1
|
Chen J, Li G, Sun D, Li H, Chen L. Research progress of hexokinase 2 in inflammatory-related diseases and its inhibitors. Eur J Med Chem 2024; 264:115986. [PMID: 38011767 DOI: 10.1016/j.ejmech.2023.115986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/14/2023] [Accepted: 11/19/2023] [Indexed: 11/29/2023]
Abstract
Hexokinase 2 (HK2) is a crucial enzyme involved in glycolysis, which converts glucose into glucose-6-phosphate and plays a significant role in glucose metabolism. HK2 can mediate glycolysis, which is linked to the release of inflammatory factors. The over-expression of HK2 increases the production of pro-inflammatory cytokines, exacerbating the inflammatory reaction. Consequently, HK2 is closely linked to various inflammatory-related diseases affecting multiple systems, including the digestive, nervous, circulatory, respiratory, reproductive systems, as well as rheumatoid arthritis. HK2 is regarded as a novel therapeutic target for inflammatory-related diseases, and this article provides a comprehensive review of its roles in these conditions. Furthermore, the development of potent HK2 inhibitors has garnered significant attention in recent years. Therefore, this review also presents a summary of potential HK2 inhibitors, offering promising prospects for the treatment of inflammatory-related diseases in the future.
Collapse
Affiliation(s)
- Jinxia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Guirong Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Dejuan Sun
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Hua Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; Institute of Structural Pharmacology & TCM Chemical Biology, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Lixia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
2
|
Sung YS, Kerimoglu B, Ooi A, Tomat E. Aroylhydrazone Glycoconjugate Prochelators Exploit Glucose Transporter 1 (GLUT1) to Target Iron in Cancer Cells. ACS Med Chem Lett 2022; 13:1452-1458. [PMID: 36105345 PMCID: PMC9465708 DOI: 10.1021/acsmedchemlett.2c00250] [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: 05/25/2022] [Accepted: 08/15/2022] [Indexed: 11/29/2022] Open
Abstract
Glycoconjugation strategies in anticancer drug discovery exploit the high expression of glucose transporters in malignant cells to achieve preferential uptake and hence attractive pharmacological characteristics of increased therapeutic windows and decreased unwanted toxicity. Here we present the design of glycoconjugated prochelators of aroylhydrazone AH1, an antiproliferative scavenger that targets the increased iron demand of rapidly proliferating malignant cells. The constructs feature a monosaccharide (d-glucose, d-glucosamine, or glycolytic inhibitor 2-deoxy-d-glucose) connected at the C2 or C6 position via a short linker, which masks the chelator through a disulfide bond susceptible to intracellular reduction. Cellular assays showed that the glycoconjugates rely on the GLUT1 transporter for uptake, lead to intracellular iron deprivation, and present antiproliferative activity. Ectopic overexpression of GLUT1 in malignant and normal cells increased the uptake and toxicity of the glycoconjugated prochelators, demonstrating that these compounds are well suited for targeting cells overexpressing glucose transporters and therefore for selective iron sequestration in malignant cells.
Collapse
Affiliation(s)
- Yu-Shien Sung
- Department
of Chemistry and Biochemistry, The University
of Arizona, 1306 E. University Blvd., Tucson, Arizona 85721-0041, United States
| | - Baris Kerimoglu
- Department
of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1703 E. Mabel St., Tucson, Arizona 85721, United
States
| | - Aikseng Ooi
- Department
of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1703 E. Mabel St., Tucson, Arizona 85721, United
States
| | - Elisa Tomat
- Department
of Chemistry and Biochemistry, The University
of Arizona, 1306 E. University Blvd., Tucson, Arizona 85721-0041, United States
| |
Collapse
|
3
|
Banerjee S, Banerjee S. Metal-Based Complexes as Potential Anti-cancer Agents. Anticancer Agents Med Chem 2022; 22:2684-2707. [PMID: 35362388 DOI: 10.2174/1871520622666220331085144] [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: 10/05/2021] [Revised: 12/16/2021] [Accepted: 02/03/2022] [Indexed: 11/22/2022]
Abstract
Metal based therapy is no new in biomedical research. In early days the biggest limitation was the inequality among therapeutical and toxicological dosages. Ever since, Barnett Rosenberg discovered cisplatin, a new era has begun to treat cancer with metal complexes. Platinum complexes such as oxaliplatin, cisplatin, and carboplatin, seem to be the foundation of metal/s-based components to challenge malignancies. With an advancement in the biomolemoecular mechanism, researchers have started developing non-classical platinum-based complexes, where a different mechanistic approach of the complexes is observed towards the biomolecular target. Till date, larger number of metal/s-based complexes was synthesized by overhauling the present structures chemically by substituting the ligand or preparing the whole novel component with improved cytotoxic and safety profiles. Howsoever, due to elevated accentuation upon the therapeutic importance of metal/s-based components, a couple of those agents are at present on clinical trials and several other are in anticipating regulatory endorsement to enter the trial. This literature highlights the detailed heterometallic multinuclear components, primarily focusing on platinum, ruthenium, gold and remarks on possible stability, synergism, mechanistic studies and structure activity relationships.
Collapse
Affiliation(s)
- Sabyasachi Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Ashram More, G.T. Road, Asansol-713301, West Bengal, India
| | - Subhasis Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Ashram More, G.T. Road, Asansol-713301, West Bengal, India
| |
Collapse
|
4
|
Design, synthesis and biological evaluation of colchicine glycoconjugates as tubulin polymerization inhibitors. Bioorg Med Chem 2022; 58:116671. [DOI: 10.1016/j.bmc.2022.116671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/29/2022] [Accepted: 02/13/2022] [Indexed: 11/30/2022]
|
5
|
Altay A, Caglar S, Caglar B. Silver(I) complexes containing diclofenac and niflumic acid induce apoptosis in human-derived cancer cell lines. Arch Physiol Biochem 2022; 128:69-79. [PMID: 31516039 DOI: 10.1080/13813455.2019.1662454] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We investigated the anti-cancer activity of [Ag(μ-dicl)]n 1 and [AgH(nif)2] 2 complexes against human cancer cell lines (MCF-7, HT-29, and HepG2) and mouse fibroblast (3T3-L1) cell line. Anti-proliferative activity was monitored by XTT cell viability and LDH leakage assays. Cell death mode was evaluated by multi-caspase activity, annexin V cytofluorimetric, MMP, cell cycle arrest, and ROS generation assays. Antioxidant capacity was evaluated on SOD, GPx, GR, and CAT enzymes. The XTT and LDH assay results showed that both complexes exhibited strong cytotoxicity against the tested cancer cell lines. The apoptotic mechanisms of the complexes were demonstrated by loss of MMP and increase in phosphatidylserine translocation, sub-G1 phase, and multi-caspase activity. Besides, both complexes induced the oxidative stress in MCF-7 cells by decreasing the activity of GPx, GR, and CAT enzymes. In conclusion, both Ag(I) complexes, especially 1, warrant for further in vivo evaluation as a new alternative in cancer treatment.HighlightsAg(I) complexes inhibited cell proliferation and induced LDH leakage in human cancer cell lines.Ag(I) complexes induced apoptosis through MMP disruption and ROS generation.Ag(I) complexes mimicked the multi-caspase activity.Ag(I) complexes increased the accumulation of sub-G1 phase.Ag(I) complexes inhibited the activity of antioxidant system enzymes.
Collapse
Affiliation(s)
- Ahmet Altay
- Department of Chemistry, Faculty of Arts and Science, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Sema Caglar
- Department of Chemistry, Faculty of Arts and Science, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Bulent Caglar
- Department of Chemistry, Faculty of Arts and Science, Erzincan Binali Yıldırım University, Erzincan, Turkey
| |
Collapse
|
6
|
Caglar S, Altay A, Kuzucu M, Caglar B. In Vitro Anticancer Activity of Novel Co(II) and Ni(II) Complexes of Non-steroidal Anti-inflammatory Drug Niflumic Acid Against Human Breast Adenocarcinoma MCF-7 Cells. Cell Biochem Biophys 2021; 79:729-746. [PMID: 33914261 DOI: 10.1007/s12013-021-00984-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2021] [Indexed: 12/16/2022]
Abstract
Herein, we report the synthesis, characterization and anticancer activity of six novel complexes of non-steroidal anti-inflammatory drug niflumic acid with Co(II) and Ni(II). In vitro cytotoxicity screening in MCF-7, HepG2 and HT-29 cancer cell lines showed that the complex 3 [Co(nif)2(met)(4-pic)] and complex 6 [Ni(nif)2(met)(4-pic)] among all the complexes exhibited the highest cytotoxicity against MCF-7 cells with IC50 values of 11.14 µM and, 41.47 µM, respectively. Besides, all the complexes exhibited significantly higher selectivity towards mouse fibroblast 3T3L1 cells. Further mechanistic studies with both complexes on MCF-7 cells revealed their cytotoxic action through the mitochondrial-dependent apoptotic pathway causing an increase oxidative/nitrosative stress, decrease in mitochondrial membrane potential (ΔΨm), inducing the multicaspase activation and arresting the cell cycle at S phase. q-PCR analysis resulted in an increase in the expression of the apoptotic marker proteins bax, p53 and caspase-3 and -8 in MCF-7 cells, but a decrease in the expression of antiapoptotic bcl-2 gene. Moreover, both complexes induced the apoptosis through the inhibition of PI3K/Akt signaling pathway by decreasing the expression of PI3K and increasing dephosphorylation form of Akt protein. These results provide a significant contribution to the explanation of the anticancer mechanisms of these complexes in MCF-7 cells.
Collapse
Affiliation(s)
- Sema Caglar
- Department of Chemistry, Faculty of Arts and Sciences, Erzincan Binali Yıldırım University, 24100, Erzincan, Turkey
| | - Ahmet Altay
- Department of Chemistry, Faculty of Arts and Sciences, Erzincan Binali Yıldırım University, 24100, Erzincan, Turkey.
| | - Mehmet Kuzucu
- Department of Biology, Faculty of Arts and Sciences, Erzincan Binali Yıldırım University, 24100, Erzincan, Turkey
| | - Bulent Caglar
- Department of Chemistry, Faculty of Arts and Sciences, Erzincan Binali Yıldırım University, 24100, Erzincan, Turkey
| |
Collapse
|
7
|
Fu J, Yang J, Seeberger PH, Yin J. Glycoconjugates for glucose transporter-mediated cancer-specific targeting and treatment. Carbohydr Res 2020; 498:108195. [PMID: 33220603 DOI: 10.1016/j.carres.2020.108195] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 12/29/2022]
Abstract
First observed in 1920s, the Warburg effects have inspired scientists to harness the unique glucose metabolism of cancer cells for targeted therapy for a century. Carbohydrate-drug conjugates are explicitly designed for selective uptake by cancer cells overexpressing glucose transporters. We summarize the progress in developing glycoconjugates for cancer-specific targeting and treatment over the past decade (2010-2020) and point to some future directions in this field.
Collapse
Affiliation(s)
- Junjie Fu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, PR China
| | - Jiaxin Yang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, PR China
| | - Peter H Seeberger
- Biomolecular Systems Department, Max Planck Institute of Colloids and Interfaces, Potsdam, 14476, Germany
| | - Jian Yin
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, PR China.
| |
Collapse
|
8
|
Altay A, Caglar S, Caglar B, Sahin ZS. Novel silver(I) complexes bearing mefenamic acid and pyridine derivatives: Synthesis, chemical characterization and in vitro anticancer evaluation. Inorganica Chim Acta 2019. [DOI: 10.1016/j.ica.2019.05.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
9
|
Pettenuzzo N, Brustolin L, Coltri E, Gambalunga A, Chiara F, Trevisan A, Biondi B, Nardon C, Fregona D. Cu II and Au III Complexes with Glycoconjugated Dithiocarbamato Ligands for Potential Applications in Targeted Chemotherapy. ChemMedChem 2019; 14:1162-1172. [PMID: 31091012 DOI: 10.1002/cmdc.201900226] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 04/29/2019] [Indexed: 11/07/2022]
Abstract
This work is focused on the synthesis, characterization, and preliminary biological evaluation of bio-conjugated AuIII and CuII complexes with the aim of overcoming the well-known side effects of chemotherapy by improving the selective accumulation of an anticancer metal payload in malignant cells. For this purpose, carbohydrates were chosen as targeting agents, exploiting the Warburg effect that accounts for the overexpression of glucose-transporter proteins (in particular GLUTs) in the phospholipid bilayer of most neoplastic cells. We linked the dithiocarbamato moiety to the C1 position of three different monosaccharides: d-glucose, d-galactose, and d-mannose. Altogether, six complexes with a 1:2 metal-to-ligand stoichiometry were synthesized and in vitro tested as anticancer agents. One of them showed high cytotoxic activity toward the HCT116 colorectal human carcinoma cell line, paving the way to future in vivo studies aimed at evaluating the role of carbohydrates in the selective delivery of whole molecules into cancerous cells.
Collapse
Affiliation(s)
- Nicolò Pettenuzzo
- Department of Chemical Sciences (DISC), University of Padova, Via Marzolo 1, 35131, Padova, Italy.,Department of Surgical, Oncological and Gastroenterological Sciences (DISCOG), University of Padova, Via Giustiniani 2, 35128, Padova, Italy
| | - Leonardo Brustolin
- Department of Chemical Sciences (DISC), University of Padova, Via Marzolo 1, 35131, Padova, Italy.,Department of Surgical, Oncological and Gastroenterological Sciences (DISCOG), University of Padova, Via Giustiniani 2, 35128, Padova, Italy
| | - Elisa Coltri
- Department of Chemical Sciences (DISC), University of Padova, Via Marzolo 1, 35131, Padova, Italy
| | - Alberto Gambalunga
- Department of Cardio-Thoraco-Vascular Sciences and Public Health (DCTV), University of Padova, Via Giustiniani 2, 35128, Padova, Italy
| | - Federica Chiara
- Department of Cardio-Thoraco-Vascular Sciences and Public Health (DCTV), University of Padova, Via Giustiniani 2, 35128, Padova, Italy
| | - Andrea Trevisan
- Department of Cardio-Thoraco-Vascular Sciences and Public Health (DCTV), University of Padova, Via Giustiniani 2, 35128, Padova, Italy
| | - Barbara Biondi
- Institute of Biomolecular Chemistry, Padova Unit, CNR, Via Marzolo 1, 35131, Padova, Italy
| | - Chiara Nardon
- Department of Chemical Sciences (DISC), University of Padova, Via Marzolo 1, 35131, Padova, Italy
| | - Dolores Fregona
- Department of Chemical Sciences (DISC), University of Padova, Via Marzolo 1, 35131, Padova, Italy
| |
Collapse
|
10
|
Transporter and protease mediated delivery of platinum complexes for precision oncology. J Biol Inorg Chem 2019; 24:457-466. [DOI: 10.1007/s00775-019-01660-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/09/2019] [Indexed: 01/03/2023]
|
11
|
Kenny RG, Marmion CJ. Toward Multi-Targeted Platinum and Ruthenium Drugs-A New Paradigm in Cancer Drug Treatment Regimens? Chem Rev 2019; 119:1058-1137. [PMID: 30640441 DOI: 10.1021/acs.chemrev.8b00271] [Citation(s) in RCA: 425] [Impact Index Per Article: 70.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
While medicinal inorganic chemistry has been practised for over 5000 years, it was not until the late 1800s when Alfred Werner published his ground-breaking research on coordination chemistry that we began to truly understand the nature of the coordination bond and the structures and stereochemistries of metal complexes. We can now readily manipulate and fine-tune their properties. This had led to a multitude of complexes with wide-ranging biomedical applications. This review will focus on the use and potential of metal complexes as important therapeutic agents for the treatment of cancer. With major advances in technologies and a deeper understanding of the human genome, we are now in a strong position to more fully understand carcinogenesis at a molecular level. We can now also rationally design and develop drug molecules that can either selectively enhance or disrupt key biological processes and, in doing so, optimize their therapeutic potential. This has heralded a new era in drug design in which we are moving from a single- toward a multitargeted approach. This approach lies at the very heart of medicinal inorganic chemistry. In this review, we have endeavored to showcase how a "multitargeted" approach to drug design has led to new families of metallodrugs which may not only reduce systemic toxicities associated with modern day chemotherapeutics but also address resistance issues that are plaguing many chemotherapeutic regimens. We have focused our attention on metallodrugs incorporating platinum and ruthenium ions given that complexes containing these metal ions are already in clinical use or have advanced to clinical trials as anticancer agents. The "multitargeted" complexes described herein not only target DNA but also contain either vectors to enable them to target cancer cells selectively and/or moieties that target enzymes, peptides, and intracellular proteins. Multitargeted complexes which have been designed to target the mitochondria or complexes inspired by natural product activity are also described. A summary of advances in this field over the past decade or so will be provided.
Collapse
Affiliation(s)
- Reece G Kenny
- Centre for Synthesis and Chemical Biology, Department of Chemistry , Royal College of Surgeons in Ireland , 123 St. Stephen's Green , Dublin 2 , Ireland
| | - Celine J Marmion
- Centre for Synthesis and Chemical Biology, Department of Chemistry , Royal College of Surgeons in Ireland , 123 St. Stephen's Green , Dublin 2 , Ireland
| |
Collapse
|
12
|
Yekke-Ghasemi Z, Takjoo R, Ramezani M, Mague JT. Molecular design and synthesis of new dithiocarbazate complexes; crystal structure, bioactivities and nano studies. RSC Adv 2018; 8:41795-41809. [PMID: 35558762 PMCID: PMC9091968 DOI: 10.1039/c8ra07100d] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 09/24/2018] [Indexed: 01/09/2023] Open
Abstract
The syntheses of a new set of metal complexes MoO2L'(CH3OH), VOL'(CH3O)(CH3OH), , , SnL'Cl2 and SnL'I2 with a new ligand (L = (2,2'(disulfanediylbis((ethylthio)methylene)bis(hydrazin-2-yl-1-ylidene)bis(methanylylidene)) diphenol; L' = S-ethyl-3-(2-hydroxyphenyl)methylenedithiocarbazate are described along with characterization by elemental analysis, mass spectrometry, spectroscopic (IR, 1H- and 13C-NMR) and TGA techniques. The crystal structures of compounds were determined by single crystal X-ray diffraction analysis and compared to powder X-ray diffraction (PXRD) patterns of the nano complexes obtained using ultrasonic methods. The PXRD results indicate that the compounds synthesized by ultrasonic methods have high crystallinity. The compounds were evaluated in an in vitro cytotoxicity study with two human cancer cell lines. The results of this study revealed that all complexes exhibit good cytotoxic activity when compared to the clinical drug, cisplatin. Interaction of the samples with human serum albumin (HSA) was investigated using fluorescence spectrophotometric methods and the Stern-Volmer quenching constant (K SV) and free energy changes (ΔG) were calculated at 298 K. The fluorescence quenching method is used to determine the number of binding sites (n) and association constants (K a) at the same temperatures.
Collapse
Affiliation(s)
- Zahra Yekke-Ghasemi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad Mashhad Iran +98 513 880 5536
| | - Reza Takjoo
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad Mashhad Iran +98 513 880 5536
| | - Mohammad Ramezani
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| | - Joel T Mague
- Department of Chemistry, Tulane University New Orleans LA 70118 USA
| |
Collapse
|
13
|
Liu R, Fu Z, Zhao M, Gao X, Li H, Mi Q, Liu P, Yang J, Yao Z, Gao Q. GLUT1-mediated selective tumor targeting with fluorine containing platinum(II) glycoconjugates. Oncotarget 2018; 8:39476-39496. [PMID: 28467806 PMCID: PMC5503626 DOI: 10.18632/oncotarget.17073] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/22/2017] [Indexed: 02/07/2023] Open
Abstract
Increased glycolysis and overexpression of glucose transporters (GLUTs) are physiological characteristics of human malignancies. Based on the so-called Warburg effect, 18flurodeoxyglucose-positron emission tomography (FDG-PET) has successfully developed as clinical modality for the diagnosis and staging of many cancers. To leverage this glucose transporter mediated metabolic disparity between normal and malignant cells, in the current report, we focus on the fluorine substituted series of glucose, mannose and galactose-conjugated (trans-R,R-cyclohexane-1,2-diamine)-2-flouromalonato-platinum(II) complexes for a comprehensive evaluation on their selective tumor targeting. Besides highly improved water solubility, these sugar-conjugates presented improved cytotoxicity than oxaliplatin in glucose tranporters (GLUTs) overexpressing cancer cell lines and exhibited no cross-resistance to cisplatin. For the highly water soluble glucose-conjugated complex (5a), two novel in vivo assessments were conducted and the results revealed that 5a was more efficacious at a lower equitoxic dose (70% MTD) than oxaliplatin (100% MTD) in HT29 xenograft model, and it was significantly more potent than oxaliplatin in leukemia-bearing DBA/2 mice as well even at equimolar dose levels (18% vs 90% MTD). GLUT inhibitor mediated cell viability analysis, GLUT1 knockdown cell line-based cytotoxicity evaluation, and platinum accumulation study demonstrated that the cellular uptake of the sugar-conjugates was regulated by GLUT1. The higher intrinsic DNA reactivity of the sugar-conjugates was confirmed by kinetic study of platinum(II)-guanosine adduct formation. The mechanistic origin of the antitumor effect of the fluorine complexes was found to be forming the bifunctional Pt-guanine-guanine (Pt-GG) intrastrand cross-links with DNA. The results provide a rationale for Warburg effect targeted anticancer drug design.
Collapse
Affiliation(s)
- Ran Liu
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Zheng Fu
- Department of Immunology, Laboratory of Immune Micro-environment, Tianjin Medical University, Tianjin 300070, P. R. China
| | - Meng Zhao
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300040, P. R. China
| | - Xiangqian Gao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Hong Li
- Affiliated Hospital, Logistics University of the Chinese People's Armed Police Force, Tianjin 300162, P. R. China
| | - Qian Mi
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Pengxing Liu
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Jinna Yang
- Department of Medicinal Chemistry, Gudui BioPharma Technology Inc.,Huayuan Industrial Park, Tianjin 300384, P. R. China
| | - Zhi Yao
- Department of Immunology, Laboratory of Immune Micro-environment, Tianjin Medical University, Tianjin 300070, P. R. China
| | - Qingzhi Gao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, P. R. China
| |
Collapse
|
14
|
Ndagi U, Mhlongo N, Soliman ME. Metal complexes in cancer therapy - an update from drug design perspective. Drug Des Devel Ther 2017; 11:599-616. [PMID: 28424538 PMCID: PMC5344412 DOI: 10.2147/dddt.s119488] [Citation(s) in RCA: 583] [Impact Index Per Article: 72.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In the past, metal-based compounds were widely used in the treatment of disease conditions, but the lack of clear distinction between the therapeutic and toxic doses was a major challenge. With the discovery of cisplatin by Barnett Rosenberg in 1960, a milestone in the history of metal-based compounds used in the treatment of cancers was witnessed. This forms the foundation for the modern era of the metal-based anticancer drugs. Platinum drugs, such as cisplatin, carboplatin and oxaliplatin, are the mainstay of the metal-based compounds in the treatment of cancer, but the delay in the therapeutic accomplishment of other metal-based compounds hampered the progress of research in this field. Recently, however, there has been an upsurge of activities relying on the structural information, aimed at improving and developing other forms of metal-based compounds and nonclassical platinum complexes whose mechanism of action is distinct from known drugs such as cisplatin. In line with this, many more metal-based compounds have been synthesized by redesigning the existing chemical structure through ligand substitution or building the entire new compound with enhanced safety and cytotoxic profile. However, because of increased emphasis on the clinical relevance of metal-based complexes, a few of these drugs are currently on clinical trial and many more are awaiting ethical approval to join the trial. In this review, we seek to give an overview of previous reviews on the cytotoxic effect of metal-based complexes while focusing more on newly designed metal-based complexes and their cytotoxic effect on the cancer cell lines, as well as on new approach to metal-based drug design and molecular target in cancer therapy. We are optimistic that the concept of selective targeting remains the hope of the future in developing therapeutics that would selectively target cancer cells and leave healthy cells unharmed.
Collapse
Affiliation(s)
- Umar Ndagi
- Molecular Modelling and Drug Design Research Group, School of Health Sciences, University of KwaZulu-Natal, Westville, Durban, South Africa
| | - Ndumiso Mhlongo
- Molecular Modelling and Drug Design Research Group, School of Health Sciences, University of KwaZulu-Natal, Westville, Durban, South Africa
| | - Mahmoud E Soliman
- Molecular Modelling and Drug Design Research Group, School of Health Sciences, University of KwaZulu-Natal, Westville, Durban, South Africa
| |
Collapse
|
15
|
Li Q, Li C, Xie L, Zhang C, Feng Y, Xie J. Characterization of a putative ArsR transcriptional regulator encoded by Rv2642 from Mycobacterium tuberculosis. J Biomol Struct Dyn 2016; 35:2031-2039. [DOI: 10.1080/07391102.2016.1206037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Qiming Li
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Ministry of Education Eco-Environment of the Three Gorges Reservoir Region, Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Chunyan Li
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Ministry of Education Eco-Environment of the Three Gorges Reservoir Region, Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Longxiang Xie
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Ministry of Education Eco-Environment of the Three Gorges Reservoir Region, Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Chenhui Zhang
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Ministry of Education Eco-Environment of the Three Gorges Reservoir Region, Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Yonghong Feng
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, China
| | - Jianping Xie
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Ministry of Education Eco-Environment of the Three Gorges Reservoir Region, Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|