A Special Emphasis on their Potential as Chemo-adjuvants in Cancer Therapy. Curr Pharm Des 2020; 26:1712-1728. [PMID: 32003663 DOI: 10.2174/1381612826666200128095248] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 01/24/2020] [Indexed: 02/06/2023]
Abstract
Flavonoids are low molecular weight, polyphenolic phytochemicals, obtained from secondary metabolism of various plant compounds. They have a spectrum of pharmacological efficacies, including potential anticancer efficacy. Natural flavonoids are present in fruits, vegetables, grains, bark, roots, stems, flowers, tea and wine. Flavonoids can attenuate or inhibit the initiation, promotion and progression of cancer by modulating various enzymes and receptors in diverse pathways that involve cellular proliferation, differentiation, apoptosis, inflammation, angiogenesis and metastasis. Furthermore, in vitro, flavonoids have been shown to reverse multidrug resistance when used as chemo-adjuvants. Flavonoids (both natural and synthetic analogues) interact with several oncogenic targets through dependent and independent mechanisms to mediate their anticancer efficacy in different types of cancer cells.
Collapse
Affiliation(s)
- Ajay G Namdeo
- Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Pune, India
| | - Sai H S Boddu
- College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
| | - Haneen Amawi
- Department of Pharmacy practice, Faculty of Pharmacy, Yarmouk University, P.O. BOX 566, Irbid 21163, Jordan
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY 11439, United States
| | - Diwakar B Tukaramrao
- Department of Pharmacology and Experimental Therapeutics, The University of Toledo, Toledo, OH 43606, United States
| | - Piyush Trivedi
- Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Pune, India
| | - R Jayachandra Babu
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, United States
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, The University of Toledo, Toledo, OH 43606, United States
| |
Collapse
|
10
|
Tuli HS, Mittal S, Aggarwal D, Parashar G, Parashar NC, Upadhyay SK, Barwal TS, Jain A, Kaur G, Savla R, Sak K, Kumar M, Varol M, Iqubal A, Sharma AK. Path of Silibinin from diet to medicine: A dietary polyphenolic flavonoid having potential anti-cancer therapeutic significance. Semin Cancer Biol 2020; 73:196-218. [PMID: 33130037 DOI: 10.1016/j.semcancer.2020.09.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/11/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023]
Abstract
In the last few decades, targeting cancer by the use of dietary phytochemicals has gained enormous attention. The plausible reason and believe or mind set behind this fact is attributed to either lesser or no side effects of natural compounds as compared to the modern chemotherapeutics, or due to their conventional use as dietary components by mankind for thousands of years. Silibinin is a naturally derived polyphenol (a flavonolignans), possess following biochemical features; molecular formula C25H22O10, Molar mass: 482.44 g/mol, Boiling point 793 °C, with strikingly high antioxidant and anti-tumorigenic properties. The anti-cancer properties of Silibinin are determined by a variety of cellular pathways which include induction of apoptosis, cell cycle arrest, inhibition of angiogenesis and metastasis. In addition, Silibinin controls modulation of the expression of aberrant miRNAs, inflammatory response, and synergism with existing anti-cancer drugs. Therefore, modulation of a vast array of cellular responses and homeostatic aspects makes Silibinin an attractive chemotherapeutic agent. However, like other polyphenols, the major hurdle to declare Silibinin a translational chemotherapeutic agent, is its lesser bioavailability. After summarizing the chemistry and metabolic aspects of Silibinin, this extensive review focuses on functional aspects governed by Silibinin in chemoprevention with an ultimate goal of summarizing the evidence supporting the chemopreventive potential of Silibinin and clinical trials that are currently ongoing, at a single platform.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India
| | - Sonam Mittal
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India
| | - Gaurav Parashar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India
| | | | - Sushil Kumar Upadhyay
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India
| | - Tushar Singh Barwal
- Department of Zoology, Central University of Punjab, Bathinda, 151 001, Punjab, India
| | - Aklank Jain
- Department of Zoology, Central University of Punjab, Bathinda, 151 001, Punjab, India
| | - Ginpreet Kaur
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's, NMIMS, Mumbai, 400 056, Maharastra, India
| | - Raj Savla
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's, NMIMS, Mumbai, 400 056, Maharastra, India
| | | | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar University, Sadopur, India
| | - Mehmet Varol
- Department of Molecular Biology and Genetics, Faculty of Science, Mugla Sitki Kocman University, Mugla, TR48000, Turkey
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research (Formerly Faculty of Pharmacy), Jamia Hamdard (Deemed to be University), Delhi, India
| | - Anil Kumar Sharma
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India.
| |
Collapse
|
11
|
Evaluation of Cytotoxicity and Taste-Masking Effect of Selected Flavors on Dental Lidocaine HCl Injection. Pharmaceuticals (Basel) 2020; 13:ph13110353. [PMID: 33138130 PMCID: PMC7693877 DOI: 10.3390/ph13110353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/26/2020] [Accepted: 10/26/2020] [Indexed: 01/04/2023] Open
Abstract
AIM Anxiety and intolerance to dental local anesthetic injections are common in patients undergoing dental procedures. This work was designed to study cytotoxicity of selected flavors in primary gingival keratinocytes (PGK), to acquire information on their suitability for use in dental lidocaine hydrochloride (LID) injection. We also evaluated the bio-mimetic taste of LID dental injection in the presence of selected flavors and sweetener using an Astree electronic tongue (ETongue). METHODS The cytotoxicity of chocolate natural and artificial flavor (CTE), raspberry flavor artificial (RAS), cherry flavor (CHR), bitterness suppressor flavor (BSF) and lemon flavor extract (LFE) at various dilutions (0.16-10% v/v) was carried out in PGK using the live cell morphological analysis and MTT cell cytotoxicity assay. Based on the cytotoxicity data, CTE and RAS were added to Xylocaine® (2%) along with 0.09% sodium saccharin and taste was assessed using an ETongue. RESULTS After three hours of treatment, a dose-dependent cell death was induced by all flavors compared to the untreated control. BSF was found to be more toxic when compared to other flavors. CTE was found to be less toxic. The mean IC50 values of CTE, RAS, CHR, BSF and LFE in PGK were found to be 9.54, 8.43, 2.21, 0.38 and 4.01 mg/mL. Taste analysis with the ETongue showed a clear taste difference between the control and test formulations containing CTE and RAS flavors along with sodium saccharin. CONCLUSION CTE and RAS flavors in combination with 0.09% sodium saccharin can achieve a significant taste-masking effect in the dental LID injection.
Collapse
|
12
|
Novel Chrysin-De-Allyl PAC-1 Hybrid Analogues as Anticancer Compounds: Design, Synthesis, and Biological Evaluation. Molecules 2020; 25:molecules25133063. [PMID: 32635530 PMCID: PMC7412250 DOI: 10.3390/molecules25133063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/16/2022] Open
Abstract
New chrysin-De-allyl-Pac-1 hybrid analogues, tethered with variable heterocyclic systems (4a–4o), were rationally designed and synthesized. The target compounds were screened for in vitro antiproliferative efficacy in the triple-negative breast cancer (TNBC) cell line, MDA-MB-231, and normal human mammary epithelial cells (HMECs). Two compounds, 4g and 4i, had the highest efficacy and selectivity towards MDA-MB-231 cells, and thus, were further evaluated by mechanistic experiments. The results indicated that both compounds 4g and 4i induced apoptosis by (1) inducing cell cycle arrest at the G2 phase in MDA-MB-231 cells, and (2) activating the intrinsic apoptotic pathways in a concentration-dependent manner. Physicochemical characterizations of these compounds suggested that they can be further optimized as potential anticancer compounds for TNBC cells. Overall, our results suggest that 4g and 4i could be suitable leads for developing novel compounds to treat TNBC.
Collapse
|
13
|
Hosseinabadi T, Lorigooini Z, Tabarzad M, Salehi B, Rodrigues CF, Martins N, Sharifi-Rad J. Silymarin antiproliferative and apoptotic effects: Insights into its clinical impact in various types of cancer. Phytother Res 2019; 33:2849-2861. [PMID: 31407422 DOI: 10.1002/ptr.6470] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 06/07/2019] [Accepted: 07/13/2019] [Indexed: 12/11/2022]
Abstract
Silymarin is a complex extract isolated from the plant Silybum marianum, widely known for its prominent antioxidant and hepatoprotective effects, although increasing evidences have reported extraordinary antiproliferative and apoptotic abilities. As a result, several signaling pathways involved in cell cycle control, cell proliferation, and cell death have been deconvoluted as critical mechanisms. In this regard, cyclin and cyclin-dependent pathways have been the most studied ones. Following that, apoptotic pathways, such as p53, Akt, STAT-3, Ras, and caspases pathways, have been extensively studied, although other mechanisms involved in inflammation and angiogenesis have also been highlighted as silymarin-likely targets in cancer therapy. Therefore, the main challenge of this review is to discuss the diverse molecular mechanisms for silymarin antiproliferative and apoptotic effects; most of them largely studied in various types of cancers so far. Clinical trials and combination therapies related to silymarin application in cancer prevention and treatment are presented as well.
Collapse
Affiliation(s)
- Tahereh Hosseinabadi
- Department of Pharmacognosy and Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Lorigooini
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Maryam Tabarzad
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahare Salehi
- Student Research Committee, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| | - Célia F Rodrigues
- LEPABE-Department of Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Natália Martins
- Faculty of Medicine, University of Porto, Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal
| | - Javad Sharifi-Rad
- Zabol Medicinal Plants Research Center, Zabol University of Medical Sciences, Zabol, Iran
| |
Collapse
|
14
|
Novel Thienopyrimidine Derivative, RP-010, Induces β-Catenin Fragmentation and Is Efficacious against Prostate Cancer Cells. Cancers (Basel) 2019; 11:cancers11050711. [PMID: 31126091 PMCID: PMC6563099 DOI: 10.3390/cancers11050711] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 12/31/2022] Open
Abstract
Thienopyrimidines containing a thiophene ring fused to pyrimidine are reported to have a wide-spectrum of anticancer efficacy in vitro. Here, we report for the first time that thieno[3,2-d]pyrimidine-based compounds, also known as the RP series, have efficacy in prostate cancer cells. The compound RP-010 was efficacious against both PC-3 and DU145 prostate cancer (PC) cells (IC50 < 1 µM). The cytotoxicity of RP-010 was significantly lower in non-PC, CHO, and CRL-1459 cell lines. RP-010 (0.5, 1, 2, and 4 µM) arrested prostate cancer cells in G2 phase of the cell cycle, and induced mitotic catastrophe and apoptosis in both PC cell lines. Mechanistic studies suggested that RP-010 (1 and 2 µM) affected the wingless-type MMTV (Wnt)/β-catenin signaling pathway, in association with β-catenin fragmentation, while also downregulating important proteins in the pathway, including LRP-6, DVL3, and c-Myc. Interestingly, RP-010 (1 and 2 µM) induced nuclear translocation of the negative feedback proteins, Naked 1 and Naked 2, in the Wnt pathway. In addition, RP-010 (0.5, 1, 2 and 4 µM) significantly decreased the migration of PC cells in vitro. Finally, RP-010 did not produce significant toxic effects in zebrafish at concentrations of up to 6 µM. In conclusion, RP-010 may be an efficacious and relatively nontoxic anticancer compound for prostate cancer. Future mechanistic and in vivo efficacy studies are needed to optimize the hit compound RP-010 for lead optimization and clinical use.
Collapse
|
15
|
Song Y, Ye M, Zhou J, Wang ZW, Zhu X. Restoring E-cadherin Expression by Natural Compounds for Anticancer Therapies in Genital and Urinary Cancers. MOLECULAR THERAPY-ONCOLYTICS 2019; 14:130-138. [PMID: 31194121 PMCID: PMC6551504 DOI: 10.1016/j.omto.2019.04.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
E-cadherin plays a pivotal role in cancer progression, including the epithelial-mesenchymal transition (EMT) process and tumor metastasis. Loss of E-cadherin contributes to enhanced invasion and metastasis in human cancers. Therefore, restoring E-cadherin could be a potential approach for cancer therapy. Multiple natural compounds have been shown to possess anti-tumor activities through the regulation of key molecules in signaling pathways, including E-cadherin. In this review, we describe the numerous compounds that restore the expression of E-cadherin in genital and urinary malignancies. We further discuss the potential anti-tumor molecular mechanisms of these agents as the activators of E-cadherin in genital and urinary cancers. Although these compounds exhibit their potential to inhibit the development and progression of cancers, there are several challenges to developing them as therapeutic drugs for cancer patients. Poor bioavailability in vivo is the main disadvantage of these compounds. Modification of compound structures has produced actual improvements in bioavailability. Nanoparticle-based delivery systems could be useful to deliver the agents to targeted organs. These compounds could be new promising therapeutic agents for the treatment of human genital and urinary cancers. Further investigations are required to determine the safety and side effects of natural compounds using animal models prior to clinical trials.
Collapse
Affiliation(s)
- Yizuo Song
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Miaomiao Ye
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Junhan Zhou
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhi-Wei Wang
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.,Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Xueqiong Zhu
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| |
Collapse
|
16
|
Karthikeyan C, Amawi H, Ashby CR, Khare VM, Jones V, Hari Narayana Moorthy NS, Trivedi P, Tiwari AK. Novel 3-((2-chloroquinolin-3-yl)methylene)indolin-2-one derivatives produce anticancer efficacy in ovarian cancer in vitro. Heliyon 2019; 5:e01603. [PMID: 31193218 PMCID: PMC6522656 DOI: 10.1016/j.heliyon.2019.e01603] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 02/24/2019] [Accepted: 04/26/2019] [Indexed: 12/24/2022] Open
Abstract
A novel series of 3-((2-chloroquinolin-3-yl)methylene)indolin-2-ones were synthesized, using the ‘molecular hybridization approach’ and evaluated for anticancer efficacy. Eleven 3-((2-chloroquinolin-3-yl)methylene)indolin-2-ones (LM01 to LM11) were synthesized and evaluated for in vitro cytotoxic efficacy in cancer (ovarian, prostate and colon) and two non-cancerous cell lines. Among the 3-((2-chloroquinolin-3-yl)methylene)indolin-2-one derivatives, LM08, with a 6-Cl substitution in the 3-quinolinyl moiety, had selective and potent cytotoxic efficacy in the ovarian cancer cell line A2780. Further mechanistic investigations indicated that LM08 significantly inhibited the clonogenic survival of A2780 cancer cells, which was mediated by inducing apoptosis.
Collapse
Affiliation(s)
- Chandrabose Karthikeyan
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, 484887, India.,School of Pharmaceutical Sciences, Rajiv Gandhi Proudyogiki Vishwavidyalaya, Bhopal, 462036, India
| | - Haneen Amawi
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, OH, USA.,Department of Pharmacy Practice, Yarmouk University, Irbid, 21163, Jordan
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, St. John's University, Queens, USA
| | - Vishwa M Khare
- Cell and Developmental Biology, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Veronica Jones
- Department of Biomedical Sciences, Tuskegee University, Tuskegee, AL, USA
| | | | - Piyush Trivedi
- School of Pharmaceutical Sciences, Rajiv Gandhi Proudyogiki Vishwavidyalaya, Bhopal, 462036, India.,Center of Innovation and Translational Research, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Pune, India
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, OH, USA.,Department of Biomedical Sciences, Tuskegee University, Tuskegee, AL, USA
| |
Collapse
|
17
|
Letrado P, de Miguel I, Lamberto I, Díez-Martínez R, Oyarzabal J. Zebrafish: Speeding Up the Cancer Drug Discovery Process. Cancer Res 2018; 78:6048-6058. [PMID: 30327381 DOI: 10.1158/0008-5472.can-18-1029] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/29/2018] [Accepted: 08/23/2018] [Indexed: 11/16/2022]
Abstract
Zebrafish (Danio rerio) is an ideal in vivo model to study a wide variety of human cancer types. In this review, we provide a comprehensive overview of zebrafish in the cancer drug discovery process, from (i) approaches to induce malignant tumors, (ii) techniques to monitor cancer progression, and (iii) strategies for compound administration to (iv) a compilation of the 355 existing case studies showing the impact of zebrafish models on cancer drug discovery, which cover a broad scope of scenarios. Finally, based on the current state-of-the-art analysis, this review presents some highlights about future directions using zebrafish in cancer drug discovery and the potential of this model as a prognostic tool in prospective clinical studies. Cancer Res; 78(21); 6048-58. ©2018 AACR.
Collapse
Affiliation(s)
- Patricia Letrado
- Ikan Biotech SL, The Zebrafish Lab Department, Centro Europeo de Empresas e Innovación de Navarra (CEIN), Noain, Spain.,Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Irene de Miguel
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Iranzu Lamberto
- Ikan Biotech SL, The Zebrafish Lab Department, Centro Europeo de Empresas e Innovación de Navarra (CEIN), Noain, Spain
| | - Roberto Díez-Martínez
- Ikan Biotech SL, The Zebrafish Lab Department, Centro Europeo de Empresas e Innovación de Navarra (CEIN), Noain, Spain.
| | - Julen Oyarzabal
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.
| |
Collapse
|
18
|
Jahanafrooz Z, Stallinger A, Anders I, Kleinegger F, Lohberger B, Durchschein C, Bauer R, Deutsch A, Rinner B, Kretschmer N. Influence of silibinin and β-β-dimethylacrylshikonin on chordoma cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 49:32-40. [PMID: 30217260 DOI: 10.1016/j.phymed.2018.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 05/17/2018] [Accepted: 06/07/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Chordoma, slow growing bone tumours originating from remnants of the notochord, leave affected patients with a median survival of six years. The high recurrence rate of chordoma, together with limited treatment options and bad overall prognosis, make the development of new treatment options urgently necessary. PURPOSE In this study, the potential of two natural products, silibinin and β-β-dimethylacrylshikonin (DMAS), was tested on clival (MUG-CC1 and UM-Chor1) as well as sacral (MUG-Chor1 and U-CH2) chordoma cell lines. The treatment was administered both as single- and combined therapy. METHODS For investigation of cell viability, the Cell Titer 96 Aqueous Non-Radioactive Cell Proliferation Assay Kit was used. Apoptosis induction was studied by flow cytometry, (Annexin V/SYTOX Green, caspase-3) and RT-qPCR. Pathway analyses were performed by western blot. RESULTS Both drugs were found to reduce cell viability alone as well as in combination in a dose dependent manner, with DMAS being more efficient than silibinin. The mode of cell death was mainly apoptosis in DMAS treated samples, while the combination therapy led to apoptosis as well as late-apoptosis/necrosis. Silibinin therapy alone, although reducing cell viability, did not lead to significant apoptotic effects in the performed assays. Focussing on the molecular mechanism of DMAS induced apoptosis, it was found that major genes of the mitochondrial apoptosis pathway, like NOXA and PUMA were overexpressed. Additionally, western blot experiments showed a decrease of ERK/pERK, STAT3/pSTAT3 (Tyr705) and AKT/pAKT expression/activation levels under DMAS treatment. CONCLUSION DMAS is a promising new candidate for chordoma therapy, while silibinin or a combination of both is less favourable.
Collapse
Affiliation(s)
- Zohreh Jahanafrooz
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biotechnology, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | | | - Ines Anders
- Division of Biomedical Research, Medical University Graz, Graz, Austria
| | | | - Birgit Lohberger
- Department of Orthopedics and Trauma, Medical University Graz, Graz, Austria
| | - Christin Durchschein
- Department of Pharmacognosy, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Rudolf Bauer
- Department of Pharmacognosy, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Alexander Deutsch
- CBmed Center for Biomarker Research in Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Beate Rinner
- Division of Biomedical Research, Medical University Graz, Graz, Austria.
| | - Nadine Kretschmer
- Department of Pharmacognosy, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| |
Collapse
|
19
|
Hussein N, Ashby CR, Amawi H, Nyinawabera A, Vij A, Khare VM, Karthikeyan C, Tiwari AK. Cariprazine, A Dopamine D₂/D₃ Receptor Partial Agonist, Modulates ABCG2-Mediated Multidrug Resistance in Cancer. Cancers (Basel) 2018; 10:E308. [PMID: 30181510 PMCID: PMC6162716 DOI: 10.3390/cancers10090308] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/13/2018] [Accepted: 08/27/2018] [Indexed: 12/20/2022] Open
Abstract
Multidrug resistance (MDR) is a continuing clinical problem that limits the efficacy of chemotherapy in cancer. The over expression of the ATP-binding cassette (ABC) family G2 (ABCG2) transporter is one of the main mechanisms that mediates MDR in cancer. Molecular modeling data indicated that cariprazine, a dopamine D₂/D₃ receptor partial agonist, had a significant binding affinity for ABCG2 transporter with a Glide XP score of -6.515. Therefore, in this in vitro study, we determined the effect of cariprazine on MDR resulting from the overexpression of ABCG2 transporters. Alone, cariprazine, at concentrations up to 20 μM, did not significantly decrease cell viability. Cariprazine, at concentrations ranging from 1 to 10 μM, did not significantly alter the cytotoxicity of mitoxantrone (MX) in the parental non-small cell cancer cell line, H460 and colon cancer cell S1. However, cariprazine (1⁻20 μM) significantly enhanced the efficacy of ABCG2 substrate antineoplastic drug MX in the ABCG2-overexpressing MDR cell line, H460-MX20 and S1M1-80, by reducing the resistance fold from 28 to 1 and from 93 to 1.33, respectively. Cariprazine, in a concentration-dependent (1⁻20 μM), significantly increased the intracellular accumulation of Rhodamine 123 in S1M1-80. Interestingly, 10 or 20 μM of cariprazine significantly decreased the expression levels of the ABCG2 protein in the colon and lung cancer cell lines, suggesting that cariprazine inhibits both the function and expression of ABCG2 transporters at nontoxic concentrations. Overall, our results suggest that cariprazine, via several distinct mechanisms, can resensitize resistant cancer cells to mitoxantrone.
Collapse
Affiliation(s)
- Noor Hussein
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA.
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy, St. John's University, Queens, NY 11439, USA.
| | - Haneen Amawi
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA.
| | - Angelique Nyinawabera
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA.
| | - Atul Vij
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA.
| | - Vishwa M Khare
- Cell and Developmental Biology, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Chandrabose Karthikeyan
- Department of Pharmacy, Indira Gandhi National Tribal University, Lalpur, Amarkantak, MP 484887, India.
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA.
| |
Collapse
|