1
|
Glomb T, Środa-Pomianek K, Palko-Łabuz A, Wesołowska O, Wikiera A, Wojtkowiak K, Jezierska A, Kochel A, Lesyk R, Świątek P. New Hydrazone Derivatives Based on Pyrazolopyridothiazine Core as Cytotoxic Agents to Colon Cancers: Design, Synthesis, Biological Evaluation, and Molecular Modeling. ChemMedChem 2025; 20:e202400687. [PMID: 39786320 DOI: 10.1002/cmdc.202400687] [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: 09/02/2024] [Revised: 12/18/2024] [Accepted: 01/08/2025] [Indexed: 01/12/2025]
Abstract
In this research, a series of novel hydrazone derivatives based on pyrazolopyridothiazinylacetohydrazide were designed, synthesized, and evaluated for their in vitro cytotoxic potency on several human colon cancer cells (HTC116, HT-29, and LoVo). After MTT and SRB assays four of the most active derivatives: hydrazide GH and hydrazones GH7, GH8, and GH11, were chosen for further investigation. Hydrazone GH11 had the highest cytotoxic activity (IC50 values of c.a. 0.5 μM). Additionally, the impact of novel derivatives on the oxidative stress level, apoptosis induction, and modulation of inflammation in colon cancer cells was examined. In all studies, the activity of the derivatives increased in order GH < GH7 < GH8 < GH11. At the same time, most of the research was conducted on compounds combined with apple pectin (PC). The most interesting observation was that all the studied derivatives applied together with PC showed significantly higher activity than observed in the case of using PC, hydrazide, or hydrazones separately. Finally, computational chemistry methods (molecular modeling and Density Functional Theory - DFT) were used to complement the experimental studies.
Collapse
Affiliation(s)
- Teresa Glomb
- Department of Medicinal Chemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556, Wroclaw, Poland
| | - Kamila Środa-Pomianek
- Department of Biophysics and Neuroscience, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 3 A, 50-368, Wroclaw, Poland
| | - Anna Palko-Łabuz
- Department of Biophysics and Neuroscience, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 3 A, 50-368, Wroclaw, Poland
| | - Olga Wesołowska
- Department of Biophysics and Neuroscience, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 3 A, 50-368, Wroclaw, Poland
| | - Agnieszka Wikiera
- Department of Medical Physiology, Faculty of Health Sciences, Jagiellonian University Medical College, Michałowskiego Kraków, 12, 31-126, Krakow, Poland
| | - Kamil Wojtkowiak
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383, Wroclaw, Poland
| | - Aneta Jezierska
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383, Wroclaw, Poland
| | - Andrzej Kochel
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383, Wroclaw, Poland
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79-010, Lviv, Ukraine
| | - Piotr Świątek
- Department of Medicinal Chemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556, Wroclaw, Poland
| |
Collapse
|
2
|
Andersson CR, Ye J, Blom K, Fryknäs M, Larsson R, Nygren P. Assessment in vitro of interactions between anti-cancer drugs and noncancer drugs commonly used by cancer patients. Anticancer Drugs 2023; 34:92-102. [PMID: 36066384 PMCID: PMC9760465 DOI: 10.1097/cad.0000000000001344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 03/11/2022] [Indexed: 11/02/2022]
Abstract
Cancer patients often suffer from cancer symptoms, treatment complications and concomitant diseases and are, therefore, often treated with several drugs in addition to anticancer drugs. Whether such drugs, here denoted as 'concomitant drugs', have anticancer effects or interact at the tumor cell level with the anticancer drugs is not very well known. The cytotoxic effects of nine concomitant drugs and their interactions with five anti-cancer drugs commonly used for the treatment of colorectal cancer were screened over broad ranges of drug concentrations in vitro in the human colon cancer cell line HCT116wt. Seven additional tyrosine kinase inhibitors were included to further evaluate key findings as were primary cultures of tumor cells from patients with colorectal cancer. Cytotoxic effects were evaluated using the fluorometric microculture cytotoxicity assay (FMCA) and interaction analysis was based on Bliss independent interaction analysis. Simvastatin and loperamide, included here as an opioid agonists, were found to have cytotoxic effects on their own at reasonably low concentrations whereas betamethasone, enalapril, ibuprofen, metformin, metoclopramide, metoprolol and paracetamol were inactive also at very high concentrations. Drug interactions ranged from antagonistic to synergistic over the concentrations tested with a more homogenous pattern of synergy between simvastatin and protein kinase inhibitors in HCT116wt cells. Commonly used concomitant drugs are mostly neither expected to have anticancer effects nor to interact significantly with anticancer drugs frequently used for the treatment of colorectal cancer.
Collapse
Affiliation(s)
| | - Jiawei Ye
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Medical Laboratory Sciences, School of Medicine, Southeast University, Nanjing, China
| | - Kristin Blom
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Mårten Fryknäs
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Rolf Larsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Peter Nygren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
3
|
Beton K, Brożek-Płuska B. Biochemistry and Nanomechanical Properties of Human Colon Cells upon Simvastatin, Lovastatin, and Mevastatin Supplementations: Raman Imaging and AFM Studies. J Phys Chem B 2022; 126:7088-7103. [PMID: 36083294 PMCID: PMC9511485 DOI: 10.1021/acs.jpcb.2c03724] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
One of the most important areas of medical science is
oncology,
which is responsible for both the diagnostics and treatment of cancer
diseases. Over the years, there has been an intensive development
of cancer diagnostics and treatment. This paper shows the comparison
of normal (CCD-18Co) and cancerous (CaCo-2) cell lines of the human
gastrointestinal tract on the basis of nanomechanical and biochemical
properties to obtain information on cancer biomarkers useful in oncological
diagnostics. The research techniques used were Raman spectroscopy
and imaging and atomic force microscopy (AFM). In addition, the studies
also included the effect of the statin compounds—mevastatin,
lovastatin, and simvastatin—and their influence on biochemical
and nanomechanical changes of cell properties using Raman imaging
and AFM techniques. The cytotoxicity of statins was determined using
XTT tests.
Collapse
Affiliation(s)
- Karolina Beton
- Laboratory of Laser Molecular Spectroscopy, Institute of Applied Radiation Chemistry, Lodz University of Technology, Wroblewskiego 15, 93-590 Lodz, Poland
| | - Beata Brożek-Płuska
- Laboratory of Laser Molecular Spectroscopy, Institute of Applied Radiation Chemistry, Lodz University of Technology, Wroblewskiego 15, 93-590 Lodz, Poland
| |
Collapse
|
4
|
Duarte D, Vale N. Combining repurposed drugs to treat colorectal cancer. Drug Discov Today 2021; 27:165-184. [PMID: 34592446 DOI: 10.1016/j.drudis.2021.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 07/19/2021] [Accepted: 09/22/2021] [Indexed: 02/08/2023]
Abstract
The drug development process, especially of antineoplastic agents, has become increasingly costly and ineffective. Drug repurposing and drug combination are alternatives to de novo drug development, being low cost, rapid, and easy to apply. These strategies allow higher efficacy, decreased toxicity, and overcoming of drug resistance. The combination of antineoplastic agents is already being applied in cancer therapy, but the combination of repurposed drugs is still under-explored in pre- and clinical development. In this review, we provide a set of pharmacological concepts focusing on drug repurposing for treating colorectal cancer (CRC) and that are relevant for the application of new drug combinations against this disease.
Collapse
Affiliation(s)
- Diana Duarte
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal; Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal; Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
| |
Collapse
|
5
|
The potential use of simvastatin for cancer treatment: A review. Biomed Pharmacother 2021; 141:111858. [PMID: 34323700 DOI: 10.1016/j.biopha.2021.111858] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/17/2021] [Accepted: 06/28/2021] [Indexed: 12/21/2022] Open
Abstract
Statins, typically used to reduce lipid levels, have been rediscovered for exhibiting anticancer activities. Among them, especially simvastatin may influence the proliferation, migration, and survival of cancer cells. The concept of using statins to treat cancer has been adopted since the 1990s In vitro and in vivo experiments and cohort studies using statins have been carried out to demonstrate their antitumor effects (such as proliferation and migration impairment) by influencing inflammatory and oxidative stress-related tumorigenesis. Nevertheless, the biological mechanisms for these actions are not fully elucidated. In this review, we present an overview of the most important studies conducted from 2015 to date on the use of simvastatin in cancer therapy. This review brings the most recent perspectives and targets in epidemiological, in vitro, and in vivo studies, regarding the use of simvastatin alone or in combination with other drugs for the treatment of various types of cancer.
Collapse
|
6
|
Upreti S, Pandey SC, Bisht I, Samant M. Evaluation of the target-specific therapeutic potential of herbal compounds for the treatment of cancer. Mol Divers 2021; 26:1823-1835. [PMID: 34240331 DOI: 10.1007/s11030-021-10271-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 07/02/2021] [Indexed: 11/25/2022]
Abstract
Cancer is among one of the most fatal diseases leading to millions of death around the globe. Chemotherapy is the most popular conventional approach for the treatment of cancer. However, this is usually associated with various side effects and puts the patients under extreme physical and mental stress. Besides, there are increasing concerns about drug resistance. Thus, to surmount these limitations, there is a need to explore some alternative treatments. Studies related to plant-derived compounds are crucial in the search for safer and more efficient treatments. Plants and their associated secondary metabolites have been a revolutionary approach in the field of cancer treatment, as they give answers to almost all the constraints faced by synthetic drugs. Various plants and associated secondary metabolites display a great prospective as cytotoxic anticancer agents due to their specific interference with validated drug targets, such as inhibitors of mitosis, topoisomerase I and II inhibitor, DNA interactive agent, protein kinase inhibitors, inhibitors of DNA synthesis. In this review, the therapeutic potential of various natural compounds and their derivatives are presented based on their molecular targets. These herbal compounds and their derivatives could provide a rich resource for novel anticancer drug development.
Collapse
Affiliation(s)
- Shobha Upreti
- Cell and Molecular Biology Laboratory, Department Of Zoology, Kumaun University, SSJ Campus, Almora, Uttarakhand, India
| | - Satish Chandra Pandey
- Cell and Molecular Biology Laboratory, Department Of Zoology, Kumaun University, SSJ Campus, Almora, Uttarakhand, India
| | - Ila Bisht
- Cell and Molecular Biology Laboratory, Department Of Zoology, Kumaun University, SSJ Campus, Almora, Uttarakhand, India
| | - Mukesh Samant
- Cell and Molecular Biology Laboratory, Department Of Zoology, Kumaun University, SSJ Campus, Almora, Uttarakhand, India.
| |
Collapse
|
7
|
Taymouri S, Ahmadi Z, Mirian M, Tavakoli N. Simvastatin nanosuspensions prepared using a combination of pH-sensitive and timed-release approaches for potential treatment of colorectal cancer. Pharm Dev Technol 2021; 26:335-348. [PMID: 33430677 DOI: 10.1080/10837450.2021.1872086] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
A dual pH- and time-dependent polymeric coated capsule was developed to achieve the site specificity of simvastatin (SIM) release in the colon. To improve the SIM solubility, soluplus-based nanosuspension of the drug were prepared by applying the anti-solvent crystallization technique; this was then followed by lyophilization. Particle size, polydispersity index, and saturation solubility were evaluated. The optimized nanosuspension was combined with SLS and freeze-dried before filling into hard gelatin capsules. Drug release characteristics of the coated capsules were studied in HCl 0.1 N, the phosphate buffers 6.8 and 7.4, and the simulated colonic fluid (pH 6.8). The in-vitro cytotoxic effects of SIM nanoparticles against HT29 cells were then evaluated using the MTT assay. The prepared nanoparticles were spherical with a mean size of 261.66 nm, the zeta potential of -18.20 and the dissolution efficiency of 59.71%. X-ray diffraction and differential scanning calorimetry studies showed that the nanosizing technique transformed the crystalline drug into the more soluble amorphous form. The coated capsules had no release in the gastric media, providing the specific delivery of SIM in the colon. The cytotoxic effect of the SIM nanoparticles was significantly increased, as compared to the free SIM. The findings, therefore, showed that the coated capsules using the two polymers of ethyl cellulose and Eudragit S100 could be suitable for the colon target delivery of SIM.
Collapse
Affiliation(s)
- Somayeh Taymouri
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zoha Ahmadi
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Mirian
- Department of Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Naser Tavakoli
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
8
|
Antimicrobial Peptides as New Combination Agents in Cancer Therapeutics: A Promising Protocol against HT-29 Tumoral Spheroids. Int J Mol Sci 2020; 21:ijms21186964. [PMID: 32971958 PMCID: PMC7555805 DOI: 10.3390/ijms21186964] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/18/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022] Open
Abstract
Antimicrobial peptides are molecules synthetized by a large variety of organisms as an innate defense against pathogens. These natural compounds have been identified as promising alternatives to widely used molecules to treat infections and cancer cells. Antimicrobial peptides could be viewed as future chemotherapeutic alternatives, having the advantage of low propensity to drug resistance. In this study, we evaluated the efficiency of the antimicrobial peptide gramicidin A (GA) and the anticancer drug, doxorubicin (Doxo) against the spheroids from colorectal cancer cells (HT-29). The two drugs were applied separately against HT-29 spheroids as well as together to determine if they can act synergistically. The spheroid evolution, cell viability, and ATP levels were monitored at 24 and 48 h after the applied treatments. The results show significant drops in cell viability and cellular ATP levels for all the experimental treatments. The simultaneous use of the two compounds (GA and Doxo) seems to cause a synergistic effect against the spheroids.
Collapse
|
9
|
Fong W, To KKW. Drug repurposing to overcome resistance to various therapies for colorectal cancer. Cell Mol Life Sci 2019; 76:3383-3406. [PMID: 31087119 PMCID: PMC11105507 DOI: 10.1007/s00018-019-03134-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 04/06/2019] [Accepted: 05/06/2019] [Indexed: 02/06/2023]
Abstract
Emergence of novel treatment modalities provides effective therapeutic options, apart from conventional cytotoxic chemotherapy, to fight against colorectal cancer. Unfortunately, drug resistance remains a huge challenge in clinics, leading to invariable occurrence of disease progression after treatment initiation. While novel drug development is unfavorable in terms of time frame and costs, drug repurposing is one of the promising strategies to combat resistance. This approach refers to the application of clinically available drugs to treat a different disease. With the well-established safety profile and optimal dosing of these approved drugs, their combination with current cancer therapy is suggested to provide an economical, safe and efficacious approach to overcome drug resistance and prolong patient survival. Here, we review both preclinical and clinical efficacy, as well as cellular mechanisms, of some extensively studied repurposed drugs, including non-steroidal anti-inflammatory drugs, statins, metformin, chloroquine, disulfiram, niclosamide, zoledronic acid and angiotensin receptor blockers. The three major treatment modalities in the management of colorectal cancer, namely classical cytotoxic chemotherapy, molecular targeted therapy and immunotherapy, are covered in this review.
Collapse
Affiliation(s)
- Winnie Fong
- Faculty of Medicine, School of Pharmacy, Room 801N, Lo Kwee-Seong Integrated Biomedical Sciences Building, The Chinese University of Hong Kong, Area 39, Shatin, New Territories, Hong Kong SAR, China
| | - Kenneth K W To
- Faculty of Medicine, School of Pharmacy, Room 801N, Lo Kwee-Seong Integrated Biomedical Sciences Building, The Chinese University of Hong Kong, Area 39, Shatin, New Territories, Hong Kong SAR, China.
| |
Collapse
|
10
|
FOLFIRI-Mediated Toxicity in Human Aortic Smooth Muscle Cells and Possible Amelioration with Curcumin and Quercetin. Cardiovasc Toxicol 2019; 20:139-154. [DOI: 10.1007/s12012-019-09541-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
11
|
Tiwari H, Karki N, Pal M, Basak S, Verma RK, Bal R, Kandpal ND, Bisht G, Sahoo NG. Functionalized graphene oxide as a nanocarrier for dual drug delivery applications: The synergistic effect of quercetin and gefitinib against ovarian cancer cells. Colloids Surf B Biointerfaces 2019; 178:452-459. [DOI: 10.1016/j.colsurfb.2019.03.037] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 03/16/2019] [Accepted: 03/16/2019] [Indexed: 12/27/2022]
|
12
|
Shi X, Ran L, Liu Y, Zhong SH, Zhou PP, Liao MX, Fang W. Knockdown of hnRNP A2/B1 inhibits cell proliferation, invasion and cell cycle triggering apoptosis in cervical cancer via PI3K/AKT signaling pathway. Oncol Rep 2018; 39:939-950. [PMID: 29328485 PMCID: PMC5802035 DOI: 10.3892/or.2018.6195] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 12/29/2017] [Indexed: 01/18/2023] Open
Abstract
Cervical cancer is currently one of the major threats to women's health. The overexpression of heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNP A2/B1) as the biomarker has been investigated in various cancers. In our previous study, we found that lobaplatin induced apoptosis and cell cycle arrest via downregulation of proteins including hnRNP A2/B1 in cervical cancer cells. However, the underlying relationship between hnRNP A2/B1 and cervical cancer remained largely unknown. hnRNP A2/B1 knock-down in HeLa and CaSki cells was performed by shRNA transfection. The expression of hnRNP A2/B1 was detected by western blot and Quantitative Real-time PCR. Cell proliferation, migration, invasion and the IC50 of lobaplatin and irinotecan were determined by MTT assay, Transwell assay, Plate colony formation assay and wound healing assay. Flow cytometry was perfomed to investigate cell apoptosis and the cell cycle. The expression of PI3K, AKT, p-AKT, p21, p27, caspase-3, cleaved caspase-3 were revealed by western blot. Nude mouse xenograft model was undertaken with HeLa cells and the xenograft tumor tissue samples were analyzed for the expression of PCNA and Ki-67 by immunohistochemistry and the cell morphology was evaluated by hematoxylin and eosin (H&E). Results revealed that hnRNP A2/B1 was successfully silenced in HeLa and CaSki cells. hnRNP A2/B1 knock-down significantly induced the suppression of proliferation, migration, invasion and also enhancement of apoptosis and reduced the IC50 of lobaplatin and irinotecan. The expression of p21, p27 and cleaved caspase-3 in shRNA group were significantly upregulated and the expression of p-AKT was reduced both in vitro and in vivo. The results of immunohistochemistry showed that PCNA and Ki-67 were significantly downregulated in vivo. The growth of nude mouse xenograft tumor was significantly reduced by hnRNP A2/B1 knock-down. Taken together, these data indicate that inhibition of hnRNP A2/B1 in cervical cancer cells can inhibit cell proliferation and invasion, induce cell-cycle arrestment and trigger apoptosis via PI3K/AKT signaling pathway. In addition, after silencing hnRNP A2/B1 can increase the sensitivity of cervical cancer cells to lobaplatin and irinotecan.
Collapse
Affiliation(s)
- Xiang Shi
- Department of Biochemistry, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Li Ran
- Department of Mammary Gland and Gynecologic Oncology, Guizhou Cancer Hospital, Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Yao Liu
- Department of Biochemistry, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Shu-Huai Zhong
- Department of Biochemistry, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Ping-Ping Zhou
- Department of Biochemistry, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Ming-Xin Liao
- Department of Biochemistry, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Wen Fang
- Department of Biochemistry, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
13
|
Alqudah MAY, Mansour HT, Mhaidat N. Simvastatin enhances irinotecan-induced apoptosis in prostate cancer via inhibition of MCL-1. Saudi Pharm J 2017; 26:191-197. [PMID: 30166915 PMCID: PMC6111232 DOI: 10.1016/j.jsps.2017.12.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 12/12/2017] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer is one of the most common malignant tumors around the world. Hyperlipidemia is considered as one of the most important risk factors for the development of prostate cancer. Simvastatin is widely used for the treatment of hyperlipidemia and was previously shown to induce apoptosis in several cancer types including lung, colon, pancreas, breast, and prostate cancer. In this study we aimed to explore the potential role of simvastatin in enhancing irinotecan-induced apoptosis in prostate cancer cells. In addition, the underlying molecular mechanisms driving this potential effect of simvastatin were also explored. PC3 cells were treated with simvastatin, irinotecan or combination. Cell viability was assessed by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) colorimetric assay. Flow cytometry technique was used to analyze apoptosis and cell cycle progression. Western blot was used for detection of protein expression. Results showed that simvastatin has a significant anti-proliferative activity on PC3 cells. Combined treatment of simvastatin with irinotecan exhibited a significant inhibition of PC3 cell growth compared to each treatment alone. Flow cytometry analysis showed that PC3 cell treatment with simvastatin and irinotecan combination demonstrated a remarkable increase in the percentage of apoptotic cells and those accumulated at G0/G1 phase when compared to each treatment alone. Moreover, induction of apoptosis was caspase-independent. Western blot showed that apoptosis was accompanied by upregulation of GRP-78 level and downregulation of Mcl-1 levels in a time-dependent manner. The results of this study demonstrated that combined treatment of simvastatin with chemotherapeutic agents such as irinotecan resulted in enhancement of growth inhibition and induction of prostate cancer cell apoptosis.
Collapse
Affiliation(s)
- Mohammad A Y Alqudah
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Hebah T Mansour
- Department of Applied Biological Sciences, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Nizar Mhaidat
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| |
Collapse
|
14
|
Koi C, Izumi H, Kurita T, Nguyen TT, Murakami M, Yoshiura Y, Hachisuga T, Morimoto Y. Lovastatin induced Kruppel like factor 2 ( KLF2), Kruppel like factor 6 ( KLF6) and Ras homolog family member B ( RHOB) genes and preferentially led to viability reduction of Cisplatin-resistant cells. Oncotarget 2017; 8:106429-106442. [PMID: 29290960 PMCID: PMC5739745 DOI: 10.18632/oncotarget.22472] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 10/27/2017] [Indexed: 11/25/2022] Open
Abstract
It was reported that statins, inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase that are used to prevent hypercholesterolemia, have antitumor activity in several cancers. In this study, we investigated the cell viability of statins in Cisplatin-resistant HCP4 and PCDP5 cells compared with their parent Hela and PC3 cells, respectively, and found that HCP4 and PCDP5 cells were 37-fold and 18-fold more resistant to Cisplatin but 13-fold and 7-fold more sensitive to Lovastatin by cell proliferation assay. Lovastatin induced the apoptosis of HCP4 cells more rapidly and to greater extent than in Hela cells as assessed by flow cytometry and western blotting analyses. The MVA pathway was not involved in this acquired Cisplatin resistance. To elucidate the mechanism underlying the reduced viability to Lovastatin, we performed cDNA microarray analysis and identified 65 and 54 genes that were induced more than 2-fold by Lovastatin in HCP4 and PCDP5 cells, respectively. Of these, only three genes, KLF2, KLF6, and RHOB, were commonly induced between HCP4 and PCDP5 cells. These mRNAs were strongly induced by Lovastatin with transcriptional regulation in HCP4 cells. Consistent with transcription, the protein expression of RHOB also was induced by Lovastatin. The induction of these genes was associated with cell cycle arrest and apoptosis. Combination treatment with Cisplatin and Lovastatin resulted in an agonistic effect in Hela and PC3 cells and an antagonistic effect in HCP4 and PCDP5 cells. These results suggest that statins might have the potential to overcome Cisplatin resistance as single-agent therapy.
Collapse
Affiliation(s)
- Chiho Koi
- Department of Obstetrics and Gynecology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 805-8555, Japan
| | - Hiroto Izumi
- Department of Occupational Pneumology, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 805-8555, Japan
| | - Tomoko Kurita
- Department of Obstetrics and Gynecology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 805-8555, Japan
| | - Thuy Thi Nguyen
- Department of Obstetrics and Gynecology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 805-8555, Japan
| | - Midori Murakami
- Department of Obstetrics and Gynecology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 805-8555, Japan
| | - Yukiko Yoshiura
- Department of Occupational Pneumology, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 805-8555, Japan
| | - Toru Hachisuga
- Department of Obstetrics and Gynecology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 805-8555, Japan
| | - Yasuo Morimoto
- Department of Occupational Pneumology, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 805-8555, Japan
| |
Collapse
|
15
|
Keyvani-Ghamsari S, Rabbani-Chadegani A, Sargolzaei J, Shahhoseini M. Effect of irinotecan on HMGB1, MMP9 expression, cell cycle, and cell growth in breast cancer (MCF-7) cells. Tumour Biol 2017; 39:1010428317698354. [DOI: 10.1177/1010428317698354] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Irinotecan is a natural alkaloid agent widely used in cancer therapy. High-mobility group protein B1 as a non-histone chromosomal protein plays a fundamental role in gene expression and inflammation. In this study, the effect of irinotecan on high-mobility group protein B1 and MMP9 content, gene expression, cell cycle, and cell growth in human breast cancer cells (MCF-7) was investigated. The cells were exposed to various concentrations of irinotecan and the viability determined by trypan blue exclusion and 3-(4,5-dimethylthiazal-2-yl)-2,5-diphenyltetrazolium bromide assays. High-mobility group B proteins were extracted from the control and drug-treated cells and analyzed by immunoblot. High-mobility group protein B1 and MMP9 messenger RNA expression was studied by reverse transcription polymerase chain reaction. The results demonstrated reduction of cell viability upon increasing irinotecan concentration, up-regulated high-mobility group protein B1 gene expression, and down-regulated MMP9 mRNA. Although the content of high-mobility group protein B1 was decreased in chromatin extract upon drug action, no high-mobility group protein B1 release to extracellular space was detected by immunoblot analysis. Irinotecan decreased H3K9 acetylation and increased poly ADP-ribose polymerase fragmentation to 89 kDa and anion superoxide production suggesting induction of apoptosis in these cells. Propidium iodide staining of the cells 24 h after the drug treatment revealed arrest of the cells in S-phase. From the results, it is concluded that overexpression of high-mobility group protein B1 in the presence of irinotecan precedes breast cancer cells into apoptosis and in this response the binding of irinotecan to chromatin or high-mobility group protein B1 may condense/aggregate chromatin, preventing high-mobility group protein B1 release from chromatin.
Collapse
Affiliation(s)
- Saeedeh Keyvani-Ghamsari
- Department of Biochemistry, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Azra Rabbani-Chadegani
- Department of Biochemistry, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Javad Sargolzaei
- Department of Biochemistry, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Maryam Shahhoseini
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, Tehran, Iran
| |
Collapse
|
16
|
Simvastatin inhibits tumor angiogenesis in HER2-overexpressing human colorectal cancer. Biomed Pharmacother 2017; 85:418-424. [DOI: 10.1016/j.biopha.2016.11.045] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 11/09/2016] [Accepted: 11/09/2016] [Indexed: 11/17/2022] Open
|
17
|
Li M, Huang Y, Dong X, Wei Q, Li J, Sun H, Li C, Qi C, Yang J. Simvastatin downregulated C35 expression and inhibited the proliferation of colon cancer cells Lovo and HT29 in vitro . Biosci Trends 2016; 10:227-30. [DOI: 10.5582/bst.2016.01025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Min Li
- Shandong Academy of Medicine Sciences, Shandong Provincial Institute of Parasitic Diseases
- School of Medicine and Life Sciences, Ji’nan University, Shandong Academy of Medicine Sciences
| | - Yong Huang
- Shandong Academy of Medicine Sciences, Shandong Provincial Institute of Parasitic Diseases
| | - Xuan Dong
- Shandong Academy of Medicine Sciences, Shandong Provincial Institute of Parasitic Diseases
| | - Qingkuan Wei
- Shandong Academy of Medicine Sciences, Shandong Provincial Institute of Parasitic Diseases
| | - Jin Li
- Shandong Academy of Medicine Sciences, Shandong Provincial Institute of Parasitic Diseases
| | - Hui Sun
- Shandong Academy of Medicine Sciences, Shandong Provincial Institute of Parasitic Diseases
| | - Chenchen Li
- Shandong Academy of Medicine Sciences, Shandong Provincial Institute of Parasitic Diseases
- School of Medicine and Life Sciences, Ji’nan University, Shandong Academy of Medicine Sciences
| | | | - Jingyu Yang
- Shandong Academy of Medicine Sciences, Shandong Provincial Institute of Parasitic Diseases
| |
Collapse
|